48 results on '"Motti, Ml"'
Search Results
2. Melanoma and immunotherapy bridge 2015 : Naples, Italy. 1-5 December 2015.
- Author
-
Nanda, VGY, Peng, W, Hwu, P, Davies, MA, Ciliberto, G, Fattore, L, Malpicci, D, Aurisicchio, L, Ascierto, PA, Croce, CM, Mancini, R, Spranger, S, Gajewski, TF, Wang, Y, Ferrone, S, Vanpouille-Box, C, Wennerberg, E, Pilones, KA, Formenti, SC, Demaria, S, Tang, H, Fu, Y-X, Dummer, R, Puzanov, I, Tarhini, A, Chauvin, J-M, Pagliano, O, Fourcade, J, Sun, Z, Wang, H, Sanders, C, Kirkwood, JM, Chen, T-HT, Maurer, M, Korman, AJ, Zarour, HM, Stroncek, DF, Huber, V, Rivoltini, L, Thurin, M, Rau, T, Lugli, A, Pagès, F, Camarero, J, Sancho, A, Jommi, C, de Coaña, YP, Wolodarski, M, Yoshimoto, Y, Gentilcore, G, Poschke, I, Masucci, GV, Hansson, J, Kiessling, R, Scognamiglio, G, Sabbatino, F, Marino, FZ, Anniciello, AM, Cantile, M, Cerrone, M, Scala, S, D’alterio, C, Ianaro, A, Cirin, G, Liguori, G, Bott, G, Chapman, PB, Robert, C, Larkin, J, Haanen, JB, Ribas, A, Hogg, D, Hamid, O, Testori, A, Lorigan, P, Sosman, JA, Flaherty, KT, Yue, H, Coleman, S, Caro, I, Hauschild, A, McArthur, GA, Sznol, M, Callahan, MK, Kluger, H, Postow, MA, Gordan, R, Segal, NH, Rizvi, NA, Lesokhin, A, Atkins, MB, Burke, MM, Ralabate, A, Rivera, A, Kronenberg, SA, Agunwamba, B, Ruisi, M, Horak, C, Jiang, J, Wolchok, J, Liszkay, G, Maio, M, Mandalà, M, Demidov, L, Stoyakovskiy, D, Thomas, L, de la Cruz-Merino, L, Atkinson, V, Dutriaux, C, Garbe, C, Wongchenko, M, Chang, I, Koralek, DO, Rooney, I, Yan, Y, Dréno, B, Sullivan, R, Patel, M, Hodi, S, Amaria, R, Boasberg, P, Wallin, J, He, X, Cha, E, Richie, N, Ballinger, M, Smith, DC, Bauer, TM, Wasser, JS, Luke, JJ, Balmanoukian, AS, Kaufman, DR, Zhao, Y, Maleski, J, Leopold, L, Gangadhar, TC, Long, GV, Michielin, O, VanderWalde, A, Andtbacka, RHI, Cebon, J, Fernandez, E, Malvehy, J, Olszanski, AJ, Gause, C, Chen, L, Chou, J, Stephen Hodi, F, Brady, B, Mortier, L, Hassel, JC, Rutkowski, P, McNeil, C, Kalinka-Warzocha, E, Lebbé, C, Ny, L, Chacon, M, Queirolo, P, Loquai, C, Cheema, P, Berrocal, A, Eizmendi, KM, Bar-Sela, G, Hardy, H, Weber, JS, Grob, J-J, Marquez-Rodas, I, Schmidt, H, Briscoe, K, Baurain, J-F, Wolchok, JD, Pinto, R, De Summa, S, Garrisi, VM, Strippoli, S, Azzariti, A, Guida, G, Guida, M, Tommasi, S, Jacquelot, N, Enot, D, Flament, C, Pitt, JM, Vimond, N, Blattner, C, Yamazaki, T, Roberti, M-P, Vetizou, M, Daillere, R, Poirier-Colame, V, la Semeraro, M, Caignard, A, Slingluff, CL, Sallusto, F, Rusakiewicz, S, Weide, B, Marabelle, A, Kohrt, H, Dalle, S, Cavalcanti, A, Kroemer, G, Di Giacomo, AM, Wong, P, Yuan, J, Umansky, V, Eggermont, A, Zitvogel, L, Anna, P, Marco, T, Stefania, S, Francesco, M, Mariaelena, C, Gabriele, M, Antonio, AP, Franco, S, Roberti, MP, Enot, DP, Semeraro, M, Jégou, S, Flores, C, Kwon, BS, Anderson, AC, Borg, C, Aubin, F, Ayyoub, M, De Presbiteris, AL, Cordaro, FG, Camerlingo, R, Fratangelo, F, Mozzillo, N, Pirozzi, G, Patriarca, EJ, Caputo, E, Motti, ML, Falcon, R, Miceli, R, Capone, M, Madonna, G, Mallardo, D, Carrier, MV, Panza, E, De Cicco, P, Armogida, C, Ercolano, G, Botti, G, Cirino, G, Sandru, A, Blank, M, Balatoni, T, Olasz, J, Farkas, E, Szollar, A, Savolt, A, Godeny, M, Csuka, O, Horvath, S, Eles, K, Shoenfeld, Y, Kasler, M, Costantini, S, Capone, F, Moradi, F, Berglund, P, Leandersson, K, Linnskog, R, Andersson, T, Prasad, CP, Nigro, CL, Lattanzio, L, Proby, C, Syed, N, Occelli, M, Cauchi, C, Merlano, M, Harwood, C, Thompson, A, Crook, T, Bifulco, K, Ingangi, V, Minopoli, M, Ragone, C, Pessi, A, Mannavola, F, D’Oronzo, S, Felici, C, Tucci, M, Doronzo, A, Silvestris, F, Ferretta, A, Guida, S, Maida, I, Cocco, T, Passarelli, A, Quaresmini, D, Franzese, O, Palermo, B, Di Donna, C, Sperduti, I, Foddai, M, Stabile, H, Gismondi, A, Santoni, A, Nisticò, P, Sponghini, AP, Platini, F, Marra, E, Rondonotti, D, Alabiso, O, Fierro, MT, Savoia, P, Stratica, F, Quaglino, P, Di Monta, G, Corrado, C, Di Marzo, M, Ugo, M, Di Cecilia, ML, Nicola, M, Fusciello, C, Marra, A, Guarrasi, R, Baldi, C, Russo, R, Di Giulio, G, Faiola, V, Zeppa, P, Pepe, S, Gambale, E, Carella, C, Di Paolo, A, De Tursi, M, Marra, L, De Murtas, F, Sorrentino, V, Voinea, S, Panaitescu, E, Bolovan, M, Stanciu, A, Cinca, S, Botti, C, Aquino, G, Anniciello, A, Fortes, C, Mastroeni, S, Caggiati, A, Passarelli, F, Zappalà, A, Capuano, M, Bono, R, Nudo, M, Marino, C, Michelozzi, P, De Biasio, V, Battarra, VC, Formenti, S, Ascierto, ML, McMiller, TL, Berger, AE, Danilova, L, Anders, RA, Netto, GJ, Xu, H, Pritchard, TS, Fan, J, Cheadle, C, Cope, L, Drake, CG, Pardoll, DM, Taube, JM, Topalian, SL, Gnjatic, S, Nataraj, S, Imai, N, Rahman, A, Jungbluth, AA, Pan, L, Venhaus, R, Park, A, Lehmann, FF, Lendvai, N, Cohen, AD, Cho, HJ, Daniel, S, Hirsh, V, Nanda, VGY, Peng, W, Hwu, P, Davies, MA, Ciliberto, G, Fattore, L, Malpicci, D, Aurisicchio, L, Ascierto, PA, Croce, CM, Mancini, R, Spranger, S, Gajewski, TF, Wang, Y, Ferrone, S, Vanpouille-Box, C, Wennerberg, E, Pilones, KA, Formenti, SC, Demaria, S, Tang, H, Fu, Y-X, Dummer, R, Puzanov, I, Tarhini, A, Chauvin, J-M, Pagliano, O, Fourcade, J, Sun, Z, Wang, H, Sanders, C, Kirkwood, JM, Chen, T-HT, Maurer, M, Korman, AJ, Zarour, HM, Stroncek, DF, Huber, V, Rivoltini, L, Thurin, M, Rau, T, Lugli, A, Pagès, F, Camarero, J, Sancho, A, Jommi, C, de Coaña, YP, Wolodarski, M, Yoshimoto, Y, Gentilcore, G, Poschke, I, Masucci, GV, Hansson, J, Kiessling, R, Scognamiglio, G, Sabbatino, F, Marino, FZ, Anniciello, AM, Cantile, M, Cerrone, M, Scala, S, D’alterio, C, Ianaro, A, Cirin, G, Liguori, G, Bott, G, Chapman, PB, Robert, C, Larkin, J, Haanen, JB, Ribas, A, Hogg, D, Hamid, O, Testori, A, Lorigan, P, Sosman, JA, Flaherty, KT, Yue, H, Coleman, S, Caro, I, Hauschild, A, McArthur, GA, Sznol, M, Callahan, MK, Kluger, H, Postow, MA, Gordan, R, Segal, NH, Rizvi, NA, Lesokhin, A, Atkins, MB, Burke, MM, Ralabate, A, Rivera, A, Kronenberg, SA, Agunwamba, B, Ruisi, M, Horak, C, Jiang, J, Wolchok, J, Liszkay, G, Maio, M, Mandalà, M, Demidov, L, Stoyakovskiy, D, Thomas, L, de la Cruz-Merino, L, Atkinson, V, Dutriaux, C, Garbe, C, Wongchenko, M, Chang, I, Koralek, DO, Rooney, I, Yan, Y, Dréno, B, Sullivan, R, Patel, M, Hodi, S, Amaria, R, Boasberg, P, Wallin, J, He, X, Cha, E, Richie, N, Ballinger, M, Smith, DC, Bauer, TM, Wasser, JS, Luke, JJ, Balmanoukian, AS, Kaufman, DR, Zhao, Y, Maleski, J, Leopold, L, Gangadhar, TC, Long, GV, Michielin, O, VanderWalde, A, Andtbacka, RHI, Cebon, J, Fernandez, E, Malvehy, J, Olszanski, AJ, Gause, C, Chen, L, Chou, J, Stephen Hodi, F, Brady, B, Mortier, L, Hassel, JC, Rutkowski, P, McNeil, C, Kalinka-Warzocha, E, Lebbé, C, Ny, L, Chacon, M, Queirolo, P, Loquai, C, Cheema, P, Berrocal, A, Eizmendi, KM, Bar-Sela, G, Hardy, H, Weber, JS, Grob, J-J, Marquez-Rodas, I, Schmidt, H, Briscoe, K, Baurain, J-F, Wolchok, JD, Pinto, R, De Summa, S, Garrisi, VM, Strippoli, S, Azzariti, A, Guida, G, Guida, M, Tommasi, S, Jacquelot, N, Enot, D, Flament, C, Pitt, JM, Vimond, N, Blattner, C, Yamazaki, T, Roberti, M-P, Vetizou, M, Daillere, R, Poirier-Colame, V, la Semeraro, M, Caignard, A, Slingluff, CL, Sallusto, F, Rusakiewicz, S, Weide, B, Marabelle, A, Kohrt, H, Dalle, S, Cavalcanti, A, Kroemer, G, Di Giacomo, AM, Wong, P, Yuan, J, Umansky, V, Eggermont, A, Zitvogel, L, Anna, P, Marco, T, Stefania, S, Francesco, M, Mariaelena, C, Gabriele, M, Antonio, AP, Franco, S, Roberti, MP, Enot, DP, Semeraro, M, Jégou, S, Flores, C, Kwon, BS, Anderson, AC, Borg, C, Aubin, F, Ayyoub, M, De Presbiteris, AL, Cordaro, FG, Camerlingo, R, Fratangelo, F, Mozzillo, N, Pirozzi, G, Patriarca, EJ, Caputo, E, Motti, ML, Falcon, R, Miceli, R, Capone, M, Madonna, G, Mallardo, D, Carrier, MV, Panza, E, De Cicco, P, Armogida, C, Ercolano, G, Botti, G, Cirino, G, Sandru, A, Blank, M, Balatoni, T, Olasz, J, Farkas, E, Szollar, A, Savolt, A, Godeny, M, Csuka, O, Horvath, S, Eles, K, Shoenfeld, Y, Kasler, M, Costantini, S, Capone, F, Moradi, F, Berglund, P, Leandersson, K, Linnskog, R, Andersson, T, Prasad, CP, Nigro, CL, Lattanzio, L, Proby, C, Syed, N, Occelli, M, Cauchi, C, Merlano, M, Harwood, C, Thompson, A, Crook, T, Bifulco, K, Ingangi, V, Minopoli, M, Ragone, C, Pessi, A, Mannavola, F, D’Oronzo, S, Felici, C, Tucci, M, Doronzo, A, Silvestris, F, Ferretta, A, Guida, S, Maida, I, Cocco, T, Passarelli, A, Quaresmini, D, Franzese, O, Palermo, B, Di Donna, C, Sperduti, I, Foddai, M, Stabile, H, Gismondi, A, Santoni, A, Nisticò, P, Sponghini, AP, Platini, F, Marra, E, Rondonotti, D, Alabiso, O, Fierro, MT, Savoia, P, Stratica, F, Quaglino, P, Di Monta, G, Corrado, C, Di Marzo, M, Ugo, M, Di Cecilia, ML, Nicola, M, Fusciello, C, Marra, A, Guarrasi, R, Baldi, C, Russo, R, Di Giulio, G, Faiola, V, Zeppa, P, Pepe, S, Gambale, E, Carella, C, Di Paolo, A, De Tursi, M, Marra, L, De Murtas, F, Sorrentino, V, Voinea, S, Panaitescu, E, Bolovan, M, Stanciu, A, Cinca, S, Botti, C, Aquino, G, Anniciello, A, Fortes, C, Mastroeni, S, Caggiati, A, Passarelli, F, Zappalà, A, Capuano, M, Bono, R, Nudo, M, Marino, C, Michelozzi, P, De Biasio, V, Battarra, VC, Formenti, S, Ascierto, ML, McMiller, TL, Berger, AE, Danilova, L, Anders, RA, Netto, GJ, Xu, H, Pritchard, TS, Fan, J, Cheadle, C, Cope, L, Drake, CG, Pardoll, DM, Taube, JM, Topalian, SL, Gnjatic, S, Nataraj, S, Imai, N, Rahman, A, Jungbluth, AA, Pan, L, Venhaus, R, Park, A, Lehmann, FF, Lendvai, N, Cohen, AD, Cho, HJ, Daniel, S, and Hirsh, V
- Abstract
MELANOMA BRIDGE 2015 KEYNOTE SPEAKER PRESENTATIONS Molecular and immuno-advances K1 Immunologic and metabolic consequences of PI3K/AKT/mTOR activation in melanoma Vashisht G. Y. Nanda, Weiyi Peng, Patrick Hwu, Michael A. Davies K2 Non-mutational adaptive changes in melanoma cells exposed to BRAF and MEK inhibitors help the establishment of drug resistance Gennaro Ciliberto, Luigi Fattore, Debora Malpicci, Luigi Aurisicchio, Paolo Antonio Ascierto, Carlo M. Croce, Rita Mancini K3 Tumor-intrinsic beta-catenin signaling mediates tumor-immune avoidance Stefani Spranger, Thomas F. Gajewski K4 Intracellular tumor antigens as a source of targets of antibody-based immunotherapy of melanoma Yangyang Wang, Soldano Ferrone Combination therapies K5 Harnessing radiotherapy to improve responses to immunotherapy in cancer Claire Vanpouille-Box, Erik Wennerberg, Karsten A. Pilones, Silvia C. Formenti, Sandra Demaria K6 Creating a T cell-inflamed tumor microenvironment overcomes resistance to checkpoint blockade Haidong Tang, Yang Wang, Yang-Xin Fu K7 Biomarkers for treatment decisions? Reinhard Dummer K8 Combining oncolytic therapies in the era of checkpoint inhibitors Igor Puzanov K9 Immune checkpoint blockade for melanoma: should we combine or sequence ipilimumab and PD-1 antibody therapy? Michael A. Postow News in immunotherapy K10 An update on adjuvant and neoadjuvant therapy for melanom Ahmad Tarhini K11 Targeting multiple inhibitory receptors in melanoma Joe-Marc Chauvin, Ornella Pagliano, Julien Fourcade, Zhaojun Sun, Hong Wang, Cindy Sanders, John M. Kirkwood, Tseng-hui Timothy Chen, Mark Maurer, Alan J. Korman, Hassane M. Zarour K12 Improving adoptive immune therapy using genetically engineered T cells David F. Stroncek Tumor microenvironment and biomarkers K13 Myeloid cells and tumor exosomes: a crosstalk for assessing immunosuppression? Veronica Huber, Licia Rivoltini K14 Update on the SITC biomarker taskforce: progress and challenges Magdalena Thurin World-wide immunosc
- Published
- 2016
3. Akt-dependent T198 phosphorylation of cicli-dependent kinase inhibitor p27kip1 in breast cancer
- Author
-
Motti ML, De Marco C, Califano D, Fusco A, and Viglietto G.
- Published
- 2004
4. Regulation of p27Kip1 protein levels contributes to mitogenic effects of the RET/PTC kinase in thyroid carcinoma cells
- Author
-
Vitagliano D, Carlomagno F, Motti ML, Viglietto G, Nikiforov YE, Nikiforova MN, Hershman JM, Ryan AJ, Fusco A, Melillo RM, and Santoro M.
- Published
- 2004
5. Loss of p27 Expression Through RAS->BRAF->MAP Kinase-Dependent Pathway in Human Thyroid Carcinomas
- Author
-
Massimo Santoro, Silvia De Gisi, Angela Persico, Donatella Malanga, Gennaro Chiappetta, Carmela De Marco, Maria Letizia Motti, Giancarlo Troncone, Daniela Califano, Giuseppe Viglietto, Simona Losito, Alfredo Fusco, Fernanda Fabiani, Motti, Ml, Marco, Cd, Califano, D, Gisi, Sd, Malanga, D, Troncone, Giancarlo, Persico, A, Losito, S, Fabiani, F, Santoro, Massimo, Fusco, Alfredo, and Viglietto, G.
- Subjects
Proto-Oncogene Proteins B-raf ,endocrine system diseases ,MAP Kinase Signaling System ,thyroid ,Cell Line, Tumor ,medicine ,SKP2 ,Humans ,Thyroid Neoplasms ,Molecular Biology ,Thyroid cancer ,Thyroid hormone receptor ,biology ,Oncogene ,Cell growth ,Thyroid ,Cell Biology ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Genes, ras ,medicine.anatomical_structure ,Mitogen-activated protein kinase ,biology.protein ,Cancer research ,cell cycle ,Mitogen-Activated Protein Kinases ,ra ,Cyclin-Dependent Kinase Inhibitor p27 ,braf ,Developmental Biology - Abstract
In the present study, we report that the RAS/BRAF/MAP kinase cascade plays a crucial role in the regulation of the Skp2/p27 pathway in thyroid cancer cells and that this is critical for cell proliferation. In vitro studies with cellular models of human thyroid carcinoma cells demonstrated that the adoptive expression of oncogenic RET/PTC1, Ha-RASV12 or BRAFV600E enhances Skp2 and reduces p27 protein expression in a MAP kinase-dependent manner; that RAS/BRAF/MAP kinase-dependent control of p27 expression in thyroid cancer cells occurs by regulating the stability of Skp2 and p27 protein; and that antisense oligonucleotides to p27 suppress growth arrest induced by MEK inhibitors. Finally, analysis of human thyroid carcinomas indicated that MAP kinase-positive thyroid tumors-as detected by immunostaining for p-ERK - presented high p27 degradative activity and low levels of p27 protein (n = 30; p < 0.05). In summary, our results indicate that constitutive signalling of the MAP kinase cascade contributes to the development of thyroid cancer promoted by activated RAS and BRAF oncogenes and that this occurs, at least in part, by compromising the inhibitory function of p27.
- Published
- 2007
6. p27Kip1 is expressed in proliferating cells in its form phosphorylated on threonine 187
- Author
-
Lucio Palombini, Daniela Califano, Maria Letizia Motti, Maria Giovanna Russo, Pio Zeppa, Alessia Caleo, Giancarlo Troncone, Ilenia Migliaccio, Antonino Iaccarino, Emiliano A. Palmieri, J C Martinez, Troncone, Giancarlo, Martinez, Jc, Iaccarino, A, Zeppa, Pio, Caleo, A, Russo, M, Migliaccio, I, Motti, Ml, Califano, D, Palmieri, Ea, and Palombini, Lucio
- Subjects
medicine.medical_specialty ,Pathology ,p27Kip1 ,Histology ,medicine.diagnostic_test ,biology ,Proteolysis ,Immunocytochemistry ,threonine 187 ,S cell ,Stain ,Molecular biology ,Pathology and Forensic Medicine ,proliferating cell ,lcsh:Pathology ,medicine ,biology.protein ,Phosphorylation ,Histopathology ,Threonine ,Antibody ,Research Article ,lcsh:RB1-214 - Abstract
Background G1/S cell cycle progression requires p27Kip1 (p27) proteolysis, which is triggered by its phosphorylation on threonine (Thr) 187. Since its levels are abundant in quiescent and scarce in cycling cells, p27 is an approved marker for quiescent cells, extensively used in histopathology and cancer research. Methods However here we showed that by using a specific phosphorylation site (pThr187) antibody, p27 is detectable also in proliferative compartments of normal, dysplastic and neoplastic tissues. Results In fact, whereas un-phosphorylated p27 and MIB-1 showed a significant inverse correlation (Spearman R = -0.55; p < 0,001), pThr187-p27 was positively and significantly correlated with MIB-1 expression (Spearman R = 0.88; p < 0,001). Thus proliferating cells only stain for pThr187-p27, whereas they are un-reactive with the regular p27 antibodies. However increasing the sensitivity of the immunocytochemistry (ICH) by the use of an ultra sensitive detection system based on tiramide signal amplification, simultaneous expression and colocalisation of both forms of p27 was shown in proliferating compartments nuclei by double immunofluorescence and laser scanning confocal microscopy studies. Conclusion Overall, our data suggest that p27 expression also occurs in proliferating cells compartments and the combined use of both regular and phospho- p27 antibodies is suggested.
- Published
- 2005
7. The transcriptional repressor DREAM is involved in thyroid gene expression
- Author
-
Barbara D'Andrea, Lucio Nitsch, Anna Mascia, Giuseppe Viglietto, Tina Di Palma, Maria Letizia Motti, Mariastella Zannini, D'Andrea, B, DI PALMA, T, Mascia, A, Motti, Ml, Viglietto, G, Nitsch, Lucio, and Zannini, Mariastella
- Subjects
Untranslated region ,endocrine system ,Transcription, Genetic ,Cellular differentiation ,Thyroid Gland ,Repressor ,Biology ,Cell Line ,Reference Values ,medicine ,Animals ,Transcription factor ,DNA Primers ,Regulation of gene expression ,Base Sequence ,Reverse Transcriptase Polymerase Chain Reaction ,Thyroid ,Calcium-Binding Proteins ,Cell Differentiation ,Epithelial Cells ,Kv Channel-Interacting Proteins ,Cell Biology ,Blotting, Northern ,Molecular biology ,TATA Box ,humanities ,Cell biology ,Rats ,Repressor Proteins ,medicine.anatomical_structure ,Mutagenesis ,Calcium ,PAX8 ,5' Untranslated Regions ,psychological phenomena and processes ,FOXE1 - Abstract
Downstream regulatory element antagonistic modulator (DREAM) was originally identified in neuroendocrine cells as a calcium-binding protein that specifically binds to downstream regulatory elements (DRE) on DNA, and represses transcription of its target genes. To explore the possibility that DREAM may regulate the endocrine activity of the thyroid gland, we analyzed its mRNA expression in undifferentiated and differentiated thyroid cells. We demonstrated that DREAM is expressed in the normal thyroid tissue as well as in differentiated thyroid cells in culture while it is absent in FRT poorly differentiated cells. In the present work, we also show that DREAM specifically binds to DRE sites identified in the 5' untranslated region (UTR) of the thyroid-specific transcription factors Pax8 and TTF-2/FoxE1 in a calcium-dependent manner. By gel retardation assays we demonstrated that thapsigargin treatment increases the binding of DREAM to the DRE sequences present in Pax8 and TTF-2/Foxe1 5' UTRs, and this correlates with a significant reduction of the expression of these genes. Interestingly, in poorly differentiated thyroid cells overexpression of exogenous DREAM strongly inhibits Pax8 expression. Moreover, we provide evidence that a mutated form of DREAM unable to bind Ca(2+) interferes with thyroid cell proliferation. Therefore, we propose that in thyroid cells DREAM is a mediator of the calcium-signaling pathway and it is involved in the regulation of thyroid cell function.
- Published
- 2005
8. Complex regulation of the cyclin-dependent kinase inhibitor p27kip1 in thyroid cancer cells by the PI3K/AKT pathway: regulation of p27kip1 expression and localization
- Author
-
Motti, M. L., Califano, D., Troncone, G., Marco, C., Migliaccio, I., Palmieri, E., Pezzullo, L., Palombini, L., Alfredo Fusco, Viglietto, G., Motti, Ml, Califano, D, Troncone, Giancarlo, DE MARCO, C, Migliaccio, I, Palmieri, E, Pezzullo, L, Palombini, Lucio, Fusco, Alfredo, and Viglietto, G.
- Subjects
Cytoplasm ,Time Factors ,Morpholines ,Blotting, Western ,Cell Cycle Proteins ,Protein Serine-Threonine Kinases ,Transfection ,Phosphatidylinositol 3-Kinases ,Cytosol ,Cell Line, Tumor ,Proto-Oncogene Proteins ,Humans ,Thyroid Neoplasms ,Phosphorylation ,S-Phase Kinase-Associated Proteins ,Reverse Transcriptase Polymerase Chain Reaction ,Tumor Suppressor Proteins ,PTEN Phosphohydrolase ,Flow Cytometry ,Phosphoric Monoester Hydrolases ,Androstadienes ,Enzyme Activation ,Gene Expression Regulation, Neoplastic ,Original Research Paper ,Bromodeoxyuridine ,Microscopy, Fluorescence ,Chromones ,Wortmannin ,Proto-Oncogene Proteins c-akt ,Cyclin-Dependent Kinase Inhibitor p27 - Abstract
Functional inactivation of the tumor suppressor p27(kip1) in human cancer occurs either through loss of expression or through phosphorylation-dependent cytoplasmic sequestration. Here we demonstrate that dysregulation of the PI3K/AKT pathway is important in thyroid carcinogenesis and that p27(kip1) is a key target of the growth-regulatory activity exerted by this pathway in thyroid cancer cells. Using specific PI3K inhibitors (LY294002, wortmannin, and PTEN) and a dominant active AKT construct (myrAKT), we demonstrated that the PI3K/AKT pathway controlled thyroid cell proliferation by regulating the expression and subcellular localization of p27. Results obtained with phospho-specific antibodies and with transfection of nonphosphorylable p27(kip1) mutant constructs demonstrated that PI3K/AKT-dependent regulation of p27(kip1) mislocalization in thyroid cancer cells occurred via phosphorylation of p27(kip1) at T157 and T198 (but not at S10 or T187). Finally, we evaluated whether these results were applicable to human tumors. Analysis of 100 thyroid carcinomas indicated that p27(kip1) phosphorylation at T157/T198 and cytoplasmic mislocalization were preferentially associated with activation of the PI3K/AKT pathway. Thus the PI3/AKT pathway and its effector p27(kip1) play major roles in thyroid carcinogenesis.
- Published
- 2005
9. Critical role of cyclin D3 in TSH-dependent growth of thyrocytes and in hyperproliferative diseases of the thyroid gland
- Author
-
Giancarlo Troncone, Barbara Belletti, Lucio Palombini, Gustavo Baldassarre, Gennaro Chiappetta, Mario Monaco, Alfredo Fusco, Letizia Cito, Paola Bruni, Giuseppe Viglietto, Angelo Boccia, Maria Letizia Motti, Motti, Ml, Boccia, A, Belletti, B, Bruni, P, Troncone, Giancarlo, Cito, L, Monaco, M, Chiappetta, G, Baldassarre, G, Palombini, Lucio, Fusco, A, and Viglietto, G.
- Subjects
endocrine system ,Cancer Research ,endocrine system diseases ,Cyclin D ,Thyroid Gland ,Cyclin B ,Thyrotropin ,Cell Line ,Thyroid-stimulating hormone ,Cyclins ,Cyclin E ,Genetics ,medicine ,Animals ,Humans ,Cyclin D3 ,Molecular Biology ,Cyclin ,biology ,Thyroid ,Contact inhibition ,Thyroid Diseases ,Cyclin-Dependent Kinases ,Rats ,medicine.anatomical_structure ,Gene Expression Regulation ,biology.protein ,Cancer research ,Protein Kinases ,Cell Division ,Cyclin A2 - Abstract
We report that cyclin D3 is rate limiting for G1 progression in thyroid follicular cells and that its constitutive upregulation by chronic stimulation of the TSH/cAMP pathway plays a role in human and experimental hyperproliferative diseases of the thyroid gland. These conclusions are supported by in vitro and in vivo studies. In rat thyrocytes (PC Cl 3 cells), cyclin D3 expression is enhanced in response to activation of the TSH/cAMP pathway. Interference with the expression of G1 cyclins (in particular cyclin D3) by the antisense methodology strongly reduced TSH-dependent proliferation of PC Cl 3 cells, indicating that proper progression through G1 requires cyclin D3. Accordingly, PC Cl 3 cells engineered to overexpress cyclin D3 (PC-D3 cells) show enhanced growth rate and elude hormone-dependence and contact inhibition. Using an animal experimental model of thyroid stimulation, we demonstrate that cyclin D3 is a key mediator of TSH-dependent proliferation of thyroid follicular cells also in vivo. Cyclin D3 protein levels were higher in the thyrocytes from glands of propylthiouracil-treated rats compared with control animals. The increase in cyclin D3 expression occurred after the propylthiouracil-induced increase in TSH levels and preceded the burst of cell proliferation. Finally, we found that cyclin D3 protein is expressed in a fraction of human goiters but it is strongly overexpressed in most follicular adenomas.
- Published
- 2003
10. Glial cell line-derived neurotrophic factor induces proliferative inhibition of NT2/D1 cells through RET-mediated up-regulation of the cyclin-dependent kinase inhibitor p27(kip1)
- Author
-
Giuliana Salvatore, Giuseppe Viglietto, Rosa Marina Melillo, Massimo Santoro, Paola Bruni, Alfredo Fusco, Maria Letizia Motti, Gustavo Baldassarre, Angelo Boccia, Barbara Belletti, Maria Napolitano, Baldassarre, G, Bruni, P, Boccia, A, Salvatore, G, Melillo, ROSA MARINA, Motti, Ml, Napolitano, M, Belletti, B, Fusco, Alfredo, Santoro, Massimo, Viglietto, G., and Santoro, M
- Subjects
Stage-Specific Embryonic Antigens ,Cancer Research ,Glial Cell Line-Derived Neurotrophic Factor Receptors ,endocrine system diseases ,Cell ,Cell Cycle Proteins ,Nerve Tissue Proteins ,Glycosphingolipids ,Cell Line ,chemistry.chemical_compound ,Neurotrophic factors ,Proto-Oncogene Proteins ,Genetics ,Glial cell line-derived neurotrophic factor ,medicine ,Animals ,Drosophila Proteins ,Antigens, Tumor-Associated, Carbohydrate ,Glial Cell Line-Derived Neurotrophic Factor ,Nerve Growth Factors ,Molecular Biology ,neoplasms ,Neurons ,biology ,Oncogene ,Kinase ,Tumor Suppressor Proteins ,Proto-Oncogene Proteins c-ret ,Receptor Protein-Tyrosine Kinases ,Cell Differentiation ,Up-Regulation ,Cell biology ,Enzyme Activation ,medicine.anatomical_structure ,chemistry ,Cell culture ,biology.protein ,Growth inhibition ,GDNF family of ligands ,Cell Division ,Cyclin-Dependent Kinase Inhibitor p27 - Abstract
Growth factors of the glial cell line-derived neurotrophic factor (GDNF) family control the differentiation of neuronal cells of the central and peripheral nervous systems. Intracellular signalling of these growth factors is, at least in part, mediated by activation of the RET receptor tyrosine kinase. Here, we demonstrate that GDNF triggering inhibits the proliferation of the embryonal carcinoma cell line NT2/D1. This anti-proliferative effect is accompanied by down-regulation of the SSEA-3 antigen, a marker typical of undifferentiated NT2/D1 cells. We show that these effects are mediated by activation of RET signalling. The block of RET by a kinase-deficient dominant negative mutant impairs GDNF-dependent growth inhibition, whereas the adoptive expression of a constitutively active RET, the RET-MEN2A oncogene, promotes effects similar to those exerted by GDNF. We show that RET signalling increases the expression of the cyclin-dependent kinase inhibitor p27(kip1) in NT2/D1 cells. Both DNA synthesis inhibition and SSEA-3 down-regulation are prevented if p27(kip1) expression is blocked by an antisense construct, which demonstrates that RET-triggered effects are mediated by p27(kip1).
- Published
- 2002
11. Cytoplasmic relocalization and inhibition of the cyclin-dependent kinase inhibitor p27(Kip1) by PKB/Akt-mediated phosphorylation in breast cancer
- Author
-
Paola Bruni, Maria Letizia Motti, Floriana Vinci, Massimo Santoro, Rosa Marina Melillo, Gennaro Chiappetta, Giuseppe Viglietto, Philip N. Tsichlis, Alfonso Bellacosa, A D'Alessio, Daniela Califano, Alfredo Fusco, Viglietto, G, Motti, Ml, Bruni, P, Melillo, ROSA MARINA, D'Alessio, A, Califano, D, Vinci, F, Chiappetta, G, Tsichlis, P, Bellacosa, A, Fusco, Alfredo, Santoro, M., and Santoro, Massimo
- Subjects
Cell cycle checkpoint ,Kinase ,Cell growth ,General Medicine ,Biology ,urologic and male genital diseases ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,Cell culture ,Cytoplasm ,Cancer cell ,Phosphorylation ,biological phenomena, cell phenomena, and immunity ,Protein kinase B ,neoplasms - Abstract
The cyclin-dependent kinase inhibitor p27(kip1) is a putative tumor suppressor for human cancer. The mechanism underlying p27(kip1) deregulation in human cancer is, however, poorly understood. We demonstrate that the serine/threonine kinase Akt regulates cell proliferation in breast cancer cells by preventing p27(kip1)-mediated growth arrest. Threonine 157 (T157), which maps within the nuclear localization signal of p27(kip1), is a predicted Akt-phosphorylation site. Akt-induced T157 phosphorylation causes retention of p27(kip1) in the cytoplasm, precluding p27(kip1)-induced G1 arrest. Conversely, the p27(kip1)-T157A mutant accumulates in cell nuclei and Akt does not affect p27(kip1)-T157A-mediated cell cycle arrest. Lastly, T157-phosphorylated p27(kip1) accumulates in the cytoplasm of primary human breast cancer cells coincident with Akt activation. Thus, cytoplasmic relocalization of p27(kip1), secondary to Akt-mediated phosphorylation, is a novel mechanism whereby the growth inhibitory properties of p27(kip1) are functionally inactivated and the proliferation of breast cancer cells is sustained.
- Published
- 2002
12. Use of nutritional supplements among south Italian students of physical training and sport university
- Author
-
Mazzeo, F., Motti, M. L., Messina, G., Monda, V., Ascione, A., Domenico Tafuri, Palmieri, F., Messina, A., Monda, M., Mazzeo, F, Motti, Ml, Messina, G, Monda, Marcellino, Ascione, A, Tafuri, D, Palmieri, F, Messina, A, and Monda, M.
- Subjects
education
13. Targeting the p90RSK/MDM2/p53 Pathway Is Effective in Blocking Tumors with Oncogenic Up-Regulation of the MAPK Pathway Such as Melanoma and Lung Cancer.
- Author
-
Maietta I, Viscusi E, Laudati S, Iannaci G, D'Antonio A, Melillo RM, Motti ML, and De Falco V
- Subjects
- Humans, Cell Line, Tumor, Up-Regulation drug effects, Up-Regulation genetics, Cell Proliferation drug effects, Phosphorylation, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Cyclin-Dependent Kinase Inhibitor p21 genetics, Proto-Oncogene Proteins c-mdm2 metabolism, Tumor Suppressor Protein p53 metabolism, Melanoma metabolism, Melanoma pathology, Melanoma genetics, Lung Neoplasms pathology, Lung Neoplasms metabolism, Lung Neoplasms genetics, MAP Kinase Signaling System drug effects, Ribosomal Protein S6 Kinases, 90-kDa metabolism, Apoptosis drug effects
- Abstract
In most human tumors, the MAPK pathway is constitutively activated. Since p90RSK is downstream of MAPK, it is often hyperactive and capable of phosphorylating oncogenic substrates. We have previously shown that p90RSK phosphorylates MDM2 at S166, promoting p53 degradation in follicular thyroid carcinomas. Thus, the inhibition of p90RSK restores p53 expression, which in turn inhibits cell proliferation and promotes apoptosis. In the present study, we demonstrated that the p90RSK/MDM2/p53 pathway proved to be an excellent target in the therapy of tumors with MAPK hyperactivation. For this purpose, we selected p53wt melanoma, lung and medullary thyroid carcinoma cell lines with high activation of p90RSK. In these cell lines, we demonstrated that the p90RSK/MDM2/p53 pathway is implicated in the regulation of the cell cycle and apoptosis through p53-dependent transcriptional control of p21 and Bcl-2 . Furthermore, with an immunohistochemical evaluation of primary melanomas and lung tumors, which exhibit highly activated p90RSK compared to corresponding normal tissue, we demonstrated that MDM2 stabilization was associated with p90RSK phosphorylation. The results indicate that p90RSK is able to control the proliferative rate and induction of apoptosis through the regulation of p53wt levels by stabilizing MDM2 in selected tumors with constitutively activated MAPKs, making p90RSK a new attractive target for anticancer therapy.
- Published
- 2024
- Full Text
- View/download PDF
14. Dietary Protein and Physical Exercise for the Treatment of Sarcopenia.
- Author
-
Nasso R, D'Errico A, Motti ML, Masullo M, and Arcone R
- Abstract
Sarcopenia is a multifactorial age-related disorder that causes a decrease in muscle mass, strength, and function, leading to alteration of movement, risk of falls, and hospitalization. This article aims to review recent findings on the factors underlying sarcopenia and the strategies required to delay and counteract its symptoms. We focus on molecular factors linked to ageing, on the role of low-grade chronic and acute inflammatory conditions such as cancer, which contributes to the onset of sarcopenia, and on the clinical criteria for its diagnosis. The use of drugs against sarcopenia is still subject to debate, and the suggested approaches to restore muscle health are based on adequate dietary protein intake and physical exercise. We also highlight the difference in the amount and quality of amino acids within animal- and plant-based diets, as studies have often shown varying results regarding their effect on sarcopenia in elderly people. In addition, many studies have reported that non-pharmacological approaches, such as an optimization of dietary protein intake and training programs based on resistance exercise, can be effective in preventing and delaying sarcopenia. These approaches not only improve the maintenance of skeletal muscle function, but also reduce health care costs and improve life expectancy and quality in elderly people.
- Published
- 2024
- Full Text
- View/download PDF
15. Emerging Targeted Therapeutic Strategies to Overcome Imatinib Resistance of Gastrointestinal Stromal Tumors.
- Author
-
Masucci MT, Motti ML, Minopoli M, Di Carluccio G, and Carriero MV
- Subjects
- Humans, Imatinib Mesylate pharmacology, Imatinib Mesylate therapeutic use, Proto-Oncogene Proteins c-kit genetics, Receptor, Platelet-Derived Growth Factor alpha genetics, Mutation, Gastrointestinal Stromal Tumors drug therapy, Gastrointestinal Stromal Tumors genetics, Gastrointestinal Stromal Tumors pathology, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Gastrointestinal Neoplasms drug therapy, Gastrointestinal Neoplasms genetics, Gastrointestinal Neoplasms pathology
- Abstract
Gastrointestinal stromal tumors (GISTs) are the most common malignant mesenchymal neoplasms of the gastrointestinal tract. The gold standard for the diagnosis of GISTs is morphologic analysis with an immunohistochemical evaluation plus genomic profiling to assess the mutational status of lesions. The majority of GISTs are driven by gain-of-function mutations in the proto-oncogene c- KIT encoding the tyrosine kinase receptor (TKR) known as KIT and in the platelet-derived growth factor-alpha receptor ( PDGFRA ) genes. Approved therapeutics are orally available as tyrosine kinase inhibitors (TKIs) targeting KIT and/or PDGFRA oncogenic activation. Among these, imatinib has changed the management of patients with unresectable or metastatic GISTs, improving their survival time and delaying disease progression. Nevertheless, the majority of patients with GISTs experience disease progression after 2-3 years of imatinib therapy due to the development of secondary KIT mutations. Today, based on the identification of new driving oncogenic mutations, targeted therapy and precision medicine are regarded as the new frontiers for GISTs. This article reviews the most important mutations in GISTs and highlights their importance in the current understanding and treatment options of GISTs, with an emphasis on the most recent clinical trials.
- Published
- 2023
- Full Text
- View/download PDF
16. Inhibition of Interleukin-6 Dependent Metalloproteinases-9/2 Expression in Cancer Cells by Diet Polyphenols.
- Author
-
Arcone R, Nasso R, Pagliara V, D'Errico A, Motti ML, D'Angelo S, Carbonara G, and Masullo M
- Abstract
Among inflammatory cytokines, Interleukin-6 (IL-6) is one of the major activators of acute phase response and is also involved in immune response and cancer progression. IL-6 is involved in the up-regulation of enzymes and growth factors acting on the extracellular matrix (ECM) remodelling components in physio-pathological processes. IL-6 enhances the expression of metalloproteases (MMP-)2/9, enzymes that play a key role in ECM degradation and therefore contribute to the process of tumor metastasis. To counteract and/or prevent cancer diseases, many efforts have been devoted to the identification of factors able to inhibit the IL-6-dependent MMP-9/2 expression. Recently, diet polyphenols have been identified as molecules manifesting anti-inflammatory and anti-cancer properties beyond their well-known capacity to promote health on the basis of their antioxidant effects. This review summarizes the recent advances in this field, focusing on the protective effects exerted by diet polyphenols on the proliferation and invasiveness of tumor cells, with specific emphasis on the ability of these molecules to inhibit the IL-6-dependent upregulation of MMP-2/9., (Copyright© Bentham Science Publishers; For any queries, please email at epub@benthamscience.net.)
- Published
- 2023
- Full Text
- View/download PDF
17. p90RSK Regulates p53 Pathway by MDM2 Phosphorylation in Thyroid Tumors.
- Author
-
Maietta I, Del Peschio F, Buonocore P, Viscusi E, Laudati S, Iannaci G, Minopoli M, Motti ML, and De Falco V
- Abstract
The expression level of the tumor suppressor p53 is controlled by the E3 ubiquitin ligase MDM2 with a regulatory feedback loop, which allows p53 to upregulate its inhibitor MDM2. In this manuscript we demonstrated that p90RSK binds and phosphorylates MDM2 on serine 166 both in vitro and in vivo by kinase assay, immunoblot, and co-immunoprecipitation assay; this phosphorylation increases the stability of MDM2 which in turn binds p53, ubiquitinating it and promoting its degradation by proteasome. A pharmacological inhibitor of p90RSK, BI-D1870, decreases MDM2 phosphorylation, and restores p53 function, which in turn transcriptionally increases the expression of cell cycle inhibitor p21 and of pro-apoptotic protein Bax and downregulates the anti-apoptotic protein Bcl-2, causing a block of cell proliferation, measured by a BrdU assay and growth curve, and promoting apoptosis, measured by a TUNEL assay. Finally, an immunohistochemistry evaluation of primary thyroid tumors, in which p90RSK is very active, confirms MDM2 stabilization mediated by p90RSK phosphorylation.
- Published
- 2022
- Full Text
- View/download PDF
18. The Role of Nutrients in Prevention, Treatment and Post-Coronavirus Disease-2019 (COVID-19).
- Author
-
Motti ML, Tafuri D, Donini L, Masucci MT, De Falco V, and Mazzeo F
- Subjects
- Humans, Nutrients, Pandemics prevention & control, SARS-CoV-2, Post-Acute COVID-19 Syndrome, COVID-19 complications, COVID-19 prevention & control
- Abstract
SARS-CoV-2 virus, infecting human cells via its spike protein, causes Coronavirus disease 2019 (COVID-19). COVID-19 is characterized by shortness of breath, fever, and pneumonia and is sometimes fatal. Unfortunately, to date, there is still no definite therapy to treat COVID-19. Therefore, the World Health Organization (WHO) approved only supportive care. During the COVID-19 pandemic, the need to maintain a correct intake of nutrients to support very weakened patients in overcoming disease arose. The literature available on nutrient intake for COVID-19 is mainly focused on prevention. However, the safe intake of micro- and/or macro-nutrients can be useful either for preventing infection and supporting the immune response during COVID-19, as well as in the post-acute phase, i.e., "long COVID", that is sometimes characterized by the onset of various long lasting and disabling symptoms. The aim of this review is to focus on the role of nutrient intake during all the different phases of the disease, including prevention, the acute phase, and finally long COVID.
- Published
- 2022
- Full Text
- View/download PDF
19. Therapeutic Strategies Targeting Urokinase and Its Receptor in Cancer.
- Author
-
Masucci MT, Minopoli M, Di Carluccio G, Motti ML, and Carriero MV
- Abstract
Several studies have ascertained that uPA and uPAR do participate in tumor progression and metastasis and are involved in cell adhesion, migration, invasion and survival, as well as angiogenesis. Increased levels of uPA and uPAR in tumor tissues, stroma and biological fluids correlate with adverse clinic-pathologic features and poor patient outcomes. After binding to uPAR, uPA activates plasminogen to plasmin, a broad-spectrum matrix- and fibrin-degrading enzyme able to facilitate tumor cell invasion and dissemination to distant sites. Moreover, uPAR activated by uPA regulates most cancer cell activities by interacting with a broad range of cell membrane receptors. These findings make uPA and uPAR not only promising diagnostic and prognostic markers but also attractive targets for developing anticancer therapies. In this review, we debate the uPA/uPAR structure-function relationship as well as give an update on the molecules that interfere with or inhibit uPA/uPAR functions. Additionally, the possible clinical development of these compounds is discussed.
- Published
- 2022
- Full Text
- View/download PDF
20. ω-3 and ω-6 Polyunsaturated Fatty Acids, Obesity and Cancer.
- Author
-
D'Angelo S, Motti ML, and Meccariello R
- Subjects
- Colorectal Neoplasms etiology, Colorectal Neoplasms prevention & control, Diet adverse effects, Eating drug effects, Energy Metabolism drug effects, Humans, Inflammation, Obesity etiology, Obesity prevention & control, Anti-Inflammatory Agents pharmacology, Endocannabinoids pharmacology, Fatty Acids, Omega-3 pharmacology, Fatty Acids, Omega-6 pharmacology, Hypolipidemic Agents pharmacology
- Abstract
Recently, nutraceutical bioactive compounds in foods have been discovered for their potential health benefits regarding the prevention of chronic disorders, such as cancer, and inflammatory, cardiovascular, and metabolic diseases. Dietary omega-3 polyunsaturated fatty acids (ω-3PUFAs), including alpha-linolenic acid, docosapentaenoic acid, and eicosapentaenoic acid, are mostly attractive. They are available for the customers worldwide from commonly used foods and/or as components of commercial food supplements. The anti-inflammatory and hypotriglyceridemic effects of these fatty acids are well known, whereas pro-inflammatory properties have been recognized in their dietary counterparts, the ω-6PUFAs. Both ω-3 and ω-6PUFAs contribute to the production of lipid mediators such as endocannabinoids that are notably involved in control of food intake, energy sensing, and food-related disorders. In this review, we present ω-3 and ω-6PUFAs and their derivatives, endocannabinoids; discuss the anti-obesity effects of ω-3PUFAs; their roles in inflammation and colorectal cancer development; and how their action can be co-preventative and co-therapeutic.
- Published
- 2020
- Full Text
- View/download PDF
21. MicroRNAs as Key Players in Melanoma Cell Resistance to MAPK and Immune Checkpoint Inhibitors.
- Author
-
Motti ML, Minopoli M, Di Carluccio G, Ascierto PA, and Carriero MV
- Subjects
- Animals, Humans, Melanoma drug therapy, Melanoma pathology, Drug Resistance, Neoplasm, Immune Checkpoint Inhibitors therapeutic use, Melanoma genetics, MicroRNAs genetics, Mitogen-Activated Protein Kinases chemistry, Protein Kinase Inhibitors therapeutic use
- Abstract
Advances in the use of targeted and immune therapies have revolutionized the clinical management of melanoma patients, prolonging significantly their overall and progression-free survival. However, both targeted and immune therapies suffer limitations due to genetic mutations and epigenetic modifications, which determine a great heterogeneity and phenotypic plasticity of melanoma cells. Acquired resistance of melanoma patients to inhibitors of BRAF (BRAFi) and MEK (MEKi), which block the mitogen-activated protein kinase (MAPK) pathway, limits their prolonged use. On the other hand, immune checkpoint inhibitors improve the outcomes of patients in only a subset of them and the molecular mechanisms underlying lack of responses are under investigation. There is growing evidence that altered expression levels of microRNAs (miRNA)s induce drug-resistance in tumor cells and that restoring normal expression of dysregulated miRNAs may re-establish drug sensitivity. However, the relationship between specific miRNA signatures and acquired resistance of melanoma to MAPK and immune checkpoint inhibitors is still limited and not fully elucidated. In this review, we provide an updated overview of how miRNAs induce resistance or restore melanoma cell sensitivity to mitogen-activated protein kinase inhibitors (MAPKi) as well as on the relationship existing between miRNAs and immune evasion by melanoma cell resistant to MAPKi.
- Published
- 2020
- Full Text
- View/download PDF
22. Targeting the Formyl Peptide Receptor type 1 to prevent the adhesion of ovarian cancer cells onto mesothelium and subsequent invasion.
- Author
-
Minopoli M, Botti G, Gigantino V, Ragone C, Sarno S, Motti ML, Scognamiglio G, Greggi S, Scaffa C, Roca MS, Stoppelli MP, Ciliberto G, Losito NS, and Carriero MV
- Subjects
- Adult, Aged, Antineoplastic Agents pharmacology, Biomarkers, Tumor, Cell Line, Tumor, Extracellular Matrix Proteins metabolism, Female, Gene Expression, Humans, Middle Aged, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology, Receptors, Formyl Peptide genetics, Cell Adhesion drug effects, Cell Movement drug effects, Ovarian Neoplasms metabolism, Receptors, Formyl Peptide antagonists & inhibitors, Receptors, Formyl Peptide metabolism
- Abstract
Background: The biological behavior of epithelial ovarian cancer (EOC) is unique since EOC cells metastasize early to the peritoneum. Thereby, new anti-target agents designed to block trans-coelomic dissemination of EOC cells may be useful as anti-metastatic drugs. The Urokinase Plasminogen Activator Receptor (uPAR) is overexpressed in EOC tissues, and its truncated forms released in sera and/or ascitic fluid are associated with poor prognosis and unfavorable clinical outcome. We documented that uPAR triggers intra-abdominal dissemination of EOC cells through the interaction of its 84-95 sequence with the Formyl Peptide Receptor type 1 (FPR1), even as short linear peptide Ser-Arg-Ser-Arg-Tyr (SRSRY). While the pro-metastatic role of uPAR is well documented, little information regarding the expression and role of FPR1 in EOC is currently available., Methods: Expression levels of uPAR and FPR1 in EOC cells and tissues were assessed by immunofluorescence, Western blot, or immunohystochemistry. Cell adhesion to extra-cellular matrix proteins and mesothelium as well as mesothelium invasion kinetics by EOC cells were monitored using the xCELLigence technology or assessed by measuring cell-associated fluorescence. Cell internalization of FPR1 was identified on multiple z-series by confocal microscopy. Data from in vitro assays were analysed by one-way ANOVA and post-hoc Dunnett t-test for multiple comparisons. Tissue microarray data were analyzed with the Pearson's Chi-square (χ
2 ) test., Results: Co-expression of uPAR and FPR1 by SKOV-3 and primary EOC cells confers a marked adhesion to vitronectin. The extent of cell adhesion decreases to basal level by pre-exposure to anti-uPAR84-95 Abs, or to the RI-3 peptide, blocking the uPAR84-95/FPR1 interaction. Furthermore, EOC cells exposed to RI-3 or desensitized with an excess of SRSRY, fail to adhere also to mesothelial cell monolayers, losing the ability to cross them. Finally, primary and metastatic EOC tissues express a high level of FPR1., Conclusions: Our findings identify for the first time FPR1 as a potential biomarker of aggressive EOC and suggests that inhibitors of the uPAR84-95/FPR1 crosstalk may be useful for the treatment of metastatic EOC.- Published
- 2019
- Full Text
- View/download PDF
23. Minireview: The Epigenetic Modulation of KISS1 in Reproduction and Cancer.
- Author
-
Motti ML and Meccariello R
- Subjects
- Animals, Epigenesis, Genetic, Humans, Kisspeptins genetics, Neoplasms genetics, Reproduction genetics
- Abstract
Epigenetics describes how both lifestyle and environment may affect human health through the modulation of genome functions and without any change to the DNA nucleotide sequence. The discovery of several epigenetic mechanisms and the possibility to deliver epigenetic marks in cells, gametes, and biological fluids has opened up new perspectives in the prevention, diagnosis, and treatment of human diseases. In this respect, the depth of knowledge of epigenetic mechanisms is fundamental to preserving health status and to developing targeted interventions. In this minireview, we summarize the epigenetic modulation of the KISS1 gene in order to provide an example of epigenetic regulation in health and disease.
- Published
- 2019
- Full Text
- View/download PDF
24. Role of Microenvironment on the Fate of Disseminating Cancer Stem Cells.
- Author
-
Ingangi V, Minopoli M, Ragone C, Motti ML, and Carriero MV
- Abstract
Disseminating Cancer Stem Cells (CSCs) initiate growth in specific niches of the host tissues, the cellular and molecular components of which sustain signaling pathways that support their survival, self-renewal dormancy and reactivation. In the metastatic niche, tumor cells may enter in a dormant state to survive and, consequently, the metastasis can remain latent for years. Despite the clinical importance of metastatic latency, little is known about what induces CSCs to enter a dormant state and what allows them to remain viable for years in this state. CSCs exhibit genetic, epigenetic and cellular adaptations that confer resistance to classical therapeutic approaches. The identification of potential CSC targets is complicated by the fact that CSCs may arise as a consequence of their relationship with the local microenvironment into the metastatic niches. Indeed, microenvironment modulates the capability of CSCs to evade the innate immune response and survive. Some new therapeutic options that include drugs targeting microenvironment components are achieving encouraging results in reducing the number of CSCs in tumors and/or overcoming their resistance in preclinical studies. This review will focus on specific CSC features with an emphasis on the role of tumor microenvironment in supporting metastatic dissemination of CSCs. In addition, it sheds light on potential microenvironment-targeted therapies aimed to counteract seeding and survival of CSCs in the metastatic niche.
- Published
- 2019
- Full Text
- View/download PDF
25. Effect of ABT-888 on the apoptosis, motility and invasiveness of BRAFi-resistant melanoma cells.
- Author
-
Fratangelo F, Camerlingo R, Carriero MV, Pirozzi G, Palmieri G, Gentilcore G, Ragone C, Minopoli M, Ascierto PA, and Motti ML
- Subjects
- Adult, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Apoptosis drug effects, Benzimidazoles therapeutic use, Cell Line, Tumor, Cell Movement drug effects, Cell Proliferation drug effects, Cell Survival drug effects, Drug Resistance, Neoplasm genetics, Female, Humans, Imidazoles pharmacology, Imidazoles therapeutic use, Melanoma genetics, Melanoma pathology, Mutation, Neoplasm Invasiveness prevention & control, Oximes pharmacology, Oximes therapeutic use, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins B-raf genetics, Signal Transduction drug effects, Skin Neoplasms genetics, Skin Neoplasms pathology, Melanoma, Cutaneous Malignant, Benzimidazoles pharmacology, Drug Resistance, Neoplasm drug effects, Melanoma drug therapy, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Skin Neoplasms drug therapy
- Abstract
Melanoma is a molecularly heterogeneous disease with many genetic mutations and altered signaling pathways. Activating mutations in the BRAF oncogene are observed in approximately 50% of cutaneous melanomas and the use of BRAF inhibitor (BRAFi) compounds has been reported to improve the outcome of patients with BRAF-mutated metastatic melanoma. However, the majority of these patients develop resistance within 6-8 months following the initiation of BRAFi treatment. In this study, we examined the possible use of the poly(ADP-ribose) polymerase 1 (PARP1) inhibitor, ABT-888 (veliparib), as a novel molecule that may be successfully employed in the treatment of BRAFi-resistant melanoma cells. Sensitive and resistant to BRAFi dabrafenib A375 cells were exposed to increasing concentrations of ABT-888. Cell viability and apoptosis were assessed by MTT assay and Annexin V-FITC analysis, respectively. The cell migratory and invasive ability was investigated using the xCELLigence technology and Boyden chamber assays, respectively. ABT-888 was found to reduce cell viability and exhibited pro-apoptotic activity in melanoma cell lines, independently from the BRAF/NRAS mutation status, in a dose-dependent manner, with the maximal effect being reached in the 25-50 µM concentration range. Moreover, ABT-888 promoted apoptosis in both the sensitive and resistant A375 cells, suggesting that ABT-888 may be useful in the treatment of BRAFi-resistant subsets of melanoma cells. Finally, in accordance with the involvement of PARP1 in actin cytoskeletal machinery, we found that the cytoskeletal organization, motility and invasive capability of both the A375 and A375R cells decreased upon exposure to 5 µM ABT-888 for 24 h. On the whole, the findings of this study highlight the pivotal role of PARP1 in the migration and invasion of melanoma cells, suggesting that ABT-888 may indeed be effective, not only as a pro-apoptotic drug for use in the treatment of BRAFi-resistant melanoma cells, but also in suppressing their migratory and invasive activities.
- Published
- 2018
- Full Text
- View/download PDF
26. Controversial Role of Kisspeptins/KiSS-1R Signaling System in Tumor Development.
- Author
-
Fratangelo F, Carriero MV, and Motti ML
- Abstract
KiSS-1 was first described as a metastasis suppressor gene in malignant melanoma. KiSS-1 encodes a 145 amino-acid residue peptide that is further processed, producing the 54 amino acid metastin and shorter peptides collectively named kisspeptins (KPs). KPs bind and activate KiSS-1R (GPR54). Although the KPs system has been extensively studied for its role in endocrinology of reproductive axis in mammals, its role in cancer is still controversial. Experimental evidences show that KP system exerts an anti-metastatic effect by the regulation of cellular migration and invasion in several cancer types. However, the role of KPs/KiSS-1R is very complex. Genomic studies suggest that KiSS-1/KiSS-1R expression might be different in the various stages of tumor development. Furthermore, overexpression of KiSS-1R has been reported to elicit drug resistance in triple negative breast cancer. In this review, we focused on multiple functions exerted by the KPs/KiSS-1R system in regulating tumor progression.
- Published
- 2018
- Full Text
- View/download PDF
27. MicroRNAs, Cancer and Diet: Facts and New Exciting Perspectives.
- Author
-
Motti ML, D Angelo S, and Meccariello R
- Subjects
- Epigenesis, Genetic, Food, Humans, MicroRNAs biosynthesis, MicroRNAs genetics, Neoplasms diagnosis, Neoplasms therapy, Diet, MicroRNAs metabolism, Neoplasms genetics
- Abstract
Background: MicroRNAs (miRNAs) are small non-coding RNAs able to regulate gene expression at multiple levels. They are detected in tissues, blood, and other body fluids with high stability and have a recognized role in maintaining of tissue homeostasis. Aberrant expression profile of miRNAs has been observed in several diseases, primarily cancer. As a consequence, the analysis of miRNA signature has recognized diagnostic and prognostic role in human diseases, and the development of miRNA-based therapies is currently under investigation. Recently, emerging but controversial data have revealed the possibility that diet-derived miRNAs might be transferred from food in living organisms to regulate gene expression. Thus, exogenous dietderived miRNAs might substantially contribute to the pool of circulating miRNAs to preserve tissue homeostasis and health status in recipient's organisms, opening new perspectives for diet in heath and disease., Objective: This brief review aims at summarizing data concerning the recognized role of miRNAs as biomarkers, drugs and therapeutic targets in cancer and the detection and the activity of diet-derived miRNAs in both physiological and pathological conditions., Conclusion: MiRNAs have emerged as crucial molecules in anticancer therapies and diet-derived miRNAs might contribute to the pool of circulating miRNAs to preserve, maintain or restore health., (Copyright© Bentham Science Publishers; For any queries, please email at epub@benthamscience.org.)
- Published
- 2018
- Full Text
- View/download PDF
28. Targeting the cross-talk between Urokinase receptor and Formyl peptide receptor type 1 to prevent invasion and trans-endothelial migration of melanoma cells.
- Author
-
Ragone C, Minopoli M, Ingangi V, Botti G, Fratangelo F, Pessi A, Stoppelli MP, Ascierto PA, Ciliberto G, Motti ML, and Carriero MV
- Subjects
- Cell Line, Tumor, Cell Movement physiology, Cell Proliferation physiology, Humans, Melanoma genetics, Melanoma pathology, Receptors, Formyl Peptide genetics, Transfection, Urokinase-Type Plasminogen Activator genetics, Melanoma metabolism, Receptors, Formyl Peptide metabolism, Urokinase-Type Plasminogen Activator metabolism
- Abstract
Background: Accumulating evidence demonstrates that the Urokinase Receptor (uPAR) regulates tumor cell migration through its assembly in composite regulatory units with transmembrane receptors, and uPAR
88-92 is the minimal sequence required to induce cell motility through the Formyl Peptide Receptor type 1 (FPR1). Both uPAR and FPR1 are involved in melanoma tumor progression, suggesting that they may be targeted for therapeutic purposes. In this study, the role of the uPAR-FPR1 cross-talk to sustain melanoma cell ability to invade extracellular matrix and cross endothelial barriers is investigated. Also, the possibility that inhibition of the uPAR mediated FPR1-dependent signaling may prevent matrix invasion and transendothelial migration of melanoma cells was investigated., Methods: Expression levels of uPAR and FPR1 were assessed by immunocytochemistry, Western Blot and qRT-PCR. Cell migration was investigated by Boyden chamber and wound-healing assays. Migration and invasion kinetics, trans-endothelial migration and proliferation of melanoma cells were monitored in real time using the xCELLigence technology. The agonist-triggered FPR1 internalization was visualized by confocal microscope. Cell adhesion to endothelium was determined by fluorometer measurement of cell-associated fluorescence or identified on multiple z-series by laser confocal microscopy. The 3D-organotypic models were set up by seeding melanoma cells onto collagen I matrices embedded dermal fibroblasts. Data were analyzed by one-way ANOVA and post-hoc Dunnett t-test for multiple comparisons., Results: We found that the co-expression of uPAR and FPR1 confers to A375 and M14 melanoma cells a clear-cut capability to move towards chemotactic gradients, to cross extracellular matrix and endothelial monolayers. FPR1 activity is required, as cell migration and invasion were abrogated by receptor desensitization. Finally, melanoma cell ability to move toward chemotactic gradients, invade matrigel or fibroblast-embedded collagen matrices and cross endothelial monolayers are prevented by anti-uPAR84-95 antibodies or by the RI-3 peptide which we have previously shown to inhibit the uPAR84-95 /FPR1 interaction., Conclusions: Collectively, our findings identify uPAR and FPR1 as relevant effectors of melanoma cell invasiveness and suggest that inhibitors of the uPAR84-95 /FPR1 cross-talk may be useful for the treatment of metastatic melanoma.- Published
- 2017
- Full Text
- View/download PDF
29. Retro-inverso Urokinase Receptor Antagonists for the Treatment of Metastatic Sarcomas.
- Author
-
Carriero MV, Bifulco K, Ingangi V, Costantini S, Botti G, Ragone C, Minopoli M, Motti ML, Rea D, Scognamiglio G, Botti G, Arra C, Ciliberto G, and Pessi A
- Subjects
- Animals, Cell Line, Tumor, Cell Movement drug effects, Cell Proliferation drug effects, Humans, Mice, Molecular Dynamics Simulation, Neoplasm Invasiveness genetics, Neoplasm Invasiveness pathology, Neoplasm Metastasis, Neovascularization, Pathologic genetics, Neovascularization, Pathologic pathology, Receptors, Formyl Peptide antagonists & inhibitors, Receptors, Formyl Peptide genetics, Receptors, Urokinase Plasminogen Activator genetics, Sarcoma genetics, Sarcoma pathology, Xenograft Model Antitumor Assays, Neovascularization, Pathologic drug therapy, Peptides administration & dosage, Receptors, Urokinase Plasminogen Activator antagonists & inhibitors, Sarcoma drug therapy
- Abstract
The development of metastases is a multistep process that requires the activation of physiological and biochemical processes that govern migration, invasion and entry of metastatic cells into blood vessels. The urokinase receptor (uPAR) promotes cell migration by interacting with the Formyl Peptide Receptors (FPRs). Since both uPAR and FPR1 are involved in tumor progression, the uPAR-FPR1 interaction is an attractive therapeutic target. We previously described peptide antagonists of the uPAR-FPR1 interaction that inhibited cell migration and angiogenesis. To develop enzyme-resistant analogues, we applied here the Retro-Inverso (RI) approach, whereby the topology of the side chains is maintained by inverting the sequence of the peptide and the chirality of all residues. Molecular dynamics suggests that peptide RI-3 adopts the turn structure typical of uPAR-FPR1 antagonists. Accordingly, RI-3 is a nanomolar competitor of N-formyl-Met-Leu-Phe for binding to FPR1 and inhibits migration, invasion, trans-endothelial migration of sarcoma cells and VEGF-triggered endothelial tube formation. When sarcoma cells were subcutaneously injected in nude mice, tumor size, intra-tumoral microvessel density, circulating tumor cells and pulmonary metastases were significantly reduced in animals treated daily with 6 mg/Kg RI-3 as compared to animals treated with vehicle only. Thus, RI-3 represents a promising lead for anti-metastatic drugs.
- Published
- 2017
- Full Text
- View/download PDF
30. The urokinase receptor-derived cyclic peptide [SRSRY] suppresses neovascularization and intravasation of osteosarcoma and chondrosarcoma cells.
- Author
-
Ingangi V, Bifulco K, Yousif AM, Ragone C, Motti ML, Rea D, Minopoli M, Botti G, Scognamiglio G, Fazioli F, Gallo M, De Chiara A, Arra C, Grieco P, and Carriero MV
- Subjects
- Animals, Cell Line, Tumor, Cell Movement, Chondrosarcoma pathology, Female, Humans, Mice, Mice, Nude, Neoplasm Invasiveness, Osteosarcoma pathology, Receptors, Formyl Peptide physiology, Bone Neoplasms blood supply, Chondrosarcoma blood supply, Neovascularization, Pathologic drug therapy, Osteosarcoma blood supply, Peptides, Cyclic therapeutic use, Receptors, Urokinase Plasminogen Activator therapeutic use
- Abstract
The receptor for the urokinase-type plasminogen activator (uPAR) is a widely recognized master regulator of cell migration and uPAR88-92 is the minimal sequence required to induce cell motility and angiogenesis by interacting with the formyl peptide receptor type 1 (FPR1). In this study, we present evidence that the cyclization of the uPAR88-92 sequence generates a new potent inhibitor of migration, and extracellular matrix invasion of human osteosarcoma and chondrosarcoma cells expressing comparable levels of FPR1 on cell surface. In vitro, the cyclized peptide [SRSRY] prevents formation of capillary-like tubes by endothelial cells co-cultured with chondrosarcoma cells and trans-endothelial migration of osteosarcoma and chondrosarcoma cells. When chondrosarcoma cells were subcutaneously injected in nude mice, tumor size, intra-tumoral microvessel density and circulating tumor cells in blood samples collected before the sacrifice, were significantly reduced in animals treated daily with i.p-administration of 6 mg/Kg [SRSRY] as compared to animals treated with vehicle only. Our findings indicate that [SRSRY] prevents three key events occurring during the metastatic process of osteosarcoma and chondrosarcoma cells: the extracellular matrix invasion, the formation of a capillary network and the entry into bloodstream.
- Published
- 2016
- Full Text
- View/download PDF
31. Cyclization of the urokinase receptor-derived ser-arg-ser-arg-tyr Peptide generates a potent inhibitor of trans-endothelial migration of monocytes.
- Author
-
Yousif AM, Minopoli M, Bifulco K, Ingangi V, Di Carluccio G, Merlino F, Motti ML, Grieco P, and Carriero MV
- Subjects
- Amino Acid Sequence, Animals, Cell Line, Cell Line, Tumor, Chromatography, High Pressure Liquid, Cyclization, Macrophages drug effects, Macrophages physiology, Peptides chemistry, Protein Binding, Rats, Monocytes drug effects, Monocytes physiology, Peptides metabolism, Peptides pharmacology, Receptors, Urokinase Plasminogen Activator chemistry, Receptors, Urokinase Plasminogen Activator metabolism, Transendothelial and Transepithelial Migration drug effects
- Abstract
The receptor for the urokinase-type plasminogen activator (uPAR) is a widely recognized master regulator of cell migration and uPAR88-92 is the minimal sequence required to induce cell motility. We and others have previously documented that the uPAR88-92 sequence, even in the form of synthetic linear peptide (SRSRY), interacts with the formyl peptide receptor type 1 (FPR1), henceforth inducing cell migration of several cell lines, including monocytes. FPR1 is mainly expressed by mammalian phagocytic leukocytes and plays a crucial role in chemotaxis. In this study, we present evidence that the cyclization of the SRSRY sequence generates a new potent and stable inhibitor of monocyte trafficking. In rat basophilic leukaemia RBL-2H3/ETFR cells expressing high levels of constitutively activated FPR1, the cyclic SRSRY peptide ([SRSRY]) blocks FPR1 mediated cell migration by interfering with both internalization and ligand-uptake of FPR1. Similarly to RBL-2H3/ETFR cells, [SRSRY] competes with fMLF for binding to FPR1 and prevents agonist-induced FPR1 internalization in human monocyte THP-1 cells. Unlike scramble [RSSYR], [SRSRY] inhibits fMLF-directed migration of monocytes in a dose-dependent manner, with IC50 value of 0.01 nM. PMA-differentiated THP-1 cell exposure to fMLF gradient causes a marked cytoskeletal re-organization with the formation of F-actin rich pseudopodia that are prevented by the addition of [SRSRY]. Furthermore, [SRSRY] prevents migration of human primary monocytes and trans-endothelial migration of monocytes. Our findings indicate that [SRSRY] is a new FPR1 inhibitor which may suggest the development of new drugs for treating pathological conditions sustained by increased motility of monocytes, such as chronic inflammatory diseases.
- Published
- 2015
- Full Text
- View/download PDF
32. AurkA inhibitors enhance the effects of B-RAF and MEK inhibitors in melanoma treatment.
- Author
-
Caputo E, Miceli R, Motti ML, Taté R, Fratangelo F, Botti G, Mozzillo N, Carriero MV, Cavalcanti E, Palmieri G, Ciliberto G, Pirozzi G, and Ascierto PA
- Subjects
- Aurora Kinase A metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Humans, Immunohistochemistry, Ki-67 Antigen metabolism, Mitogen-Activated Protein Kinase Kinases metabolism, Models, Biological, Proto-Oncogene Proteins B-raf metabolism, Skin drug effects, Aurora Kinase A antagonists & inhibitors, Melanoma drug therapy, Melanoma enzymology, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins B-raf antagonists & inhibitors
- Abstract
Background: Aurora kinase A (AurkA) is over-expressed in melanoma and its inhibition has been observed to limit tumor growth, suggesting a potential role in melanoma treatment., Methods: A human melanoma cell line with the B-RAF (V600E) mutation (A375mel) was exposed to B-RAF inhibitor (GSK2118436), MEK inhibitor (GSK1120212) and AurkA inhibitor (MLN8054) as single agents or in various combinations (BRAF plus AurkA inhibitor, MEK plus AurkA inhibitor or triple combination BRAF plus MEK plus AurkA inhibitor). Cell proliferation was assessed using xCELLigence technology. Total protein extracts were examined for p53 and c-Myc protein expression by Western blot analysis. Drug anti-tumor effects were further assessed using a 3D-human melanoma skin reconstruction model, in which tissues were incubated with serum-free medium containing control, B-RAF plus MEK inhibitor, MEK plus AurkA inhibitor or the triple combination., Results: AurkA inhibitor plus B-RAF inhibitor, AurkA inhibitor plus MEK inhibitor or triple combination had a markedly greater anti-proliferative effect on A375 (BRAFV600E) melanoma cells than single agents. In the 3D human skin model, the triple combination had a greater anti-tumor effect at the epidermal/dermal junction than control or either double combination. However, S-100 and Ki-67 positively stained spindle-shaped cells were detected in the dermal stratum, suggesting the presence of alive and proliferating melanoma cells., Conclusions: These findings provide new prospects for melanoma research, including combined B-RAF/AurkA inhibition for B-RAF mutated melanomas and MEK/AurkA inhibitor combination for patients without B-RAF mutations. Moreover, for the first time, we have shown that a B-RAF, MEK and AurkA inhibitor triple drug combination offers increased efficacy against melanoma cell growth and might be considered as a potential treatment strategy for enhancing clinical response in melanoma. However, although this triple drug combination was more effective at the epidermal/dermal junction, the suggested presence of alive and proliferating melanoma cells in the dermal stratum could result in drug resistance and disease recurrence. Molecular characterization of these dermal cells may be critical for the development of novel therapeutic strategies.
- Published
- 2014
- Full Text
- View/download PDF
33. Loss of p27 expression through RAS-->BRAF-->MAP kinase-dependent pathway in human thyroid carcinomas.
- Author
-
Motti ML, De Marco C, Califano D, De Gisi S, Malanga D, Troncone G, Persico A, Losito S, Fabiani F, Santoro M, Chiappetta G, Fusco A, and Viglietto G
- Subjects
- Cell Line, Tumor, Cyclin-Dependent Kinase Inhibitor p27 genetics, Gene Expression Regulation, Neoplastic physiology, Humans, MAP Kinase Signaling System genetics, Thyroid Neoplasms enzymology, Thyroid Neoplasms genetics, Cyclin-Dependent Kinase Inhibitor p27 antagonists & inhibitors, Genes, ras physiology, MAP Kinase Signaling System physiology, Mitogen-Activated Protein Kinases physiology, Proto-Oncogene Proteins B-raf physiology, Thyroid Neoplasms metabolism
- Abstract
In the present study, we report that the RAS/BRAF/MAP kinase cascade plays a crucial role in the regulation of the Skp2/p27 pathway in thyroid cancer cells and that this is critical for cell proliferation. In vitro studies with cellular models of human thyroid carcinoma cells demonstrated that the adoptive expression of oncogenic RET/PTC1, Ha-RASV12 or BRAFV600E enhances Skp2 and reduces p27 protein expression in a MAP kinase-dependent manner; that RAS/BRAF/MAP kinase-dependent control of p27 expression in thyroid cancer cells occurs by regulating the stability of Skp2 and p27 protein; and that antisense oligonucleotides to p27 suppress growth arrest induced by MEK inhibitors. Finally, analysis of human thyroid carcinomas indicated that MAP kinase-positive thyroid tumors-as detected by immunostaining for p-ERK - presented high p27 degradative activity and low levels of p27 protein (n = 30; p < 0.05). In summary, our results indicate that constitutive signalling of the MAP kinase cascade contributes to the development of thyroid cancer promoted by activated RAS and BRAF oncogenes and that this occurs, at least in part, by compromising the inhibitory function of p27.
- Published
- 2007
- Full Text
- View/download PDF
34. Overexpression of the S-phase kinase-associated protein 2 in thyroid cancer.
- Author
-
Chiappetta G, De Marco C, Quintiero A, Califano D, Gherardi S, Malanga D, Scrima M, Montero-Conde C, Cito L, Monaco M, Motti ML, Pasquinelli R, Agosti V, Robledo M, Fusco A, and Viglietto G
- Subjects
- Carcinoma chemistry, Cell Line, Tumor, Cyclin-Dependent Kinase Inhibitor p27 analysis, Cyclin-Dependent Kinase Inhibitor p27 metabolism, Gene Amplification, Humans, RNA, Messenger analysis, RNA, Messenger metabolism, S-Phase Kinase-Associated Proteins analysis, S-Phase Kinase-Associated Proteins genetics, Thyroid Neoplasms chemistry, Carcinoma metabolism, Carcinoma pathology, S-Phase Kinase-Associated Proteins metabolism, Thyroid Neoplasms metabolism, Thyroid Neoplasms pathology
- Abstract
Loss of expression of the cyclin-dependent kinase inhibitor p27 through enhanced protein degradation frequently occurs in human cancer. Degradation of p27 requires ubiquitination by the S-phase kinase-associated protein 2 (Skp2), a member of the F-box family of Skp1-Cullin-F-box protein ubiquitin ligases. In the present study, we have investigated the role of Skp2 in human thyroid tumours. Immunohistochemistry analysis showed that Skp2 was overexpressed significantly in thyroid carcinomas (26 out of 51) compared with goitres (0 out of 12, P<0.001) or adenomas (1 out of 10, P<0.05), and that high Skp2 expression was detected more often in anaplastic thyroid (ATC; 83%, n=12) than follicular thyroid (FTC; 40%, n=20) or papillary thyroid (PTC; 42%, n=19) carcinomas (P<0.05). Thyroid cancer cell lines and tissues with high levels of Skp2 protein presented high p27 degradation activity and there was an inverse correlation between Skp2 and p27 expression in thyroid cancer tissues (n=68; P<0.05). In most cases, the observed overexpression of Skp2 protein was paralleled by an increase in the levels of Skp2 mRNA, and we observed Skp2 gene amplification at 5p13 in 2 out of 6 cell lines and in 9 out of 23 primary tumours (six out of eight ATCs, two out of nine PTCs and one out of six FTCs) using Q-PCR and/or fluorescence in situ hybridization analysis. Finally, in vitro experiments demonstrated that suppression of Skp2 expression drastically reduced proliferation of thyroid cancer cells and, conversely, forced expression of Skp2 circumvented serum dependency and contact inhibition in Skp2-negative cells by promoting p27 degradation. These findings indicate that Skp2 plays an important role for the development of thyroid cancer.
- Published
- 2007
- Full Text
- View/download PDF
35. Reduced E-cadherin expression contributes to the loss of p27kip1-mediated mechanism of contact inhibition in thyroid anaplastic carcinomas.
- Author
-
Motti ML, Califano D, Baldassarre G, Celetti A, Merolla F, Forzati F, Napolitano M, Tavernise B, Fusco A, and Viglietto G
- Subjects
- Adenocarcinoma, Follicular metabolism, Adenocarcinoma, Follicular pathology, Carcinoma pathology, Carcinoma, Papillary metabolism, Carcinoma, Papillary pathology, Cell Communication, Cell Count, Cell Line, Tumor, Cell Proliferation, Cyclin A metabolism, Cyclin-Dependent Kinase Inhibitor p27, Cyclin-Dependent Kinases metabolism, Cyclins metabolism, Cytoskeletal Proteins biosynthesis, Gene Expression Regulation, Neoplastic, Humans, Thyroid Neoplasms pathology, Trans-Activators biosynthesis, beta Catenin, Cadherins biosynthesis, Carcinoma metabolism, Cell Cycle Proteins physiology, Contact Inhibition physiology, Thyroid Neoplasms metabolism, Tumor Suppressor Proteins physiology
- Abstract
In the present study, we have characterized several human thyroid cancer cell lines of different histotypes for their responsiveness to contact inhibition. We found that cells derived from differentiated carcinoma (TPC-1, WRO) arrest in G(1) phase at confluence, whereas cells derived from anaplastic carcinoma (ARO, FRO and FB1) continue to grow after reaching confluence. Furthermore, we provide experimental evidence that the axis, E-cadherin/beta-catenin/p27(Kip1), represents an integral part of the regulatory mechanism that controls proliferation at a high cell density, whose disruption may play a key role in determining the clinical behaviour of thyroid cancer. This conclusion derives from the finding that: (i) the expression of p27(Kip1) is enhanced at high cell density only in cells responsive to contact inhibition (TPC-1, WRO), but not in contact-inhibition resistant cells (ARO, FRO or FB1 cells); (ii) the increase in p27(Kip1) also resulted in increased levels of p27(Kip1) bound to cyclin E-Cdk2 complex, a reduction in cyclin E-Cdk2 activity and dephosphorylation of the retinoblastoma protein; (iii) antisense inhibition of p27(Kip1) upregulation at high cell density in confluent-sensitive cells completely prevents the confluence-induced growth arrest; (iv) proper expression and/or membrane localization of E-cadherin is observed only in cells responsive to contact inhibition (TPC-1, NPA, WRO) but not in unresponsive cells (ARO, FRO or FB1); (v) disruption of E-cadherin-mediated cell-cell contacts at high cell density induced by an anti-E-cadherin neutralizing antibody, inhibits the induction of p27(kip1) and restores proliferation in contact-inhibited cells; (vi) re-expression of E-cadherin into cells unresponsive to contact inhibition (ARO, FB1) induces a p27(kip1) expression and growth arrest. In summary, our data indicate that the altered response to contact inhibition exhibited by thyroid anaplastic cancer cells is due to the failure to upregulate p27(Kip1) in response to cell-cell interactions.
- Published
- 2005
- Full Text
- View/download PDF
36. The transcriptional repressor DREAM is involved in thyroid gene expression.
- Author
-
D'Andrea B, Di Palma T, Mascia A, Motti ML, Viglietto G, Nitsch L, and Zannini M
- Subjects
- 5' Untranslated Regions genetics, Animals, Base Sequence, Blotting, Northern, Calcium metabolism, Calcium-Binding Proteins genetics, Cell Differentiation, Cell Line, DNA Primers, Epithelial Cells, Kv Channel-Interacting Proteins, Mutagenesis, Rats, Reference Values, Repressor Proteins genetics, Reverse Transcriptase Polymerase Chain Reaction, TATA Box, Thyroid Gland cytology, Transcription, Genetic, Calcium-Binding Proteins physiology, Repressor Proteins physiology, Thyroid Gland physiology
- Abstract
Downstream regulatory element antagonistic modulator (DREAM) was originally identified in neuroendocrine cells as a calcium-binding protein that specifically binds to downstream regulatory elements (DRE) on DNA, and represses transcription of its target genes. To explore the possibility that DREAM may regulate the endocrine activity of the thyroid gland, we analyzed its mRNA expression in undifferentiated and differentiated thyroid cells. We demonstrated that DREAM is expressed in the normal thyroid tissue as well as in differentiated thyroid cells in culture while it is absent in FRT poorly differentiated cells. In the present work, we also show that DREAM specifically binds to DRE sites identified in the 5' untranslated region (UTR) of the thyroid-specific transcription factors Pax8 and TTF-2/FoxE1 in a calcium-dependent manner. By gel retardation assays we demonstrated that thapsigargin treatment increases the binding of DREAM to the DRE sequences present in Pax8 and TTF-2/Foxe1 5' UTRs, and this correlates with a significant reduction of the expression of these genes. Interestingly, in poorly differentiated thyroid cells overexpression of exogenous DREAM strongly inhibits Pax8 expression. Moreover, we provide evidence that a mutated form of DREAM unable to bind Ca(2+) interferes with thyroid cell proliferation. Therefore, we propose that in thyroid cells DREAM is a mediator of the calcium-signaling pathway and it is involved in the regulation of thyroid cell function.
- Published
- 2005
- Full Text
- View/download PDF
37. Loss of the tumor suppressor gene PTEN marks the transition from intratubular germ cell neoplasias (ITGCN) to invasive germ cell tumors.
- Author
-
Di Vizio D, Cito L, Boccia A, Chieffi P, Insabato L, Pettinato G, Motti ML, Schepis F, D'Amico W, Fabiani F, Tavernise B, Venuta S, Fusco A, and Viglietto G
- Subjects
- Animals, Breast Neoplasms, Cell Line, Tumor, Chromosomes, Human, Pair 10, Female, Flow Cytometry, Humans, In Situ Hybridization, Loss of Heterozygosity, Male, Mice, PTEN Phosphohydrolase, RNA, Messenger genetics, Testis cytology, Testis embryology, Testis pathology, Cell Transformation, Neoplastic, Genes, Tumor Suppressor, Germinoma genetics, Phosphoric Monoester Hydrolases genetics, Testicular Neoplasms genetics, Tumor Suppressor Proteins genetics
- Abstract
PTEN/MMAC1/TEP1: (hereafter PTEN) is a tumor suppressor gene (located at 10q23) that is frequently mutated or deleted in sporadic human tumors. PTEN encodes a multifunctional phosphatase, which negatively regulates cell growth, migration and survival via the phosphatidylinositol 3'-kinase/AKT signalling pathway. Accordingly, Pten+/- mice develop various types of tumors including teratocarcinomas and teratomas. We have investigated PTEN expression in 60 bioptic specimens of germ cell tumors (32 seminomas, 22 embryonal carcinomas and six teratomas) and 22 intratubular germ cell neoplasias (ITGCN) adjacent to the tumors for PTEN protein and mRNA expression. In total, 10 testicular biopsies were used as controls. In the testis, PTEN was abundantly expressed in germ cells whereas it was virtually absent from 56% of seminomas as well as from 86% of embryonal carcinomas and virtually all teratomas. On the contrary, ITGCN intensely expressed PTEN, indicating that loss of PTEN expression is not an early event in testicular tumor development. The loss of PTEN expression occurs mainly at the RNA level as determined by in situ hybridization of cellular mRNA (17/22) but also it may involve some kind of post-transcriptional mechanisms in the remaining 25% of cases. Analysis of microsatellites D10S551, D10S541 and D10S1765 in GCTs (n=22) showed LOH at the PTEN locus at 10q23 in at least 36% of GCTs (three embryonal carcinoma, three seminoma, two teratoma); one seminoma and one embryonal (9%) carcinoma presented an inactivating mutation in the PTEN gene (2/22). Finally, we demonstrated that the phosphatidylinositol 3'-kinase/AKT pathway, which is regulated by the PTEN phosphatase, is crucial in regulating the proliferation of the NT2/D1 embryonal carcinoma cells, and that the cyclin-dependent kinase inhibitor p27(kip1) is a key downstream target of this pathway.
- Published
- 2005
- Full Text
- View/download PDF
38. Complex regulation of the cyclin-dependent kinase inhibitor p27kip1 in thyroid cancer cells by the PI3K/AKT pathway: regulation of p27kip1 expression and localization.
- Author
-
Motti ML, Califano D, Troncone G, De Marco C, Migliaccio I, Palmieri E, Pezzullo L, Palombini L, Fusco A, and Viglietto G
- Subjects
- Androstadienes pharmacology, Blotting, Western, Bromodeoxyuridine pharmacology, Cell Cycle Proteins metabolism, Cell Line, Tumor, Chromones pharmacology, Cyclin-Dependent Kinase Inhibitor p27, Cytoplasm metabolism, Cytosol metabolism, Enzyme Activation, Flow Cytometry, Humans, Microscopy, Fluorescence, Morpholines pharmacology, PTEN Phosphohydrolase, Phosphoric Monoester Hydrolases metabolism, Phosphorylation, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-akt, Reverse Transcriptase Polymerase Chain Reaction, S-Phase Kinase-Associated Proteins metabolism, Time Factors, Transfection, Tumor Suppressor Proteins metabolism, Wortmannin, Cell Cycle Proteins biosynthesis, Gene Expression Regulation, Neoplastic, Phosphatidylinositol 3-Kinases metabolism, Thyroid Neoplasms metabolism, Tumor Suppressor Proteins biosynthesis
- Abstract
Functional inactivation of the tumor suppressor p27(kip1) in human cancer occurs either through loss of expression or through phosphorylation-dependent cytoplasmic sequestration. Here we demonstrate that dysregulation of the PI3K/AKT pathway is important in thyroid carcinogenesis and that p27(kip1) is a key target of the growth-regulatory activity exerted by this pathway in thyroid cancer cells. Using specific PI3K inhibitors (LY294002, wortmannin, and PTEN) and a dominant active AKT construct (myrAKT), we demonstrated that the PI3K/AKT pathway controlled thyroid cell proliferation by regulating the expression and subcellular localization of p27. Results obtained with phospho-specific antibodies and with transfection of nonphosphorylable p27(kip1) mutant constructs demonstrated that PI3K/AKT-dependent regulation of p27(kip1) mislocalization in thyroid cancer cells occurred via phosphorylation of p27(kip1) at T157 and T198 (but not at S10 or T187). Finally, we evaluated whether these results were applicable to human tumors. Analysis of 100 thyroid carcinomas indicated that p27(kip1) phosphorylation at T157/T198 and cytoplasmic mislocalization were preferentially associated with activation of the PI3K/AKT pathway. Thus the PI3/AKT pathway and its effector p27(kip1) play major roles in thyroid carcinogenesis.
- Published
- 2005
- Full Text
- View/download PDF
39. p27Kip1 is expressed in proliferating cells in its form phosphorylated on threonine 187.
- Author
-
Troncone G, Martinez JC, Iaccarino A, Zeppa P, Caleo A, Russo M, Migliaccio I, Motti ML, Califano D, Palmieri EA, and Palombini L
- Abstract
BACKGROUND: G1/S cell cycle progression requires p27Kip1 (p27) proteolysis, which is triggered by its phosphorylation on threonine (Thr) 187. Since its levels are abundant in quiescent and scarce in cycling cells, p27 is an approved marker for quiescent cells, extensively used in histopathology and cancer research. METHODS: However here we showed that by using a specific phosphorylation site (pThr187) antibody, p27 is detectable also in proliferative compartments of normal, dysplastic and neoplastic tissues. RESULTS: In fact, whereas un-phosphorylated p27 and MIB-1 showed a significant inverse correlation (Spearman R = -0.55; p < 0,001), pThr187-p27 was positively and significantly correlated with MIB-1 expression (Spearman R = 0.88; p < 0,001). Thus proliferating cells only stain for pThr187-p27, whereas they are un-reactive with the regular p27 antibodies. However increasing the sensitivity of the immunocytochemistry (ICH) by the use of an ultra sensitive detection system based on tiramide signal amplification, simultaneous expression and colocalisation of both forms of p27 was shown in proliferating compartments nuclei by double immunofluorescence and laser scanning confocal microscopy studies. CONCLUSION: Overall, our data suggest that p27 expression also occurs in proliferating cells compartments and the combined use of both regular and phospho- p27 antibodies is suggested.
- Published
- 2005
- Full Text
- View/download PDF
40. Critical role of cyclin D3 in TSH-dependent growth of thyrocytes and in hyperproliferative diseases of the thyroid gland.
- Author
-
Motti ML, Boccia A, Belletti B, Bruni P, Troncone G, Cito L, Monaco M, Chiappetta G, Baldassarre G, Palombini L, Fusco A, and Viglietto G
- Subjects
- Animals, Cell Division drug effects, Cell Line, Cyclin D3, Cyclin E, Cyclin-Dependent Kinases metabolism, Gene Expression Regulation drug effects, Humans, Protein Kinases metabolism, Rats, Thyroid Gland metabolism, Thyroid Gland pathology, Cyclins metabolism, Thyroid Diseases metabolism, Thyroid Diseases pathology, Thyroid Gland cytology, Thyroid Gland drug effects, Thyrotropin pharmacology
- Abstract
We report that cyclin D3 is rate limiting for G1 progression in thyroid follicular cells and that its constitutive upregulation by chronic stimulation of the TSH/cAMP pathway plays a role in human and experimental hyperproliferative diseases of the thyroid gland. These conclusions are supported by in vitro and in vivo studies. In rat thyrocytes (PC Cl 3 cells), cyclin D3 expression is enhanced in response to activation of the TSH/cAMP pathway. Interference with the expression of G1 cyclins (in particular cyclin D3) by the antisense methodology strongly reduced TSH-dependent proliferation of PC Cl 3 cells, indicating that proper progression through G1 requires cyclin D3. Accordingly, PC Cl 3 cells engineered to overexpress cyclin D3 (PC-D3 cells) show enhanced growth rate and elude hormone-dependence and contact inhibition. Using an animal experimental model of thyroid stimulation, we demonstrate that cyclin D3 is a key mediator of TSH-dependent proliferation of thyroid follicular cells also in vivo. Cyclin D3 protein levels were higher in the thyrocytes from glands of propylthiouracil-treated rats compared with control animals. The increase in cyclin D3 expression occurred after the propylthiouracil-induced increase in TSH levels and preceded the burst of cell proliferation. Finally, we found that cyclin D3 protein is expressed in a fraction of human goiters but it is strongly overexpressed in most follicular adenomas.
- Published
- 2003
- Full Text
- View/download PDF
41. Understanding p27(kip1) deregulation in cancer: down-regulation or mislocalization.
- Author
-
Viglietto G, Motti ML, and Fusco A
- Subjects
- Animals, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Cell Transformation, Neoplastic genetics, Cyclin-Dependent Kinase Inhibitor p27, Cytoplasm enzymology, Humans, Neoplasms genetics, Tumor Suppressor Proteins genetics, Cell Compartmentation physiology, Cell Cycle physiology, Cell Transformation, Neoplastic metabolism, Down-Regulation physiology, Neoplasms enzymology, Tumor Suppressor Proteins deficiency
- Abstract
There is considerable evidence that the inactivation of the cyclin-dependent kinase inhibitor p27(kip1) is a fundamental step for the development of human malignancies. In particular, reduced expression of p27(kip1), due to increased protein degradation, correlates with poor prognosis of patients affected by various types of cancer. The purpose of this mini-review is to present an overview of the current understanding of the alteration of p27(kip1) function in human cancer and to describe the different mechanisms that contributes to it. Particular emphasis is placed on the novel finding of p27(kip1) mislocalization in tumor cells and on the biochemical pathways responsible for p27(kip1) cytosolic accumulation. Finally, we review the possible clinical implications of these observations with respect to prognosis and novel anticancer therapies.
- Published
- 2002
- Full Text
- View/download PDF
42. Cytoplasmic relocalization and inhibition of the cyclin-dependent kinase inhibitor p27(Kip1) by PKB/Akt-mediated phosphorylation in breast cancer.
- Author
-
Viglietto G, Motti ML, Bruni P, Melillo RM, D'Alessio A, Califano D, Vinci F, Chiappetta G, Tsichlis P, Bellacosa A, Fusco A, and Santoro M
- Subjects
- Breast Neoplasms pathology, Cell Cycle Proteins genetics, Cell Line, Cell Nucleus metabolism, Cyclin-Dependent Kinase Inhibitor p27, Enzyme Inhibitors metabolism, Female, Genes, Tumor Suppressor, Humans, Phosphatidylinositol 3-Kinases metabolism, Phosphorylation, Proto-Oncogene Proteins c-akt, Threonine metabolism, Tumor Suppressor Proteins genetics, Breast Neoplasms metabolism, Cell Cycle Proteins metabolism, Cyclin-Dependent Kinases antagonists & inhibitors, Protein Serine-Threonine Kinases, Proto-Oncogene Proteins metabolism, Tumor Suppressor Proteins metabolism
- Abstract
The cyclin-dependent kinase inhibitor p27(kip1) is a putative tumor suppressor for human cancer. The mechanism underlying p27(kip1) deregulation in human cancer is, however, poorly understood. We demonstrate that the serine/threonine kinase Akt regulates cell proliferation in breast cancer cells by preventing p27(kip1)-mediated growth arrest. Threonine 157 (T157), which maps within the nuclear localization signal of p27(kip1), is a predicted Akt-phosphorylation site. Akt-induced T157 phosphorylation causes retention of p27(kip1) in the cytoplasm, precluding p27(kip1)-induced G1 arrest. Conversely, the p27(kip1)-T157A mutant accumulates in cell nuclei and Akt does not affect p27(kip1)-T157A-mediated cell cycle arrest. Lastly, T157-phosphorylated p27(kip1) accumulates in the cytoplasm of primary human breast cancer cells coincident with Akt activation. Thus, cytoplasmic relocalization of p27(kip1), secondary to Akt-mediated phosphorylation, is a novel mechanism whereby the growth inhibitory properties of p27(kip1) are functionally inactivated and the proliferation of breast cancer cells is sustained.
- Published
- 2002
- Full Text
- View/download PDF
43. Characterization of 2 novel and 2 recurring BRCA1 germline mutations in breast and/or ovarian carcinoma patients from the area of Naples.
- Author
-
Curci A, Capasso I, Romano A, Bruni P, Motti ML, Pignata S, D'Aiuto G, Casamassimi A, D'Urso M, Fusco A, and Viglietto G
- Subjects
- Adult, Age Factors, Aged, Family Health, Female, Humans, Italy, Male, Middle Aged, Pedigree, Polymorphism, Single-Stranded Conformational, Sequence Analysis, DNA, Breast Neoplasms genetics, Genes, BRCA1, Mutation, Ovarian Neoplasms genetics
- Abstract
We have analyzed 18 families with high incidence of breast cancer or breast and ovarian cancer for the presence of BRCA1 mutations. We identified 4 mutations in the BRCA1 gene in 4 unrelated probands who belong to families with at least 1 case of breast and 1 case of ovarian cancer. Two of the mutations reported in this study are novel (GAA(1172)-->TAA in family Naples 14, GAA(1765)-->TAA in family Naples 20) whereas the others are already present in the Breast Cancer Information Core Electronic Database (http://nchgr.nih.gov/ Intramural research/Lab transfer/Bic/) (5382insC in family Naples 18 and 2080delA in family Naples 19). Conversely, no mutation in the BRCA1 gene was detected in 14 families characterized by 2 or more cases of breast cancer only, even if bilateral and with early-onset. These results indicate that germline mutations in the BRCA1 gene highly predispose for a cancer syndrome that involves the presence of both breast and ovarian cancer.
- Published
- 2002
- Full Text
- View/download PDF
44. Glial cell line-derived neurotrophic factor induces proliferative inhibition of NT2/D1 cells through RET-mediated up-regulation of the cyclin-dependent kinase inhibitor p27(kip1).
- Author
-
Baldassarre G, Bruni P, Boccia A, Salvatore G, Melillo RM, Motti ML, Napolitano M, Belletti B, Fusco A, Santoro M, and Viglietto G
- Subjects
- Animals, Antigens, Tumor-Associated, Carbohydrate, Cell Differentiation, Cell Division drug effects, Cell Line, Cyclin-Dependent Kinase Inhibitor p27, Enzyme Activation, Glial Cell Line-Derived Neurotrophic Factor, Glial Cell Line-Derived Neurotrophic Factor Receptors, Glycosphingolipids analysis, Neurons physiology, Proto-Oncogene Proteins c-ret, Stage-Specific Embryonic Antigens, Up-Regulation, Cell Cycle Proteins biosynthesis, Drosophila Proteins, Nerve Growth Factors, Nerve Tissue Proteins pharmacology, Proto-Oncogene Proteins physiology, Receptor Protein-Tyrosine Kinases physiology, Tumor Suppressor Proteins biosynthesis
- Abstract
Growth factors of the glial cell line-derived neurotrophic factor (GDNF) family control the differentiation of neuronal cells of the central and peripheral nervous systems. Intracellular signalling of these growth factors is, at least in part, mediated by activation of the RET receptor tyrosine kinase. Here, we demonstrate that GDNF triggering inhibits the proliferation of the embryonal carcinoma cell line NT2/D1. This anti-proliferative effect is accompanied by down-regulation of the SSEA-3 antigen, a marker typical of undifferentiated NT2/D1 cells. We show that these effects are mediated by activation of RET signalling. The block of RET by a kinase-deficient dominant negative mutant impairs GDNF-dependent growth inhibition, whereas the adoptive expression of a constitutively active RET, the RET-MEN2A oncogene, promotes effects similar to those exerted by GDNF. We show that RET signalling increases the expression of the cyclin-dependent kinase inhibitor p27(kip1) in NT2/D1 cells. Both DNA synthesis inhibition and SSEA-3 down-regulation are prevented if p27(kip1) expression is blocked by an antisense construct, which demonstrates that RET-triggered effects are mediated by p27(kip1).
- Published
- 2002
- Full Text
- View/download PDF
45. Detection of occult melanoma cells in paraffin-embedded histologically negative sentinel lymph nodes using a reverse transcriptase polymerase chain reaction assay.
- Author
-
Palmieri G, Ascierto PA, Cossu A, Mozzillo N, Motti ML, Satriano SM, Botti G, Caracò C, Celentano E, Satriano RA, Lissia A, Tanda F, Pirastu M, and Castello G
- Subjects
- Biomarkers, Tumor analysis, DNA, Neoplasm analysis, False Negative Reactions, Humans, Lymphatic Metastasis diagnosis, Melanoma pathology, Paraffin Embedding, Prognosis, Sensitivity and Specificity, Skin Neoplasms pathology, Specimen Handling, Melanoma diagnosis, Reverse Transcriptase Polymerase Chain Reaction standards, Sentinel Lymph Node Biopsy, Skin Neoplasms diagnosis
- Abstract
Purpose: Detection of occult metastasis before the development of clinical disease could allow more accurate staging, appropriate follow-up procedures, and adjuvant therapies in patients with malignant melanoma (MM). The sentinel lymph node (SLN) has been proposed as a reliable predictor of metastatic disease in the lymphatic basin draining the primary melanoma. In this study, we screened both paraffin-embedded SLNs and peripheral-blood (PB) samples from MM patients at various stage of disease using a multimarker reverse transcriptase polymerase chain reaction (RT-PCR) assay. The prognostic significance of the presence of PCR-positive markers was also evaluated., Patients and Methods: Total RNA was obtained from paraffin-embedded SLN sections and PB samples of 75 MM patients. RT-PCR was performed using tyrosinase and MelanA/MART1 as melanoma-associated markers. Radiolabeled PCR products were analyzed on denaturing polyacrylamide gels., Results: Good sensitivity of the RT-PCR assay on archival tissues was demonstrated after comparison of RT-PCR results on frozen and paraffin-embedded SLNs from 16 MM patients. Significant correlation between the disease stage and marker expression in both PB and SLN samples was observed; the highest value was for patients who were positive for both markers in SLN (P =.006). Progression of disease was significantly associated with the total number of PCR-positive markers in both PB (P =.034) and SLN (P =.001) samples., Conclusion: Although sensitivity is lowered by the use of paraffin-embedded specimens, our data indicate that RT-PCR analysis of serial sections from archival SLNs may be helpful in improving detection of occult micrometastases, thus improving staging of patients with melanoma.
- Published
- 2001
- Full Text
- View/download PDF
46. Clinical significance of PCR-positive mRNA markers in peripheral blood and regional nodes of malignant melanoma patients. Melanoma Cooperative Group.
- Author
-
Palmieri G, Pirastu M, Strazzullo M, Ascierto PA, Satriano SM, Motti ML, Botti G, Mozzillo N, Castello G, Cossu A, Lissia A, and Tanda F
- Subjects
- Biomarkers, Tumor genetics, Blotting, Southern, Disease Progression, Follow-Up Studies, Humans, Neoplasm Proteins genetics, Neoplasm Staging, Reverse Transcriptase Polymerase Chain Reaction, Biomarkers, Tumor blood, Lymph Nodes pathology, Melanoma blood, Neoplasm Proteins blood, Neoplastic Cells, Circulating, RNA, Messenger analysis, Skin Neoplasms blood
- Abstract
Reverse-transcriptase polymerase chain reaction (RT-PCR) with multiple markers has been demonstrated to be highly sensitive in detecting metastatic cells in peripheral blood of malignant melanoma (MM) patients, and the circulating MM cells to be significantly correlated with disease stages. We further evaluated the presence of specific PCR-positive mRNA markers in peripheral blood as well as in regional nodes as an expression of tumor progression. Peripheral blood samples from 317 MM patients with either localized (n = 219) or metastatic (n = 98) disease were processed to obtain total cellular RNA. RT-PCR was performed using tyrosinase (TYR), p97, and MelanA/MART1 as mRNA markers. PCR products were analyzed by gel electrophoresis and Southern blot hybridization. In addition, paraffin-embedded samples of histologically proven tumor-negative lymph nodes from the subset of patients with localized disease were analyzed by RT-PCR, using radiolabeled primers for TYR and MelanA/MART1. The presence of mRNA markers was significantly correlated with tumor burden with a good correlation between risk of recurrence (evaluated in stage I-III patients) and increasing number of PCR-positive markers (p = 0.0002). Currently, for each patient, PCR results obtained at different times during follow-up are being analyzed, and any variation in the number of PCR-positive markers is being correlated to the clinical status. Molecular screening of histologically negative nodes for the presence of metastatic MM cells is also under evaluation. Preliminary assessment of a subset of MM patients with higher risk of recurrence will require longer follow-up in order to define the role of RT-PCR in monitoring these patients.
- Published
- 2001
- Full Text
- View/download PDF
47. Retinoic acid induces neuronal differentiation of embryonal carcinoma cells by reducing proteasome-dependent proteolysis of the cyclin-dependent inhibitor p27.
- Author
-
Baldassarre G, Boccia A, Bruni P, Sandomenico C, Barone MV, Pepe S, Angrisano T, Belletti B, Motti ML, Fusco A, and Viglietto G
- Subjects
- Antineoplastic Agents therapeutic use, Carcinoma, Embryonal genetics, Carcinoma, Embryonal metabolism, Cell Differentiation drug effects, Cyclin-Dependent Kinase Inhibitor p27, Cyclin-Dependent Kinases antagonists & inhibitors, Cysteine Endopeptidases genetics, Cysteine Endopeptidases metabolism, Gene Expression Regulation, Neoplastic drug effects, Humans, Microtubule-Associated Proteins genetics, Microtubule-Associated Proteins metabolism, Multienzyme Complexes genetics, Multienzyme Complexes metabolism, Neurons metabolism, Proteasome Endopeptidase Complex, Tretinoin therapeutic use, Tumor Cells, Cultured, Antineoplastic Agents pharmacology, Carcinoma, Embryonal drug therapy, Carcinoma, Embryonal pathology, Cell Cycle Proteins, Neurons pathology, Tretinoin pharmacology, Tumor Suppressor Proteins
- Abstract
Retinoic acid (RA) treatment of embryonal carcinoma cell line NTERA-2 clone D1 (NT2/D1) induces growth arrest and terminal differentiation along the neuronal pathway. In the present study, we provide a functional link between RA and p27 function in the control of neuronal differentiation in NT2/D1 cells. We report that RA enhances p27 expression, which results in increased association with cyclin E/cyclin-dependent kinase 2 complexes and suppression of their activity; however, antisense clones, which have greatly reduced RA-dependent p27 inducibility (NT2-p27AS), continue to synthesize DNA and are unable to differentiate properly in response to RA as determined by lack of neurite outgrowth and by the failure to modify surface antigens. As to the mechanism involved in RA-dependent p27 upregulation, our data support the concept that RA reduces p27 protein degradation through the ubiquitin/proteasome-dependent pathway. Taken together, these findings demonstrate that in embryonal carcinoma cells, p27 expression is required for growth arrest and proper neuronal differentiation.
- Published
- 2000
48. Low doses interferon-alpha in the treatment of high-risk cutaneous melanoma. Melanoma Cooperative Group.
- Author
-
Ascierto PA, Palmieri G, Strazzullo M, Daponte A, Botti G, Satriano SM, Motti ML, and Mozzillo N
- Subjects
- Adult, Disease-Free Survival, Humans, Lymphatic Metastasis, Male, Melanoma secondary, Risk Factors, Skin Neoplasms pathology, Antineoplastic Agents therapeutic use, Interferon-alpha therapeutic use, Melanoma drug therapy, Skin Neoplasms drug therapy
- Published
- 2000
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.