Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Giusy Tornillo is active.

Publication


Featured researches published by Giusy Tornillo.


Advances in Experimental Medicine and Biology | 2010

Integrins and Signal Transduction

Sara Cabodi; Paola Di Stefano; Maria del Pilar Camacho Leal; Agata Tinnirello; Brigitte Bisaro; Virginia Morello; Laura Damiano; Simona Aramu; Daniele Repetto; Giusy Tornillo; Paola Defilippi

Integrin signaling has a critical function in organizing cells in tissues during both embryonic development and tissue repair. Following their binding to the extracellular ligands, the intracellular signaling pathways triggered by integrins are directed to two major functions: organization of the actin cytoskeleton and regulation of cell behaviour including survival, differentiation and growth. Basic research conducted in the past twelve years has lead to remarkable breakthroughs in this field. Integrins are catalytically inactive and translate positional cues into biochemical signals by direct and/or functional association with intracellular adaptors, cytosolic tyrosine kinases or growth factor and cytokine receptors. The purpose of this chapter is to highlight recent experimental and conceptual advances in integrin signaling with particular emphasis on the ability of integrins to regulate Fak/Src family kinases (SFKs) activation and the cross-talk with soluble growth factors receptors and cytokines.


The FASEB Journal | 2010

p130Cas is an essential transducer element in ErbB2 transformation

Sara Cabodi; Agata Tinnirello; Brigitte Bisaro; Giusy Tornillo; Maria del Pilar Camacho-Leal; Guido Forni; Rodica Cojoca; Manuela Iezzi; Augusto Amici; Maura Montani; Alessandra Eva; Paola Di Stefano; Senthil K. Muthuswamy; Guido Tarone; Emilia Turco; Paola Defilippi

The ErbB2 oncogene is often overexpressed in breast tumors and associated with poor clinical outcome. p130Cas represents a nodal scaffold protein regulating cell survival, migration, and proliferation in normal and pathological cells. The functional role of p130Cas in ErbB2‐dependent breast tumorigenesis was assessed by its silencing in breast cancer cells derived from mouse mammary tumors overexpressing ErbB2 (N202‐1A cells), and by its reexpression in ErbB2‐transformed p130Cas‐null mouse embryonic fibroblasts. We demonstrate that p130Cas is necessary for ErbB2‐dependent foci formation, anchorage‐independent growth, and in vivo growth of orthotopic N202‐1A tumors. Moreover, intranipple injection of p130Cas‐stabilized siRNAs in the mammary gland of Balbc‐NeuT mice decreases the growth of spontaneous tumors. In ErbB2‐transformed cells, p130Cas is a crucial component of a functional molecular complex consisting of ErbB2, c‐Src, and Fak. In human mammary cells, MCF10A.B2, the concomitant activation of ErbB2, and p130Cas overexpression sustain and strengthen signaling, leading to Rac1 activation and MMP9 secretion, thus providing invasive properties. Consistently, p130Cas drives N202‐1A cell in vivo lung metastases colonization. These results demonstrate that p130Cas is an essential transducer in ErbB2 transformation and highlight its potential use as a novel therapeutic target in ErbB2 positive human breast cancers.—Cabodi, S., Tinnirello, A., Bisaro, B., Tornillo, G., Camacho‐Leal, M. P., Forni, G., Cojoca, R., Iezzi, M., Amici, A., Montani, M., Eva, A., Di Stefano, P., Muthuswamy, S. K., Tarone, G., Turco, E., Defilippi, P. p130Cas is an essential transducer element in ErbB2 transformation. FASEB J. 24, 3796–3808 (2010). www.fasebj.org


European Journal of Cell Biology | 2011

p130Cas promotes invasiveness of three-dimensional ErbB2-transformed mammary acinar structures by enhanced activation of mTOR/p70S6K and Rac1

Giusy Tornillo; Brigitte Bisaro; Maria del Pilar Camacho-Leal; Mirco Galiè; Paolo Provero; Paola Di Stefano; Emilia Turco; Paola Defilippi; Sara Cabodi

ErbB2 over-expression is detected in approximately 25% of invasive breast cancers and is strongly associated with poor patient survival. We have previously demonstrated that p130Cas adaptor is a crucial mediator of ErbB2 transformation. Here, we analysed the molecular mechanisms through which p130Cas controls ErbB2-dependent invasion in three-dimensional cultures of mammary epithelial cells. Concomitant p130Cas over-expression and ErbB2 activation enhance PI3K/Akt and Erk1/2 MAPK signalling pathways and promote invasion of mammary acini. By using pharmacological inhibitors, we demonstrate that both signalling cascades are required for the invasive behaviour of p130Cas over-expressing and ErbB2 activated acini. Erk1/2 MAPK and PI3K/Akt signalling triggers invasion through distinct downstream effectors involving mTOR/p70S6K and Rac1 activation, respectively. Moreover, in silico analyses indicate that p130Cas expression in ErbB2 positive human breast cancers significantly correlates with higher risk to develop distant metastasis, thus underlying the value of the p130Cas/ErbB2 synergism in regulating breast cancer invasion. In conclusion, high levels of p130Cas favour progression of ErbB2-transformed cells towards an invasive phenotype.


Breast Cancer Research | 2012

p130Cas/Cyclooxygenase-2 axis in the control of mesenchymal plasticity of breast cancer cells

Brigitte Bisaro; Maura Montani; Georgia Konstantinidou; Cristina Marchini; Lucia Pietrella; Manuela Iezzi; Mirco Galiè; Francesca Orso; Annalisa Camporeale; Shana Colombo; Paola Di Stefano; Giusy Tornillo; Maria del Pilar Camacho-Leal; Emilia Turco; Daniela Taverna; Sara Cabodi; Augusto Amici; Paola Defilippi

IntroductionIntrinsic plasticity of breast carcinoma cells allows them to undergo a transient and reversible conversion into mesenchymal cells to disseminate into distant organs, where they can re-differentiate to an epithelial-like status to form a cohesive secondary mass. The p130Cas scaffold protein is overexpressed in human ER+ and HER2+ breast cancer where it contributes to cancer progression, invasion and resistance to therapy. However, its role in regulating mesenchymal aggressive breast cancer cells remains to be determined. The aim of this study was to investigate the molecular and functional involvement of this adaptor protein in breast cancer cell plasticity.MethodsWe used silencing strategies and rescue experiments to evaluate phenotypic and biochemical changes from mesenchymal to epithelial traits in breast tumor cell lines. In the mouse A17 cell model previously related to mesenchymal cancer stem cells and basal-like breast cancer, we biochemically dissected the signaling pathways involved and performed functional in vivo tumor growth ability assays. The significance of the signaling platform was assessed in a human setting through the use of specific inhibitors in aggressive MDA-MB-231 subpopulation LM2-4175 cells. To evaluate the clinical relevance of the results, we analyzed publicly available microarray data from the Netherlands Cancer Institute and from the Koo Foundation Sun Yat-Sen Cancer Center.ResultsWe show that p130Cas silencing induces loss of mesenchymal features, by downregulating Vimentin, Snail, Slug and Twist transcriptional factors, resulting in the acquirement of epithelial-like traits. Mechanistically, p130Cas controls Cyclooxygenase-2 transcriptional expression, which in turn contributes to p130Cas-dependent maintenance of mesenchymal phenotype. This cascade of events also compromises in vivo tumor growth through inhibition of cell signaling controlling cell cycle progression. c-Src and JNK kinases are sequential players in p130Cas/ Cyclooxygenase-2 axis and their pharmacological inhibition is sufficient to downregulate Cyclooxygenase-2 leading to an epithelial phenotype. Finally, in silico microarray data analysis indicates that p130Cas and Cyclooxygenase-2 concomitant overexpression predicts poor survival and high probability of breast tumor recurrence.ConclusionsOverall, these data identify a new p130Cas/Cyclooxygenase-2 axis as a crucial element in the control of breast tumor plasticity, opening new therapeutic strategies leading to inhibition of these pathways in aggressive breast carcinoma.


Cell Cycle | 2013

Identification of p130Cas/ErbB2-dependent invasive signatures in transformed mammary epithelial cells

Alessandra Pincini; Giusy Tornillo; Francesca Orso; Marianna Sciortino; Brigitte Bisaro; Maria del Pilar Camacho Leal; Antonio Lembo; Maria Felice Brizzi; Emilia Turco; Cristiano De Pittà; Paolo Provero; Enzo Medico; Paola Defilippi; Daniela Taverna; Sara Cabodi

Understanding transcriptional changes during cancer progression is of crucial importance to develop new and more efficacious diagnostic and therapeutic approaches. It is well known that ErbB2 is overexpressed in about 25% of human invasive breast cancers. We have previously demonstrated that p130Cas overexpression synergizes with ErbB2 in mammary cell transformation and promotes ErbB2-dependent invasion in three-dimensional (3D) cultures of human mammary epithelial cells. Here, by comparing coding and non-coding gene expression profiles, we define the invasive signatures associated with concomitant p130Cas overexpression and ErbB2 activation in 3D cultures of mammary epithelial cells. Specifically, we have found that genes involved in amino acids synthesis (CBS, PHGDH), cell motility, migration (ITPKA, PRDM1), and angiogenesis (HEY1) are upregulated, while genes involved in inflammatory response (SAA1, S100A7) are downregulated. In parallel, we have shown that the expression of specific miRNAs is altered. Among these, miR-200b, miR-222, miR-221, miR-R210, and miR-424 are upregulated, while miR-27a, miR-27b, and miR-23b are downregulated. Overall, this study presents, for the first time, the gene expression changes underlying the invasive behavior following p130Cas overexpression in an ErbB2 transformed mammary cell model.


The Journal of Pathology | 2014

Morgana acts as a proto‐oncogene through inhibition of a ROCK–PTEN pathway

Federica Fusella; Roberta Ferretti; Daniele Recupero; Stefania Rocca; Augusta Di Savino; Giusy Tornillo; Lorenzo Silengo; Emilia Turco; Sara Cabodi; Paolo Provero; Pier Paolo Pandolfi; Anna Sapino; Guido Tarone; Mara Brancaccio

Morgana/CHP‐1 is a ubiquitously expressed protein able to inhibit ROCK II kinase activity. We have previously demonstrated that morgana haploinsufficiency leads to multiple centrosomes, genomic instability, and higher susceptibility to tumour development. While a large fraction of human cancers has shown morgana down‐regulation, a small subset of tumours was shown to express high morgana levels. Here we demonstrate that high morgana expression in different breast cancer subtypes correlates with high tumour grade, mitosis number, and lymph node positivity. Moreover, morgana overexpression induces transformation in NIH‐3T3 cells and strongly protects them from various apoptotic stimuli. From a mechanistic point of view, we demonstrate that morgana causes PTEN destabilization, by inhibiting ROCK activity, hence triggering the PI3K/AKT survival pathway. In turn, morgana down‐regulation in breast cancer cells that express high morgana levels increases PTEN expression and leads to sensitization of cells to chemotherapy. Copyright


Stem Cells | 2013

P130Cas alters the differentiation potential of mammary luminal progenitors by deregulating C-Kit activity

Giusy Tornillo; Angela Rita Elia; Isabella Castellano; Michela Spadaro; Paola Bernabei; Brigitte Bisaro; Maria del Pilar Camacho-Leal; Alessandra Pincini; Paolo Provero; Anna Sapino; Emilia Turco; Paola Defilippi; Sara Cabodi

It has recently been proposed that defective differentiation of mammary luminal progenitors predisposes to basal‐like breast cancer. However, the molecular and cellular mechanisms involved are still unclear. Here, we describe that the adaptor protein p130Cas is a crucial regulator of mouse mammary epithelial cell (MMEC) differentiation. Using a transgenic mouse model, we show that forced p130Cas overexpression in the luminal progenitor cell compartment results in the expansion of luminal cells, which aberrantly display basal cell features and reduced differentiation in response to lactogenic stimuli. Interestingly, MMECs overexpressing p130Cas exhibit hyperactivation of the tyrosine kinase receptor c‐Kit. In addition, we demonstrate that the constitutive c‐Kit activation alone mimics p130Cas overexpression, whereas c‐Kit downregulation is sufficient to re‐establish proper differentiation of p130Cas overexpressing cells. Overall, our data indicate that high levels of p130Cas, via abnormal c‐Kit activation, promote mammary luminal cell plasticity, thus providing the conditions for the development of basal‐like breast cancer. Consistently, p130Cas is overexpressed in human triple‐negative breast cancer, further suggesting that p130Cas upregulation may be a priming event for the onset of basal‐like breast cancer. STEM Cells2013;31:1422–1433


Stem Cells | 2014

Nuclear Akt2 opposes limbal keratinocyte stem cell self-renewal by repressing a FOXO-mTORC1 signaling pathway.

Stefania Saoncella; Beatrice Tassone; Erika Deklic; Fabio Avolio; Cristina Jon; Giusy Tornillo; Elisa De Luca; Enzo Di Iorio; Roberto Piva; Sara Cabodi; Emilia Turco; Pier Paolo Pandolfi; Enzo Calautti

Signals downstream of Akt can either favor or oppose stem cell (SC) maintenance, but how this dual role can be achieved is still undefined. Using human limbal keratinocyte stem cells (LKSCs), a SC type used in transplantation therapies for corneal regeneration, we show that Akt signaling is prominent in SC populations both in vivo and in vitro, and that Akt1 promotes while Akt2 opposes SC self‐renewal. Noteworthy, loss of Akt2 signaling enhances LKSC maintenance ex vivo, whereas Akt1 depletion anticipates SC exhaustion. Mechanistically, the antagonistic functions of Akt1 and Akt2 in SC control are mainly dictated by their differential subcellular distribution, being nuclear Akt2 selectively implicated in FOXO inhibition. Akt2 downregulation favors LKSC maintenance as a result of a gain of FOXO functions, which attenuates the mechanistic target of rapamycin complex one signaling via tuberous sclerosis one gene induction, and promotes growth factor signaling through Akt1. Consistently, Akt2 deficiency also enhances limbal SCs in vivo. Thus, our findings reveal distinct roles for nuclear versus cytosolic Akt signaling in normal epithelial SC control and suggest that the selective Akt2 inhibition may provide novel pharmacological strategies for human LKSC expansion in therapeutic settings and mechanistic research. Stem Cells 2014;32:754–769


PLOS ONE | 2012

p130Cas over-expression impairs mammary branching morphogenesis in response to estrogen and EGF.

Maria del Pilar Camacho Leal; Alessandra Pincini; Giusy Tornillo; Elisa Fiorito; Brigitte Bisaro; Elisa Di Luca; Emilia Turco; Paola Defilippi; Sara Cabodi

p130Cas adaptor protein regulates basic processes such as cell cycle control, survival and migration. p130Cas over-expression has been related to mammary gland transformation, however the in vivo consequences of p130Cas over-expression during mammary gland morphogenesis are not known. In ex vivo mammary explants from MMTV-p130Cas transgenic mice, we show that p130Cas impairs the functional interplay between Epidermal Growth Factor Receptor (EGFR) and Estrogen Receptor (ER) during mammary gland development. Indeed, we demonstrate that p130Cas over-expression upon the concomitant stimulation with EGF and estrogen (E2) severely impairs mammary morphogenesis giving rise to enlarged multicellular spherical structures with altered architecture and absence of the central lumen. These filled acinar structures are characterized by increased cell survival and proliferation and by a strong activation of Erk1/2 MAPKs and Akt. Interestingly, antagonizing the ER activity is sufficient to re-establish branching morphogenesis and normal Erk1/2 MAPK activity. Overall, these results indicate that high levels of p130Cas expression profoundly affect mammary morphogenesis by altering epithelial architecture, survival and unbalancing Erk1/2 MAPKs activation in response to growth factors and hormones. These results suggest that alteration of morphogenetic pathways due to p130Cas over-expression might prime mammary epithelium to tumorigenesis.


Archive | 2011

p130Cas overexpression regulates normal mammary branching morphogenesis through estrogen receptor-alpha/ERk1/2 dependent signaling

M. P. Camacho Leal; E. Fiorito; Alessandra Pincini; Giusy Tornillo

Resumen del poster presentado al 36th FEBS Congress celebrado en Torino (Italia) del 25 al 30 de Junio de 2011.-- et al.

Collaboration


Dive into the Giusy Tornillo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge