Network


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

Hotspot


Dive into the research topics where Manuela Vecchi is active.

Publication


Featured researches published by Manuela Vecchi.


Cell | 2010

Biological and Molecular Heterogeneity of Breast Cancers Correlates with Their Cancer Stem Cell Content

Salvatore Pece; Daniela Tosoni; Stefano Confalonieri; Giovanni Mazzarol; Manuela Vecchi; Simona Ronzoni; Loris Bernard; Giuseppe Viale; Pier Giuseppe Pelicci; Pier Paolo Di Fiore

Pathways that govern stem cell (SC) function are often subverted in cancer. Here, we report the isolation to near purity of human normal mammary SCs (hNMSCs), from cultured mammospheres, on the basis of their ability to retain the lipophilic dye PKH26 as a consequence of their quiescent nature. PKH26-positive cells possess all the characteristics of hNMSCs. The transcriptional profile of PKH26-positive cells (hNMSC signature) was able to predict biological and molecular features of breast cancers. By using markers of the hNMSC signature, we prospectively isolated SCs from the normal gland and from breast tumors. Poorly differentiated (G3) cancers displayed higher content of prospectively isolated cancer SCs (CSCs) than did well-differentiated (G1) cancers. By comparing G3 and G1 tumors in xenotransplantation experiments, we directly demonstrated that G3s are enriched in CSCs. Our data support the notion that the heterogeneous phenotypical and molecular traits of human breast cancers are a function of their CSC content.


Journal of Biological Chemistry | 1996

Selective Cleavage of the Heregulin Receptor ErbB-4 by Protein Kinase C Activation

Manuela Vecchi; Josep Baulida; Graham Carpenter

The 180-kDa transmembrane tyrosine kinase ErbB-4 is a receptor for the growth factor heregulin. 125I-Heregulin binding to NIH 3T3 cells overexpressing the ErbB-4 receptor is rapidly decreased by 12-O-tetradecanoylphorbol-13-acetate (TPA) pretreatment. Immunologic analysis demonstrates that TPA treatment of cells induces the proteolytic cleavage of ErbB-4, producing an 80-kDa cytoplasmic domain fragment, which contains a low level of phosphotyrosine, and a 120-kDa ectodomain fragment, which is released into the extracellular medium. Cleavage of ErbB-4 was also enhanced by other protein kinase C activators, i.e. platelet-derived growth factor, ionomycin, and synthetic diacylglycerol, while protein kinase C inhibition or down-regulation suppressed the TPA stimulation of ErbB-4 degradation. TPA did not induce the degradation of related receptors (ErbB-1, ErbB-2, and ErbB-3) in the EGF receptor family. The phorbol ester-induced cleavage of ErbB-4 occurs within or close to the ectodomain, as the 80-kDa cytoplasmic domain fragment is recognized by antibody to the ErbB-4 carboxyl terminus and is membrane-associated. Coprecipitation experiments show that, while the 80-kDa ErbB-4 fragment is associated with the SH2-containing molecules PLC-γ1 and Shc, TPA did not induce the phosphorylation of these substrates in intact cells. In addition, kinase assays in vitro indicate that the 80-kDa fragment is not an active tyrosine kinase. These results show that protein kinase C negatively regulates heregulin signaling through the ErbB-4 receptor by the activation of a selective proteolytic mechanism.


Molecular and Cellular Biology | 2004

Np95 Is a Histone-Binding Protein Endowed with Ubiquitin Ligase Activity

Elisabetta Citterio; Roberto Papait; Francesco Nicassio; Manuela Vecchi; Paola Gomiero; Roberto Mantovani; Pier Paolo Di Fiore; Ian Marc Bonapace

ABSTRACT Np95 is an important determinant in cell cycle progression. Its expression is tightly regulated and becomes detectable shortly before the entry of cells into S phase. Accordingly, Np95 is absolutely required for the G1/S transition. Its continued expression throughout the S/G2/M phases further suggests additional roles. Indeed, Np95 has been implicated in DNA damage response. Here, we show that Np95 is tightly bound to chromatin in vivo and that it binds to histones in vivo and in vitro. The binding to histones is direct and shows a remarkable preference for histone H3 and its N-terminal tail. A novel protein domain, the SRA-YDG domain, contained in Np95 is indispensable both for the interaction with histones and for chromatin binding in vivo. Np95 contains a RING finger. We show that this domain confers E3 ubiquitin ligase activity on Np95, which is specific for core histones, in vitro. Finally, Np95 shows specific E3 activity for histone H3 when the endogenous core octamer, coimmunoprecipitating with Np95, is used as a substrate. Histone ubiquitination is an important determinant in the regulation of chromatin structure and gene transcription. Thus, the demonstration that Np95 is a chromatin-associated ubiquitin ligase suggests possible molecular mechanisms for its action as a cell cycle regulator.


Journal of Clinical Investigation | 2007

Survival prediction of stage I lung adenocarcinomas by expression of 10 genes

Fabrizio Bianchi; Paolo Nuciforo; Manuela Vecchi; Loris Bernard; Laura Tizzoni; Antonio Marchetti; Fiamma Buttitta; Lara Felicioni; Francesco Nicassio; Pier Paolo Di Fiore

Adenocarcinoma is the predominant histological subtype of lung cancer, the leading cause of cancer deaths in the world. At stage I, the tumor is cured by surgery alone in about 60% of cases. Markers are needed to stratify patients by prognostic outcomes and may help in devising more effective therapies for poor prognosis patients. To achieve this goal, we used an integrated strategy combining meta-analysis of published lung cancer microarray data with expression profiling from an experimental model. The resulting 80-gene model was tested on an independent cohort of patients using RT-PCR, resulting in a 10-gene predictive model that exhibited a prognostic accuracy of approximately 75% in stage I lung adenocarcinoma when tested on 2 additional independent cohorts. Thus, we have identified a predictive signature of limited size that can be analyzed by RT-PCR, a technology that is easy to implement in clinical laboratories.


Molecular and Cellular Biology | 1999

Physiological Requirement for Both SH2 Domains for Phospholipase C-γ1 Function and Interaction with Platelet-Derived Growth Factor Receptors

Qun Sheng Ji; Ansuman Chattopadhyay; Manuela Vecchi; Graham Carpenter

ABSTRACT Two approaches have been utilized to investigate the role of individual SH2 domains in growth factor activation of phospholipase C-γ1 (PLC-γ1). Surface plasmon resonance analysis indicates that the individual N-SH2 and C-SH2 domains are able to specifically recognize a phosphotyrosine-containing peptide corresponding to Tyr 1021 of the platelet-derived growth factor (PDGF) β receptor. To assess SH2 function in the context of the full-length PLC-γ1 molecule as well as within the intact cell, PLC-γ1 SH2 domain mutants, disabled by site-directed mutagenesis of the N-SH2 and/or C-SH2 domain(s), were expressed in Plcg1−/− fibroblasts. Under equilibrium incubation conditions (4°C, 40 min), the N-SH2 domain, but not the C-SH2 domain, was sufficient to mediate significant PLC-γ1 association with the activated PDGF receptor and PLC-γ1 tyrosine phosphorylation. When both SH2 domains in PLC-γ1 were disabled, the double mutant did not associate with activated PDGF receptors and was not tyrosine phosphorylated. However, no single SH2 mutant was able to mediate growth factor activation of Ca2+mobilization or inositol 1,4,5-trisphosphate (IP3) formation. Subsequent kinetic experiments demonstrated that each single SH2 domain mutant was significantly impaired in its capacity to mediate rapid association with activated PDGF receptors and become tyrosine phosphorylated. Hence, when assayed under physiological conditions necessary to achieve a rapid biological response (Ca2+mobilization and IP3 formation), both SH2 domains of PLC-γ1 are essential to growth factor responsiveness.


Embo Molecular Medicine | 2013

The Fragile X Protein binds mRNAs involved in cancer progression and modulates metastasis formation

Rossella Luca; Michele Averna; Francesca Zalfa; Manuela Vecchi; Fabrizio Bianchi; Giorgio La Fata; Franca Del Nonno; Roberta Nardacci; Marco Bianchi; Paolo Nuciforo; Sebastian Munck; Paola Parrella; Rute Moura; Emanuela Signori; Robert D. Alston; Anna Kuchnio; Maria Giulia Farace; Vito Michele Fazio; Mauro Piacentini; Bart De Strooper; Tilmann Achsel; Giovanni Neri; Patrick Neven; D. Gareth Evans; Peter Carmeliet; Massimiliano Mazzone; Claudia Bagni

The role of the fragile X mental retardation protein (FMRP) is well established in brain, where its absence leads to the fragile X syndrome (FXS). FMRP is almost ubiquitously expressed, suggesting that, in addition to its effects in brain, it may have fundamental roles in other organs. There is evidence that FMRP expression can be linked to cancer. FMR1 mRNA, encoding FMRP, is overexpressed in hepatocellular carcinoma cells. A decreased risk of cancer has been reported in patients with FXS while a patient‐case with FXS showed an unusual decrease of tumour brain invasiveness. However, a role for FMRP in regulating cancer biology, if any, remains unknown. We show here that FMRP and FMR1 mRNA levels correlate with prognostic indicators of aggressive breast cancer, lung metastases probability and triple negative breast cancer (TNBC). We establish that FMRP overexpression in murine breast primary tumours enhances lung metastasis while its reduction has the opposite effect regulating cell spreading and invasion. FMRP binds mRNAs involved in epithelial mesenchymal transition (EMT) and invasion including E‐cadherin and Vimentin mRNAs, hallmarks of EMT and cancer progression.


Science Signaling | 2014

Memo Is a Copper-Dependent Redox Protein with an Essential Role in Migration and Metastasis

Gwen MacDonald; Ivan Nalvarte; Tatiana Smirnova; Manuela Vecchi; Nicola Aceto; Arno Doelemeyer; Anna Frei; Susanne Lienhard; Jeffrey Wyckoff; Daniel Hess; Jan Seebacher; Jeremy J. Keusch; Heinz Gut; Danièle Salaun; Giovanni Mazzarol; Davide Disalvatore; Mohamed Bentires-Alj; Pier Paolo Di Fiore; Ali Badache; Nancy E. Hynes

Copper chelators could help to reduce metastasis from breast tumors. Copper for Breast Cancer Metastasis Many patients with breast cancer die from metastases, the spread of cancer cells from the primary tumor to other sites. Activation of certain proteins by oxidation, a chemical modification involving reactive oxygen species, promotes the movement of cells from one location to another. MacDonald et al. discovered that the enzyme Memo bound copper and enhanced the oxidation of proteins involved in cell movement. Mice with tumors formed from breast cancer cells lacking Memo had fewer lung metastases. High Memo abundance predicted metastasis in breast cancer patients, and copper chelation therapy, which is in clinical trials for breast cancer treatment, may inhibit this metastatic process. Memo is an evolutionarily conserved protein with a critical role in cell motility. We found that Memo was required for migration and invasion of breast cancer cells in vitro and spontaneous lung metastasis from breast cancer cell xenografts in vivo. Biochemical assays revealed that Memo is a copper-dependent redox enzyme that promoted a more oxidized intracellular milieu and stimulated the production of reactive oxygen species (ROS) in cellular structures involved in migration. Memo was also required for the sustained production of the ROS O2− by NADPH (reduced form of nicotinamide adenine dinucleotide phosphate) oxidase 1 (NOX1) in breast cancer cells. Memo abundance was increased in >40% of the primary breast tumors tested, was correlated with clinical parameters of aggressive disease, and was an independent prognostic factor of early distant metastasis.


Oncogene | 2010

Copine III interacts with ERBB2 and promotes tumor cell migration

C Heinrich; C Keller; A Boulay; Manuela Vecchi; M Bianchi; R Sack; Susanne Lienhard; S Duss; J Hofsteenge; Nancy E. Hynes

ErbB2 amplification and overexpression in breast cancer correlates with aggressive disease and poor prognosis. To find novel ErbB2-interacting proteins, we used stable isotope labeling of amino acids in cell culture followed by peptide affinity pull-downs and identified specific binders using relative quantification by mass spectrometry. Copine-III, a member of a Ca2+-dependent phospholipid-binding protein family, was identified as binding to phosphorylated Tyr1248 of ErbB2. In breast cancer cells, Copine-III requires Ca2+ for binding to the plasma membrane, where it interacts with ErbB2 upon receptor stimulation, an interaction that is dependent on receptor activity. Copine-III also binds receptor of activated C kinase 1 and colocalizes with phosphorylated focal adhesion kinase at the leading edge of migrating cells. Importantly, knockdown of Copine-III in T47D breast cancer cells causes a decrease in Src kinase activation and ErbB2-dependent wound healing. Our data suggest that Copine-III is a novel player in the regulation of ErbB2-dependent cancer cell motility. In primary breast tumors, high CPNE3 RNA levels significantly correlate with ERBB2 amplification. Moreover, in an in situ tissue microarray analysis, we detected differential protein expression of Copine-III in normal versus breast, prostate and ovarian tumors, suggesting a more general role for Copine-III in carcinogenesis.


EMBO Reports | 2016

RAB2A controls MT1‐MMP endocytic and E‐cadherin polarized Golgi trafficking to promote invasive breast cancer programs

Hiroaki Kajiho; Yuko Kajiho; Emanuela Frittoli; Stefano Confalonieri; Giovanni Bertalot; Giuseppe Viale; Pier Paolo Di Fiore; Amanda Oldani; Massimiliano Garrè; Galina V. Beznoussenko; Andrea Palamidessi; Manuela Vecchi; Philippe Chavrier; Frank Perez; Giorgio Scita

The mechanisms of tumor cell dissemination and the contribution of membrane trafficking in this process are poorly understood. Through a functional siRNA screening of human RAB GTPases, we found that RAB2A, a protein essential for ER‐to‐Golgi transport, is critical in promoting proteolytic activity and 3D invasiveness of breast cancer (BC) cell lines. Remarkably, RAB2A is amplified and elevated in human BC and is a powerful and independent predictor of disease recurrence in BC patients. Mechanistically, RAB2A acts at two independent trafficking steps. Firstly, by interacting with VPS39, a key component of the late endosomal HOPS complex, it controls post‐endocytic trafficking of membrane‐bound MT1‐MMP, an essential metalloprotease for matrix remodeling and invasion. Secondly, it further regulates Golgi transport of E‐cadherin, ultimately controlling junctional stability, cell compaction, and tumor invasiveness. Thus, RAB2A is a novel trafficking determinant essential for regulation of a mesenchymal invasive program of BC dissemination.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Selective high-level expression of epsin 3 in gastric parietal cells, where it is localized at endocytic sites of apical canaliculi

Genevieve Ko; Summer Paradise; Hong Chen; Morven Graham; Manuela Vecchi; Fabrizio Bianchi; Ottavio Cremona; Pier Paolo Di Fiore; Pietro De Camilli

Epsin is a ubiquitin-binding endocytic adaptor, which is highly concentrated at clathrin-coated pits and coordinates acquisition of bilayer curvature with coat recruitment and cargo selection. Epsin is encoded by three distinct genes in mammals. Epsin 1 and 2 have broad tissue distribution with high-level expression in the brain. In contrast, epsin 3 was reported to be expressed primarily in immature keratinocytes. Here, we show that epsin 3 is selectively expressed at high levels in the stomach (including the majority of gastric cancers), where it is concentrated in parietal cells. In these cells, epsin 3 is enriched and colocalized with clathrin around apical canaliculi, the sites that control acidification of the stomach lumen via the exo-endocytosis of vesicles containing the H/K ATPase. Deletion of the epsin 3 gene in mice did not result in obvious pathological phenotypes in either the stomach or other organs, possibly because of overlapping functions of the other two epsins. However, levels of EHD1 and EHD2, two membrane tubulating proteins with a role in endocytic recycling, were elevated in epsin 3 knock-out stomachs, pointing to a functional interplay of epsin 3 with EHD proteins in the endocytic pathway of parietal cells. We suggest that epsin 3 cooperates with other bilayer binding proteins with curvature sensing/generating properties in the specialized traffic and membrane remodeling processes typical of gastric parietal cells.

Collaboration


Dive into the Manuela Vecchi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fabrizio Bianchi

European Institute of Oncology

View shared research outputs
Top Co-Authors

Avatar

Giovanni Bertalot

European Institute of Oncology

View shared research outputs
Top Co-Authors

Avatar

Giuseppe Viale

European Institute of Oncology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Francesco Nicassio

European Institute of Oncology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Elisa Dama

European Institute of Oncology

View shared research outputs
Top Co-Authors

Avatar

Flavia Troglio

European Institute of Oncology

View shared research outputs
Top Co-Authors

Avatar

Francesca Zalfa

University of Rome Tor Vergata

View shared research outputs
Researchain Logo
Decentralizing Knowledge