Network


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

Hotspot


Dive into the research topics where Giovanni Perconti is active.

Publication


Featured researches published by Giovanni Perconti.


Journal of Cellular Physiology | 2007

The PVT-1 oncogene is a myc protein target that is overexpressed in transformed cells

Letizia Carramusa; Flavia Contino; Arianna Ferro; Luigi Minafra; Giovanni Perconti; Agata Giallongo; Salvatore Feo

The human PVT‐1 gene is located on chromosome 8 telomeric to the c‐Myc gene and it is frequently involved in the translocations occurring in variant Burkitts lymphomas and murine plasmacytomas. It has been proposed that PVT‐1 regulates c‐Myc gene transcription over a long distance. To get new insights into the functional relationships between the two genes, we have investigated PVT‐1 and c‐Myc expression in normal human tissues and in transformed cells. Our findings indicate that PVT‐1 expression is restricted to a relative low number of normal tissues compared to the wide distribution of c‐Myc mRNA, whereas the gene is highly expressed in many transformed cell types including neuroblastoma cells that do not express c‐Myc. Reporter gene assays were used to dissect the PVT‐1 promoter and to identify the region responsible for the elevated expression observed in transformed cells. This region contains two putative binding sites for Myc proteins. The results of transfection experiments in RAT1‐MycER cells and chromatin immunoprecipitation (ChIP) assays in proliferating and differentiated neuroblastoma cells indicate that PVT‐1 is a downstream target of Myc proteins. J. Cell. Physiol. 213: 511–518, 2007.


International Journal of Cancer | 2009

An integrated humoral and cellular response is elicited in pancreatic cancer by α-enolase, a novel pancreatic ductal adenocarcinoma-associated antigen†

Paola Cappello; Barbara Tomaino; Roberto Chiarle; Patrizia Ceruti; Anna Novarino; Carlotta Castagnoli; Paola Migliorini; Giovanni Perconti; Agata Giallongo; Michele Milella; Vladia Monsurrò; Stefano Barbi; Aldo Scarpa; Paola Nisticò; Mirella Giovarelli; Francesco Novelli

Pancreatic ductal adenocarcinoma (PDAC) is a fatal disease with a very poor 5‐year survival rate. α‐Enolase is a glycolytic enzyme that also acts as a surface plasminogen receptor. We find that it is overexpressed in PDAC and present on the cell surface of PDAC cell lines. The clinical correlation of its expression with tumor status has been reported for lung and hepatocellular carcinoma. We have previously demonstrated that sera from PDAC patients contain IgG autoantibodies to α‐enolase. The present work was intended to assess the ability of α‐enolase to induce antigen‐specific T cell responses. We show that α‐enolase‐pulsed dendritic cells (DC) specifically stimulate healthy autologous T cells to proliferate, secrete IFN‐γ and lyse PDAC cells but not normal cells. In vivo, α‐enolase‐specific T cells inhibited the growth of PDAC cells in immunodeficient mice. In 8 out of 12 PDAC patients with circulating IgG to α‐enolase, the existence of α‐enolase‐specific T cells was also demonstrated. Taken as a whole, these results indicate that α‐enolase elicits a PDAC‐specific, integrated humoral and cellular response. It is thus a promising and clinically relevant molecular target candidate for immunotherapeutic approaches as new adjuvants to conventional treatments in pancreatic cancer.


Gastroenterology | 2013

Vaccination With ENO1 DNA Prolongs Survival of Genetically Engineered Mice With Pancreatic Cancer

Paola Cappello; Simona Rolla; Roberto Chiarle; Moitza Principe; Federica Cavallo; Giovanni Perconti; Salvatore Feo; Mirella Giovarelli; Francesco Novelli

BACKGROUND & AIMS Pancreatic ductal adenocarcinoma (PDA) is an aggressive tumor, and patients typically present with late-stage disease; rates of 5-year survival after pancreaticoduodenectomy are low. Antibodies against α-enolase (ENO1), a glycolytic enzyme, are detected in more than 60% of patients with PDA, and ENO1-specific T cells inhibit the growth of human pancreatic xenograft tumors in mice. We investigated whether an ENO1 DNA vaccine elicits antitumor immune responses and prolongs survival of mice that spontaneously develop autochthonous, lethal pancreatic carcinomas. METHODS We injected and electroporated a plasmid encoding ENO1 (or a control plasmid) into Kras(G12D)/Cre (KC) mice and Kras(G12D)/Trp53(R172H)/Cre (KPC) mice at 4 weeks of age (when pancreatic intraepithelial lesions are histologically evident). Antitumor humoral and cellular responses were analyzed by histology, immunohistochemistry, enzyme-linked immunosorbent assays, flow cytometry, and enzyme-linked immunosorbent spot and cytotoxicity assays. Survival was analyzed by Kaplan-Meier analysis. RESULTS The ENO1 vaccine induced antibody and a cellular response and increased survival times by a median of 138 days in KC mice and 42 days in KPC mice compared with mice given the control vector. On histologic analysis, the vaccine appeared to slow tumor progression. The vaccinated mice had increased serum levels of anti-ENO1 immunoglobulin G, which bound the surface of carcinoma cells and induced complement-dependent cytotoxicity. ENO1 vaccination reduced numbers of myeloid-derived suppressor cells and T-regulatory cells and increased T-helper 1 and 17 responses. CONCLUSIONS In a genetic model of pancreatic carcinoma, vaccination with ENO1 DNA elicits humoral and cellular immune responses against tumors, delays tumor progression, and significantly extends survival. This vaccination strategy might be developed as a neoadjuvant therapy for patients with PDA.


PLOS ONE | 2010

Myc Promoter-Binding Protein-1 (MBP-1) Is a Novel Potential Prognostic Marker in Invasive Ductal Breast Carcinoma

Mariavera Lo Presti; Arianna Ferro; Flavia Contino; Claudia Mazzarella; Silvia Sbacchi; Elena Roz; Carmelo Lupo; Giovanni Perconti; Agata Giallongo; Paola Migliorini; Antonio Marrazzo; Salvatore Feo

Background Alpha-enolase is a glycolytic enzyme that catalyses the formation of phosphoenolpyruvate in the cell cytoplasm. α-Enolase and the predominantly nuclear Myc promoter-binding protein-1 (MBP-1) originate from a single gene through the alternative use of translational starting sites. MBP-1 binds to the P2 c-myc promoter and competes with TATA-box binding protein (TBP) to suppress gene transcription. Although several studies have shown an antiproliferative effect of MBP-1 overexpression on several human cancer cells, to date detailed observations of α-enolase and MBP-1 relative expression in primary tumors versus normal tissues and their correlation with clinicopathological features have not been undertaken. Methodology and Findings We analyzed α-enolase and MBP-1 expression in normal breast epithelium and primary invasive ductal breast carcinoma (IDC) from 177 patients by Western blot and immunohistochemical analyses, using highly specific anti-α-enolase monoclonal antibodies. A significant increase in the expression of cytoplasmic α-enolase was observed in 98% of the tumors analysed, compared to normal tissues. Nuclear MBP-1 was found in almost all the normal tissues while its expression was retained in only 35% of the tumors. Statistically significant associations were observed among the nuclear expression of MBP-1 and ErbB2 status, Ki-67 expression, node status and tumor grade. Furthermore MBP-1 expression was associated with good survival of patients with IDC. Conclusions MBP-1 functions in repressing c-myc gene expression and the results presented indicate that the loss of nuclear MBP-1 expression in a large number of IDC may be a critical step in the development and progression of breast cancer and a predictor of adverse outcome. Nuclear MBP-1 appears to be a novel and valuable histochemical marker with potential prognostic value in breast cancer.


BMC Cancer | 2013

Negative transcriptional control of ERBB2 gene by MBP-1 and HDAC1: diagnostic implications in breast cancer

Flavia Contino; Claudia Mazzarella; Arianna Ferro; Mariavera Lo Presti; Elena Roz; Carmelo Lupo; Giovanni Perconti; Agata Giallongo; Salvatore Feo

BackgroundThe human ERBB2 gene is frequently amplified in breast tumors, and its high expression is associated with poor prognosis. We previously reported a significant inverse correlation between Myc promoter-binding protein-1 (MBP-1) and ERBB2 expression in primary breast invasive ductal carcinoma (IDC). MBP-1 is a transcriptional repressor of the c-MYC gene that acts by binding to the P2 promoter; only one other direct target of MBP-1, the COX2 gene, has been identified so far.MethodsTo gain new insights into the functional relationship linking MBP-1 and ERBB2 in breast cancer, we have investigated the effects of MBP-1 expression on endogenous ERBB2 transcript and protein levels, as well as on transcription promoter activity, by transient-transfection of SKBr3 cells. Reporter gene and chromatin immunoprecipitation assays were used to dissect the ERBB2 promoter and identify functional MBP-1 target sequences. We also investigated the relative expression of MBP-1 and HDAC1 in IDC and normal breast tissues by immunoblot analysis and immunohistochemistry.ResultsTransfection experiments and chromatin immunoprecipitation assays in SKBr3 cells indicated that MBP-1 negatively regulates the ERBB2 gene by binding to a genomic region between nucleotide −514 and −262 of the proximal promoter; consistent with this, a concomitant recruitment of HDAC1 and loss of acetylated histone H4 was observed. In addition, we found high expression of MBP-1 and HDAC1 in normal tissues and a statistically significant inverse correlation with ErbB2 expression in the paired tumor samples.ConclusionsAltogether, our in vitro and in vivo data indicate that the ERBB2 gene is a novel MBP-1 target, and immunohistochemistry analysis of primary tumors suggests that the concomitant high expression of MBP-1 and HDAC1 may be considered a diagnostic marker of cancer progression for breast IDC.


Scientific Reports | 2017

Pro-invasive stimuli and the interacting protein Hsp70 favour the route of alpha-enolase to the cell surface

Giovanni Perconti; Cristina Maranto; Daniele P. Romancino; Patrizia Rubino; Salvatore Feo; Antonella Bongiovanni; Agata Giallongo

Cell surface expression of alpha-enolase, a glycolytic enzyme displaying moonlighting activities, has been shown to contribute to the motility and invasiveness of cancer cells through the protein non-enzymatic function of binding plasminogen and enhancing plasmin formation. Although a few recent records indicate the involvement of protein partners in the localization of alpha-enolase to the plasma membrane, the cellular mechanisms underlying surface exposure remain largely elusive. Searching for novel interactors and signalling pathways, we used low-metastatic breast cancer cells, a doxorubicin-resistant counterpart and a non-tumourigenic mammary epithelial cell line. Here, we demonstrate by a combination of experimental approaches that epidermal growth factor (EGF) exposure, like lipopolysaccharide (LPS) exposure, promotes the surface expression of alpha-enolase. We also establish Heat shock protein 70 (Hsp70), a multifunctional chaperone distributed in intracellular, plasma membrane and extracellular compartments, as a novel alpha-enolase interactor and demonstrate a functional involvement of Hsp70 in the surface localization of alpha-enolase. Our results contribute to shedding light on the control of surface expression of alpha-enolase in non-tumourigenic and cancer cells and suggest novel targets to counteract the metastatic potential of tumours.


FEBS Letters | 2015

Cellular stress induces cap‐independent alpha‐enolase/MBP‐1 translation

Cristina Maranto; Giovanni Perconti; Flavia Contino; Patrizia Rubino; Salvatore Feo; Agata Giallongo

Myc promoter‐binding protein‐1 (MBP‐1) is a shorter protein variant of the glycolytic enzyme alpha‐enolase. Although several lines of evidence indicate that MBP‐1 acts as a tumor suppressor, the cellular mechanisms and signaling pathways underlying MBP‐1 expression still remain largely elusive. To dissect these pathways, we used the SkBr3 breast cancer cell line and non‐tumorigenic HEK293T cells ectopically overexpressing alpha‐enolase/MBP‐1. Here, we demonstrate that induced cell stresses promote MBP‐1 expression through the AKT/PERK/eIF2α signaling axis. Our results contribute to shedding light on the molecular mechanisms underlying MBP‐1 expression in non‐tumorigenic and cancer cells.


Biochimica et Biophysica Acta | 2007

The kelch protein NS1-BP interacts with alpha-enolase/MBP-1 and is involved in c-Myc gene transcriptional control

Giovanni Perconti; Arianna Ferro; Felice Amato; Patrizia Rubino; Davide Randazzo; Thorsten Wolff; Salvatore Feo; Agata Giallongo


Anticancer Research | 2015

Gene Expression Profiling of MCF10A Breast Epithelial Cells Exposed to IOERT

Luigi Minafra; Valentina Bravatà; Giorgio Ivan Russo; Giusi Irma Forte; Francesco Paolo Cammarata; Marilena Ripamonti; Giuliana Candiano; Melchiorre Cervello; Agata Giallongo; Giovanni Perconti; Cristina Messa; Maria Carla Gilardi


PeerJ | 2017

Detecting significant features in modeling microRNA-target interactions

Claudia Coronnello; Giovanni Perconti; Patrizia Rubino; Flavia Contino; Serena Bivona; Salvatore Feo; Agata Giallongo

Collaboration


Dive into the Giovanni Perconti's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Agata Giallongo

National Research Council

View shared research outputs
Top Co-Authors

Avatar

Patrizia Rubino

National Research Council

View shared research outputs
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