Network


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

Hotspot


Dive into the research topics where Antonia Bellizzi is active.

Publication


Featured researches published by Antonia Bellizzi.


The FASEB Journal | 2000

Na+/H+ exchanger-dependent intracellular alkalinization is an early event in malignant transformation and plays an essential role in the development of subsequent transformation-associated phenotypes

Stephan J. Reshkin; Antonia Bellizzi; Sandra Caldeira; Ilaria Malanchi; Manuela Poignee; Marianna Alunni-Fabbroni; Valeria Casavola; Massimo Tommasino

In this study we investigate the mechanism of intracellular pH change and its role in malignant transformation using the E7 oncogene of human papillomavirus type 16 (HPV16). Infecting NIH3T3 cells with recombinant retroviruses expressing the HPV16 E7 or a transformation deficient mutant we show that alkalinization is transformation specific. In NIH3T3 cells in which transformation can be turned on and followed by induction of the HPV16 E7 oncogene expression, we demonstrate that cytoplasmic alkalinization is an early event and was driven by stimulation of Na+/H+ exchanger activity via an increase in the affinity of the intracellular NHE‐1 proton regulatory site. Annulment of the E7‐induced cytoplasmic alkalinization by specific inhibition of the NHE‐1, acidification of culture medium, or clamping the pHi to nontransformed levels prevented the development of later transformed phenotypes such as increased growth rate, serum‐independent growth, anchorage‐independent growth, and glycolytic metabolism. These findings were verified in human keratinocytes (HPKIA), the natural host of HPV. Results from both NIH3T3 and HPKIA cells show that alkalinization acts on pathways that are independent of the E2F‐mediated transcriptional activation of cell cycle regulator genes. Moreover, we show that the transformationdependent increase in proliferation is independent of the concomitant stimulation of glycolysis. Finally, treatment of nude mice with the specific inhibitor of NHE‐1, DMA, delayed the development of HPV16‐keratinocyte tumors. Our data confirm that activation of the NHE‐1 and resulting cellular alkalinization is a key mechanism in oncogenic transformation and is necessary for the development and maintenance of the transformed phenotype.—Reshkin, S. J., Bellizzi, A., Caldeira, S., Albarani, V., Malanchi, I., Poignee, M., Alunni‐Fabbroni, M., Casavola, V., Tommasino, M. Na+/H+ exchanger‐dependent intracellular alkalinization is an early event in malignant transformation and plays an essential role in the development of subsequent transformation‐associated phenotypes.


International Journal of Cancer | 2007

Cytoskeleton and paclitaxel sensitivity in breast cancer : the role of β-tubulins

Stefania Tommasi; Anita Mangia; Rosanna Lacalamita; Antonia Bellizzi; Vita Fedele; Annalisa Chiriatti; Christopher Thomssen; Nancy Kendzierski; A. Latorre; Vito Lorusso; Francesco Schittulli; Francesco Zito; Maria Kavallaris; Angelo Paradiso

The antineoplastic effect of paclitaxel is mainly related to its ability to bind the β subunit of tubulin, thus preventing tubulin chain depolarization and inducing apoptosis. The relevance of the Class I β‐tubulin characteristics have also been confirmed in the clinical setting where mutations of paclitaxel‐binding site of β‐tubulin Class I have been related to paclitaxel resistance in non small cell lung and ovarian cancers. In the present study, we verified the hypothesis of a relationship between molecular alterations of β‐tubulin Class I and paclitaxel sensitivity in a panel of breast cell lines with different drug IC50. The Class I β‐tubulin gene cDNA has been sequenced detecting heterozygous missense mutations (exon 1 and 4) only in MCF‐7 and SK‐BR‐3 lines. Furthermore, the expression (at both mRNA and protein level) of the different isotypes have been analyzed demonstrating an association between low cell sensitivity to paclitaxel and Class III β‐tubulin expression increasing. Antisense oligonucleotide (ODN) experiments confirmed that the inhibition of Class III β‐tubulin could at least partially increase paclitaxel‐chemosensitivity. The hypothesis of a relationship between β‐tubulin tumor expression and paclitaxel clinical response has been finally verified in a series of 92 advanced breast cancer patients treated with a first line paclitaxel‐based chemotherapy. Thirty‐five percent (95% CI: 45–31) of patients with high Class III β‐tubulin expression showed a disease progression vs. only 7% of patients with low expression (35% vs. 7%, p < 0.002). Our study suggests that Class III β‐tubulin tumor expression could be considered a predictive biomarker of paclitaxel‐clinical resistance for breast cancer patients.


Breast Cancer Research | 2004

The Na + -H + exchanger-1 induces cytoskeletal changes involving reciprocal RhoA and Rac1 signaling, resulting in motility and invasion in MDA-MB-435 cells

Angelo Paradiso; Rosa Angela Cardone; Antonia Bellizzi; A. Bagorda; Lorenzo Guerra; Massimo Tommasino; Valeria Casavola; Stephan J. Reshkin

IntroductionAn increasing body of evidence shows that the tumour microenvironment is essential in driving neoplastic progression. The low serum component of this microenvironment stimulates motility/invasion in human breast cancer cells via activation of the Na+–H+ exchanger (NHE) isoform 1, but the signal transduction systems that underlie this process are still poorly understood. We undertook the present study to elucidate the role and pattern of regulation by the Rho GTPases of this serum deprivation-dependent activation of both NHE1 and subsequent invasive characteristics, such as pseudopodia and invadiopodia protrusion, directed cell motility and penetration of normal tissues.MethodsThe present study was performed in a well characterized human mammary epithelial cell line representing late stage metastatic progression, MDA-MB-435. The activity of RhoA and Rac1 was modified using their dominant negative and constitutively active mutants and the activity of NHE1, cell motility/invasion, F-actin content and cell shape were measured.ResultsWe show for the first time that serum deprivation induces NHE1-dependent morphological and cytoskeletal changes in metastatic cells via a reciprocal interaction of RhoA and Rac1, resulting in increased chemotaxis and invasion. Deprivation changed cell shape by reducing the amount of F-actin and inducing the formation of leading edge pseudopodia. Serum deprivation inhibited RhoA activity and stimulated Rac1 activity. Rac1 and RhoA were antagonistic regulators of both basal and stimulated tumour cell NHE1 activity. The regulation of NHE1 activity by RhoA and Rac1 in both conditions was mediated by an alteration in intracellular proton affinity of the exchanger. Interestingly, the role of each of these G-proteins was reversed during serum deprivation; basal NHE1 activity was regulated positively by RhoA and negatively by Rac1, whereas RhoA negatively and Rac1 positively directed the stimulation of NHE1 during serum deprivation. Importantly, the same pattern of RhoA and Rac1 regulation found for NHE1 activity was observed in both basal and serum deprivation dependent increases in motility, invasion and actin cytoskeletal organization.ConclusionOur findings suggest that the reported antagonistic roles of RhoA and Rac1 in cell motility/invasion and cytoskeletal organization may be due, in part, to their concerted action on NHE1 activity as a convergence point.


Histopathology | 2009

Biological role of NHERF1 protein expression in breast cancer.

Anita Mangia; Annalisa Chiriatti; Antonia Bellizzi; Andrea Malfettone; Baldassarre Stea; Francesso Alfredo Zito; Stephen Joel Reshkin; Giovanni Simone; Angelo Paradiso

Aims:  To determine the role of Na+/H+ exchanger regulatory factor (NHERF1) in breast cancerogenesis and progression.


Journal of Cellular Physiology | 2013

Co-expression of CD133+/CD44+ in human colon cancer and liver metastasis

Antonia Bellizzi; Sinto Sebastian; Pasquale Ceglia; Matteo Centonze; Rosa Divella; Elvira Foglia Manzillo; Amalia Azzariti; Nicola Silvestris; Severino Montemurro; Cosimo Caliandro; Raffaele De Luca; Giuseppe Cicero; Sergio Rizzo; Antonio Russo; Michele Quaranta; Giovanni Simone; Angelo Paradiso

Although relatively good therapeutic results are achieved in non‐advanced cancer, the prognosis of the advanced colon cancer still remains poor, dependent on local or distant recurrence of the disease. One of the factors responsible for recurrence is supposed to be cancer stem cells (CSCs) or tumor‐initiating cells, which are a population of cancer cells with ability to perpetuate themselves through self‐renewal and to generate differentiated cells, thought to be responsible for tumor recurrence. This study globally approach the possible role of tissue‐derived stem cells in the initiation of colon cancer and its metastatic process in the liver. Fresh surgical specimens from colon cancer, non‐tumor tissue and liver metastasis were obtained directly from the operating room, examined, and immediately processed. CSCs were selected under serum‐free conditions and characterized by CD44 and CD133 expression levels. CD133+/CD44+ cell populations were then investigated in paraffin‐embedded tissues and circulating tumor cells isolated from peripheral blood of the same group of colon cancer patients. Our data demonstrate that metastatic properties of cell populations from blood and liver metastasis, differently from primitive tumors, seem to be strictly related to the phenotype CD133 positive and CD44 positive. J. Cell. Physiol. 228: 408–415, 2013.


Breast Care | 2010

NHERF1/EBP50 in Breast Cancer: Clinical Perspectives

Antonia Bellizzi; Andrea Malfettone; Rosa Angela Cardone; Anita Mangia

Na+/H+ exchanger regulatory factor 1 (NHERF1) is a postsynaptic density 95/disc-large/zona occludens (PDZ) domain-containing protein that recruits membrane receptors/transporters and cytoplasmic signaling proteins into functional complexes. NHERF1 expression has been demonstrated to be altered in breast cancer, but its role in mammary cancerogenesis and progression remains still undefined. In this paper, we review what is known on the pathological role and the potential clinical application of NHERF1 protein in breast cancer. Recent evidence shows that an increased cytoplasmic expression of NHERF1 suggests a key role of its localization/compartmentalization in defining cancerogenesis, progression, and invasion. NHERF1 overexpression is associated with increasing tumor cytohistological grade, aggressive clinical behavior, unfavorable prognosis, and increased tumor hypoxia. Moreover, NHERF1 co-localizes with the oncogenic receptor HER2/neu in HER2/neu-overexpressing carcinoma and in distant metastases. These data make NHERF1 also a potential candidate of clinical relevance for anti-HER2/neu therapy.


Neoplasia | 2015

A novel NHE1-centered signaling cassette drives epidermal growth factor receptor-dependent pancreatic tumor metastasis and is a target for combination therapy

Rosa Angela Cardone; Maria Raffaella Greco; Katrine Zeeberg; Angela Zaccagnino; Mara Saccomano; Antonia Bellizzi; Philipp Bruns; Marta Menga; Christian Pilarsky; Albrecht Schwab; Frauke Alves; Holger Kalthoff; Valeria Casavola; Stephan J. Reshkin

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers principally because of early invasion and metastasis. The epidermal growth factor receptor (EGFR) is essential for PDAC development even in the presence of Kras, but its inhibition with erlotinib gives only a modest clinical response, making the discovery of novel EGFR targets of critical interest. Here, we revealed by mining a human pancreatic gene expression database that the metastasis promoter Na+/H+ exchanger (NHE1) associates with the EGFR in PDAC. In human PDAC cell lines, we confirmed that NHE1 drives both basal and EGF-stimulated three-dimensional growth and early invasion via invadopodial extracellular matrix digestion. EGF promoted the complexing of EGFR with NHE1 via the scaffolding protein Na +/H + exchanger regulatory factor 1, engaging EGFR in a negative transregulatory loop that controls the extent and duration of EGFR oncogenic signaling and stimulates NHE1. The specificity of NHE1 for growth or invasion depends on the segregation of the transient EGFR/Na +/H + exchanger regulatory factor 1/NHE1 signaling complex into dimeric subcomplexes in different lipid raftlike membrane domains. This signaling complex was also found in tumors developed in orthotopic mice. Importantly, the specific NHE1 inhibitor cariporide reduced both three-dimensional growth and invasion independently of PDAC subtype and synergistically sensitized these behaviors to low doses of erlotinib.


Histopathology | 2011

Na+/H+ exchanger regulatory factor 1 expression levels in blood and tissue predict breast tumour clinical behaviour

Antonia Bellizzi; Anita Mangia; Andrea Malfettone; Rosa Angela Cardone; Giovanni Simone; Stephan J. Reshkin; Angelo Paradiso

Bellizzi A, Mangia A, Malfettone A, Cardone R A, Simone G, Reshkin S J & Paradiso A
(2011) Histopathology58, 1086–1095
Na+/H+exchanger regulatory factor 1 expression levels in blood and tissue predict breast tumour clinical behaviour


Archive | 2011

NHERF1 expression levels in blood and tissue predict breast tumor clinical behaviour

Antonia Bellizzi; Anita Mangia; Andrea Malfettone; Rosa Angela Cardone; Giovanni Simone; Stephan J. Reshkin; A. Paradiso

Bellizzi A, Mangia A, Malfettone A, Cardone R A, Simone G, Reshkin S J & Paradiso A
(2011) Histopathology58, 1086–1095
Na+/H+exchanger regulatory factor 1 expression levels in blood and tissue predict breast tumour clinical behaviour


International Journal of Oncology | 2015

The scaffolding protein NHERF1 sensitizes EGFR-dependent tumor growth, motility and invadopodia function to gefitinib treatment in breast cancer cells

Antonia Bellizzi; Maria Raffaella Greco; Rosa Rubino; Angelo Paradiso; Stefania Forciniti; Katrine Zeeberg; Rosa Angela Cardone; Stephan J. Reshkin

Triple negative breast cancer (TNBC) patients cannot be treated with endocrine therapy or targeted therapies due to lack of related receptors. These patients overexpress the epidermal growth factor receptor (EGFR), but are resistant to tyrosine kinase inhibitors (TKIs) and anti-EGFR therapies. Mechanisms suggested for resistance to TKIs include EGFR independence, mutations and alterations in EGFR and in its downstream signalling pathways. Ligand-induced endocytosis and degradation of EGFR play important roles in the downregulation of the EGFR signal suggesting that its activity could be regulated by targeting its trafficking. Evidence in normal cells showing that the scaffolding protein Na+/H+ exchanger regulatory factor 1 (NHERF1) can associate with EGFR to regulate its trafficking, led us to hypothesize that NHERF1 expression levels could regulate EGFR trafficking and functional expression in TNBC cells and, in this way, modulate its role in progression and response to treatment. We investigated the subcellular localization of NHERF1 and its interaction with EGFR in a metastatic basal like TNBC cell model, MDA-MB‑231, and the role of forced NHERF1 overexpression and/or stimulation with EGF on the sensitivity to EGFR specific TKI treatment with gefitinib. Stimulation with EGF induces an interaction of NHERF1 with EGFR to regulate its localization, degradation and function. NHERF1 overexpression is sufficient to drive its interaction with EGFR in non-stimulated conditions, inhibits EGFR degradation and increases its retention time in the plasma membrane. Importantly, NHERF1 overexpression strongly sensitized the cell to the pharmacological inhibition by gefitinib of EGFR-driven growth, motility and invadopodia-dependent ECM proteolysis. The further determination of how the NHERF1‑EGFR interaction is regulated may improve our understanding of TNBC resistance to the action of existing anticancer drugs.

Collaboration


Dive into the Antonia Bellizzi'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

Giovanni Simone

National Cancer Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge