Network


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

Hotspot


Dive into the research topics where Veronica Finisguerra is active.

Publication


Featured researches published by Veronica Finisguerra.


Nature | 2011

Macrophage skewing by Phd2 haplodeficiency prevents ischaemia by inducing arteriogenesis

Yukiji Takeda; Sandra Costa; Estelle Delamarre; Carmen Roncal; Rodrigo Leite de Oliveira; Mario Leonardo Squadrito; Veronica Finisguerra; Sofie Deschoemaeker; Françoise Bruyère; Mathias Wenes; Alexander Hamm; Jens Serneels; Julie Magat; Tapan Bhattacharyya; Andrey Anisimov; Bénédicte F. Jordan; Kari Alitalo; Patrick H. Maxwell; Bernard Gallez; Zhen W. Zhuang; Yoshihiko Saito; Michael Simons; Michele De Palma; Massimiliano Mazzone

PHD2 serves as an oxygen sensor that rescues blood supply by regulating vessel formation and shape in case of oxygen shortage. However, it is unknown whether PHD2 can influence arteriogenesis. Here we studied the role of PHD2 in collateral artery growth by using hindlimb ischaemia as a model, a process that compensates for the lack of blood flow in case of major arterial occlusion. We show that Phd2 (also known as Egln1) haplodeficient (Phd2+/−) mice displayed preformed collateral arteries that preserved limb perfusion and prevented tissue necrosis in ischaemia. Improved arteriogenesis in Phd2+/− mice was due to an expansion of tissue-resident, M2-like macrophages and their increased release of arteriogenic factors, leading to enhanced smooth muscle cell (SMC) recruitment and growth. Both chronic and acute deletion of one Phd2 allele in macrophages was sufficient to skew their polarization towards a pro-arteriogenic phenotype. Mechanistically, collateral vessel preconditioning relied on the activation of canonical NF-κB pathway in Phd2+/− macrophages. These results unravel how PHD2 regulates arteriogenesis and artery homeostasis by controlling a specific differentiation state in macrophages and suggest new treatment options for ischaemic disorders.


Journal of Clinical Investigation | 2010

Sema3E–Plexin D1 signaling drives human cancer cell invasiveness and metastatic spreading in mice

Andrea Casazza; Veronica Finisguerra; Lorena Capparuccia; Andrea Camperi; Jakub M. Swiercz; Sabrina Rizzolio; Charlotte Rolny; Claus Christensen; Andrea Bertotti; Ivana Sarotto; Mauro Risio; Livio Trusolino; Jürgen Weitz; Martin Schneider; Massimiliano Mazzone; Paolo M. Comoglio; Luca Tamagnone

Semaphorin 3E (Sema3E) is a secreted molecule implicated in axonal path finding and inhibition of developmental and postischemic angiogenesis. Sema3E is also highly expressed in metastatic cancer cells, but its mechanistic role in tumor progression was not understood. Here we show that expression of Sema3E and its receptor Plexin D1 correlates with the metastatic progression of human tumors. Consistent with the clinical data, knocking down endogenous expression of either Sema3E or Plexin D1 in human metastatic carcinoma cells hampered their metastatic potential when injected into mice, while tumor growth was not markedly affected. Conversely, overexpression of exogenous Sema3E in cancer cells increased their invasiveness, transendothelial migration, and metastatic spreading, although it inhibited tumor vessel formation, resulting in reduced tumor growth in mice. The proinvasive and metastatic activity of Sema3E in tumor cells was dependent on transactivation of the Plexin D1-associated ErbB2/Neu oncogenic kinase. In sum, Sema3E-Plexin D1 signaling in cancer cells is crucially implicated in their metastatic behavior and may therefore be a promising target for strategies aimed at blocking tumor metastasis.


Nature | 2015

MET is required for the recruitment of anti-tumoural neutrophils

Veronica Finisguerra; Giusy Di Conza; Mario Di Matteo; Jens Serneels; Sandra Costa; A. A. Roger Thompson; Els Wauters; Sarah R. Walmsley; Hans Prenen; Zvi Granot; Andrea Casazza; Massimiliano Mazzone

Mutations or amplification of the MET proto-oncogene are involved in the pathogenesis of several tumours, which rely on the constitutive engagement of this pathway for their growth and survival. However, MET is expressed not only by cancer cells but also by tumour-associated stromal cells, although its precise role in this compartment is not well characterized. Here we show that MET is required for neutrophil chemoattraction and cytotoxicity in response to its ligand hepatocyte growth factor (HGF). Met deletion in mouse neutrophils enhances tumour growth and metastasis. This phenotype correlates with reduced neutrophil infiltration to both the primary tumour and metastatic sites. Similarly, Met is necessary for neutrophil transudation during colitis, skin rash or peritonitis. Mechanistically, Met is induced by tumour-derived tumour necrosis factor (TNF)-α or other inflammatory stimuli in both mouse and human neutrophils. This induction is instrumental for neutrophil transmigration across an activated endothelium and for inducible nitric oxide synthase production upon HGF stimulation. Consequently, HGF/MET-dependent nitric oxide release by neutrophils promotes cancer cell killing, which abates tumour growth and metastasis. After systemic administration of a MET kinase inhibitor, we prove that the therapeutic benefit of MET targeting in cancer cells is partly countered by the pro-tumoural effect arising from MET blockade in neutrophils. Our work identifies an unprecedented role of MET in neutrophils, suggests a potential ‘Achilles’ heel’ of MET-targeted therapies in cancer, and supports the rationale for evaluating anti-MET drugs in certain inflammatory diseases.


Oncogene | 2014

Tumor stroma: a complexity dictated by the hypoxic tumor microenvironment

Andrea Casazza; G Di Conza; Mathias Wenes; Veronica Finisguerra; Sofie Deschoemaeker; Max Mazzone

A lot of effort has been done to study how cancer cells react to low-oxygen tension, a condition known as hypoxia. Indeed, abnormal and dysfunctional blood vessels in the tumor are incapable to restore oxygenation, therefore perpetuating hypoxia, which, in turn, will fuel tumor progression, metastasis and resistance to antitumor therapies. Nevertheless, how stromal components including blood and lymphatic endothelial cells, pericytes and fibroblasts, as well as hematopoietic cells, respond to low-oxygen tension in comparison with their normoxic counterparts has been a matter of investigation in the last few years only and, to date, this field of research remains poorly understood. In general, opposing phenotypes can arise from the same stromal component when embedded in different tumor microenvironments, and, vice versa, different stromal components can have opposite reaction to the same tumor microenvironment. In this article, we will discuss the emerging link between tumor stroma and hypoxia, and how this complexity is translated at the molecular level.


Cancer Cell | 2012

Gene-Targeting of Phd2 Improves Tumor Response to Chemotherapy and Prevents Side-Toxicity

Rodrigo Leite de Oliveira; Sofie Deschoemaeker; Anne-Theres Henze; Koen Debackere; Veronica Finisguerra; Yukiji Takeda; Carmen Roncal; Daniela Dettori; Evelyne Tack; Yannick Jönsson; Lorenzo Veschini; Annelies Peeters; Andrey Anisimov; Matthias Hofmann; Kari Alitalo; Myriam Baes; Jan D'hooge; Peter Carmeliet; Massimiliano Mazzone

The success of chemotherapy in cancer treatment is limited by scarce drug delivery to the tumor and severe side-toxicity. Prolyl hydroxylase domain protein 2 (PHD2) is an oxygen/redox-sensitive enzyme that induces cellular adaptations to stress conditions. Reduced activity of PHD2 in endothelial cells normalizes tumor vessels and enhances perfusion. Here, we show that tumor vessel normalization by genetic inactivation of Phd2 increases the delivery of chemotherapeutics to the tumor and, hence, their antitumor and antimetastatic effect, regardless of combined inhibition of Phd2 in cancer cells. In response to chemotherapy-induced oxidative stress, pharmacological inhibition or genetic inactivation of Phd2 enhances a hypoxia-inducible transcription factor (HIF)-mediated detoxification program in healthy organs, which prevents oxidative damage, organ failure, and tissue demise. Altogether, our study discloses alternative strategies for chemotherapy optimization.


Oncogene | 2016

Preclinical and clinical evaluation of MET functions in cancer cells and in the tumor stroma

Veronica Finisguerra; Hans Prenen; Massimiliano Mazzone

A lot of attention has been dedicated to investigate the role of the tyrosine kinase receptor MET in tumors. The acquired notion that cancer cells from different histological origin strictly rely on the engagement of this specific oncogene for their growth and survival has certainly justified the development and the use of MET-targeted therapies in the clinic. However, the function and involvement of this pathway in the stroma (that often constitutes >50% of the global cellularity of the tumor) may offer the opportunity to conceive new patient stratification criteria, rational drug design and guided trials of new combination treatments. In this review, we will summarize and discuss the role of MET in cancer cells but especially in different stromal compartments, in light of the results showed by past and recent preclinical and clinical trials with anti-MET drugs.


Blood Cells Molecules and Diseases | 2009

Regulation of transferrin receptor 2 in human cancer cell lines

Alessia Calzolari; Veronica Finisguerra; Isabella Oliviero; Silvia Deaglio; Gualtiero Mariani; Fabio Malavasi; Ugo Testa

In a recent study we have explored TfR2 expression in a panel of cancer cell lines and we observed that about 40% of these cell lines clearly express TfR2. Taking advantage of this observation and considering the frequent overexpression of c-Myc in cancer cells we have explored the existence of a possible relationship between c-Myc and TfR2 in these cell lines. Our results provided evidence that TfR2(+) cell lines express low c-Myc levels and low TfR1 levels, while TfR2(-) cell lines express high c-Myc and TfR1 levels. Using the erythroleukemic K562 TfR2(+) cells as a model, we observed that agents that enhance c-Myc expression, such as iron, determine a decrease of TfR2 expression, while molecules that induce a decreased c-Myc expression, such as the iron chelator desferoxamine or the kinase inhibitor ST 1571, induce an enhanced TfR2 expression. On the other hand, we have evaluated a possible effect of hypoxia and nitric oxide on TfR2 expression in erythroleukemia K526 and hepatoma HepG2 cells, providing evidence that: (i) agents inducing cellular hypoxia, such as CoCl(2), elicited a marked upmodulation of TfR1, but a downmodulation of TfR2 expression; (ii) NO(+) donors, such as sodium nitroprusside (SNP), induced a moderate decrease of TfR1, associated with a marked decline of TfR2 expression; (iii) NO donors, such as S-Nitroso-N-Acetylpenicillamine (SNAP), induced a clear increase of TfR1, associated with a moderate upmodulation of TfR2 expression. The ensemble of these observations suggests that in cancer cell lines TfR2 expression can be modulated through stimuli similar to those known to act on TfR1 and these findings may have important implications for our understanding of the role of TfR2 in the regulation of iron homeostasis.


Cancer Research | 2010

Abstract 3398: Double-faced Sema3E-PlexinD1 signaling inhibits tumor growth but promotes metastatic spreading

Andrea Casazza; Lorena Capparuccia; Veronica Finisguerra; Massimiliano Mazzone; Paolo M. Comoglio; Luca Tamagnone

Semaphorins comprise a wide family of secreted and membrane-bound signals, originally found in neural development, and then implicated in a range of functions: from angiogenesis, to the immune response, to cancer progression. We have previously shown that high-affinity semaphorin receptors are found in the families of the plexins and of the neuropilins. Moreover, additional molecules interact with semaphorin receptors in cell-context dependent manner, featuring a complex scenario of multiple potential signaling pathways. The role of semaphorin signaling in tumor progression is currently under close scrutiny; it is known that cancer cells release semaphorins regulating their own behavior as well as that of cells in the tumor microenvironment, such as endothelial cells and recruited leukocytes. In this work, we focused on the function of Semaphorin 3E (Sema3E) and its high affinity receptor PlexinD1, and on their potential role to regulate cancer progression. Here we found that Sema3E and PlexinD1 are highly expressed in human metastatic tumors. Moreover, we provided experimental evidences that Sema3E released by tumor cells is a double-faced signal, acting via two distinctive signaling pathways. In paracrine manner, Sema3E-PlexinD1 signaling can elicit endothelial cell repulsion, thereby reducing vessel density and tumor growth. Conversely, Sema3E autocrine signaling in tumor cells thrusts their invasiveness, trans-endothelial migration, and metastatic spreading, through the trans-activation of PlexinD1-associated oncogenic kinases. Notably, by silencing the endogenous expression of either Sema3E or PlexinD1 the metastatic potential of cancer cells is significantly reduced, identifying this pathway as a major driver of the metastatic spreading. In sum, Sema3E autocrine signaling in cancer cells is crucially implicated to induce a metastatic behavior, and it appears as a promising target for strategies aimed at blocking tumor metastasis. Note: This abstract was not presented at the AACR 101st Annual Meeting 2010 because the presenter was unable to attend. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3398.


Translational Oncology | 2010

Transferrin receptor 2 is frequently and highly expressed in glioblastomas

Alessia Calzolari; Luigi Maria Larocca; Silvia Deaglio; Veronica Finisguerra; Alessandra Boe; Carla Raggi; Lucia Ricci-Vitani; Francesco Pierconti; Fabio Malavasi; Ruggero De Maria; Ugo Testa; Roberto Pallini


AACR Conference Cellular heterogeneity in the tumour microenvironment | 2014

Met expression in neutrophils is required to enhance their recruitment to the tumor and cytotoxic activity

Veronica Finisguerra; Andrea Casazza; Giusy Di Conza; Rocco Stirparo; Sandra Costa; A. A. Roger Thompson; Sarah R. Walmsley; Massimiliano Mazzone

Collaboration


Dive into the Veronica Finisguerra's collaboration.

Top Co-Authors

Avatar

Massimiliano Mazzone

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sofie Deschoemaeker

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Estelle Delamarre

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Jens Serneels

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Mathias Wenes

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge