Network


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

Hotspot


Dive into the research topics where Daniela Belloni is active.

Publication


Featured researches published by Daniela Belloni.


The FASEB Journal | 2004

Chromogranin A protects vessels against tumor necrosis factor alpha-induced vascular leakage

Elisabetta Ferrero; Silvia Scabini; Elena Magni; Chiara Foglieni; Daniela Belloni; Barbara Colombo; Flavio Curnis; Antonello Villa; Maria Elena Ferrero; Angelo Corti

Elevated levels of circulating chromogranin A (CgA), a protein stored in the secretory granules of many neuroendocrine cells and neurons, have been detected in the blood of patients with neuroendocrine tumors or heart failure. The pathophysiological role of increased secretion of CgA is unknown. Using mice bearing subcutaneous tumors genetically engineered to secrete CgA in circulation, we have found that increased blood levels of this protein prevent vascular leakage induced by tumor necrosis factor‐α (TNF) in the liver venous system. Structure–activity studies, carried out with CgA fragments administered to normal mice, showed that an active site is located within residues 7–57 of CgA. Accordingly, an anti‐CgA antibody directed to residues 53–57 inhibited the effect of circulating CgA, either endogenously produced or exogenously administered, on liver vessels. Studies of the mechanism of action showed that CgA inhibits TNF‐induced VE‐cadherin down‐regulation and barrier alteration of cultured endothelial cells, in an indirect manner. Other effectors, such as thrombin and vascular endothelial growth factor were partially inhibited by CgA N‐terminal fragments in in vitro permeability assays. These findings suggest that circulating CgA could help regulate the endothelial barrier function and to protect vessels against TNF‐induced plasma leakage in pathological conditions characterized by increased production of TNF and CgA, such as cancer or heart failure.


The FASEB Journal | 2007

The vasostatin-I fragment of chromogranin A inhibits VEGF-induced endothelial cell proliferation and migration

Daniela Belloni; Silvia Scabini; Chiara Foglieni; Lorenzo Veschini; Alessio Giazzon; Barbara Colombo; Alessandro Fulgenzi; Karen B. Helle; Maria Elena Ferrero; Angelo Corti; Elisabetta Ferrero

A growing body of evidence suggests that chromogranin A (CgA), a secretory protein released by many neuroendocrine cells and frequently used as a diagnostic and prognostic serum marker for a range of neuroendocrine tumors, is a precursor of several bioactive fragments. This work was undertaken to assess whether the N‐terminal fragment CgA1–76 (called vasostatin I) can inhibit the proangiogenic activity of vascular endothelial growth factor (VEGF), a factor involved in tumor growth. The effect of recom‐binant human vasostatin I (VS‐1) on VEGF‐induced human umbilical endothelial cells (HUVEC) signaling, proliferation, migration, and organization has been investigated. We have found that VS‐1 (3 μg/ml;330 nM) can inhibit VEGF‐induced ERK phosphorylation, as well as cell migration, proliferation, morphogenesis, and invasion of collagen gels in various in vitro assays. In addition, VS‐1 could inhibit the formation of capillary‐like structures in Matrigel plugs in a rat model. VS‐1 could also inhibit basal ERK phosphorylation and motility of HUVEC, leading to a more quiescent state in the absence of VEGF, without inducing apoptotic or necrotic effects. Conclusion: These findings suggest that vasostatin I may play a novel role as a regulator of endothelial cell function and homeostasis.—Belloni, D., Scabini, S., Foglieni, C., Veschini, L., Giazzon, A., Colombo, B., Fulgenzi, A., Helle, K. B., Ferrero, M. E., Corti, A., Ferrero, E. The vasostatin‐I fragment of chromogranin A inhibits VEGF‐induced endothelial cell proliferation and migration. FASEB J. 21, 3052–3062 (2007)


Cancer Research | 2005

MICA Expressed by Multiple Myeloma and Monoclonal Gammopathy of Undetermined Significance Plasma Cells Costimulates Pamidronate-Activated γδ Lymphocytes

Stefania Girlanda; Claudio Fortis; Daniela Belloni; Elisabetta Ferrero; Paolo Ticozzi; Clara Sciorati; Moreno Tresoldi; Aurelio Vicari; Thomas Spies; Veronika Groh; Federico Caligaris Cappio; Marina Ferrarini

Amino-biphosphonates (like pamidronate) activate human Vgamma9/Vdelta2 T lymphocytes and promote their cytotoxicity against multiple myeloma cells. T-cell receptor (TCR)-mediated effector functions of gammadelta cells are enhanced upon triggering of the activating receptor NKG2D by MICA, a stress-inducible antigen expressed by epithelial and some hematopoietic tumors, including multiple myeloma. Here we show that MICA was expressed not only by myeloma cell lines and by 6 of 10 primary multiple myeloma cells from patients but also by bone marrow plasma cells from all (six of six) patients with preneoplastic gammopathy (monoclonal gammopathy of undetermined significance, MGUS), a direct precursor of multiple myeloma. Moreover, compared with multiple myeloma plasma cells, MICA was expressed by MGUS plasma cells at significantly (P < 0.05) higher levels. MICA expressed by myeloma cell lines contributed to killing and IFN-gamma production by Vgamma9/Vdelta2 cells only upon pamidronate treatment, suggesting a dual interaction between Vgamma9/Vdelta2 lymphocytes and multiple myeloma plasma cells involving both TCR triggering and NKG2D-mediated signals. Finally, MICA enhanced killing of freshly derived, pamidronate-treated multiple myeloma cells from patients by gammadelta cells, as indicated by the significantly (P < 0.05) higher gammadelta cytotoxicity against MICA-positive rather than MICA-negative multiple myeloma cells. Our results indicate that MICA expressed by monoclonal plasma cells is functional and correlates with disease stages, suggesting a role for the molecule in the immune surveillance against multiple myeloma. Moreover, pamidronate-activated Vgamma9/Vdelta2 lymphocytes can be exploited in the immune therapy of early stages multiple myeloma and possibly of premalignant disease.


Experimental Cell Research | 2010

Bortezomib induces autophagic death in proliferating human endothelial cells.

Daniela Belloni; Lorenzo Veschini; Chiara Foglieni; Giacomo Dell'Antonio; Federico Caligaris-Cappio; Marina Ferrarini; Elisabetta Ferrero

The proteasome inhibitor Bortezomib has been approved for the treatment of relapsed/refractory multiple myeloma (MM), thanks to its ability to induce MM cell apoptosis. Moreover, Bortezomib has antiangiogenic properties. We report that endothelial cells (EC) exposed to Bortezomib undergo death to an extent that depends strictly on their activation state. Indeed, while quiescent EC are resistant to Bortezomib, the drug results maximally toxic in EC switched toward angiogenesis with FGF, and exerts a moderate effect on subconfluent HUVEC. Moreover, EC activation state deeply influences the death pathway elicited by Bortezomib: after treatment, angiogenesis-triggered EC display typical features of apoptosis. Conversely, death of subconfluent EC is preceded by ROS generation and signs typical of autophagy, including intense cytoplasmic vacuolization with evidence of autophagosomes at electron microscopy, and conversion of the cytosolic MAP LC3 I form toward the autophagosome-associated LC3 II form. Treatment with the specific autophagy inhibitor 3-MA prevents both LC3 I/LC3 II conversion and HUVEC cell death. Finally, early removal of Bortezomib is accompanied by the recovery of cell shape and viability. These findings strongly suggest that Bortezomib induces either apoptosis or autophagy in EC; interfering with the autophagic response may potentiate the antiangiogenic effect of the drug.


PLOS ONE | 2013

Ex-Vivo Dynamic 3-D Culture of Human Tissues in the RCCS™ Bioreactor Allows the Study of Multiple Myeloma Biology and Response to Therapy

Marina Ferrarini; Nathalie Steimberg; Maurilio Ponzoni; Daniela Belloni; Angiola Berenzi; Stefania Girlanda; Federico Caligaris-Cappio; Giovanna Mazzoleni; Elisabetta Ferrero

Three-dimensional (3-D) culture models are emerging as invaluable tools in tumor biology, since they reproduce tissue-specific structural features and cell-cell interactions more accurately than conventional 2-D cultures. Multiple Myeloma, which depends on myeloma cell-Bone Marrow microenvironment interactions for development and response to drugs, may particularly benefit from such an approach. An innovative 3-D dynamic culture model based on the use of the RCCS™ Bioreactor was developed to allow long-term culture of myeloma tissue explants. This model was first validated with normal and pathological explants, then applied to tissues from myeloma patients. In all cases, histological examination demonstrated maintenance of viable myeloma cells inside their native microenvironment, with an overall well preserved histo-architecture including bone lamellae and vessels. This system was then successfully applied to evaluate the cytotoxic effects exerted by the proteasome inhibitor Bortezomib not only on myeloma cells but also on angiogenic vessels. Moreover, as surrogate markers of specialized functions expressed by myeloma cells and microenvironment, β2 microglobulin, VEGF and Angiopoietin-2 levels, as well as Matrix Metalloproteases activity, were evaluated in supernatants from 3D cultures and their levels reflected the effects of Bortezomib treatment. Notably, determination of β2 microglobulin levels in supernatants from Bortezomib-treated samples and in patientssera following Bortezomib-based therapies disclosed an overall concordance in the response to the drug ex vivo and in vivo. Our findings indicate, as a proof of principle, that 3-D, RCCS™ bioreactor-based culture of tissue explants can be exploited for studying myeloma biology and for a pre-clinical approach to patient-targeted therapy.


BMC Nephrology | 2006

Protective effect of EDTA preadministration on renal ischemia

Chiara Foglieni; Alessandro Fulgenzi; Paolo Ticozzi; Fabio Pellegatta; Clara Sciorati; Daniela Belloni; Elisabetta Ferrero; Maria Elena Ferrero

BackgroundChelation therapy with sodium edetate (EDTA) improved renal function and slowed the progression of renal insufficiency in patients subjected to lead intoxication. This study was performed to identify the underlying mechanism of the ability of EDTA treatment to protect kidneys from damage.MethodsThe effects of EDTA administration were studied in a rat model of acute renal failure induced by 60 minutes ischemia followed or not by 60 minutes reperfusion. Renal ischemic damage was evaluated by histological studies and by functional studies, namely serum creatinine and blood urea nitrogen levels. Treatment with EDTA was performed 30 minutes before the induction of ischemia. Polymorphonuclear cell (PMN) adhesion capability, plasmatic nitric oxide (NO) levels and endothelial NO synthase (eNOS) renal expression were studied as well as the EDTA protection from the TNFα-induced vascular leakage in the kidneys. Data was compared by two-way analysis of variance followed by a post hoc test.ResultsEDTA administration resulted in the preservation of both functional and histological parameters of rat kidneys. PMN obtained from peripheral blood of EDTA-treated ischemized rats, displayed a significant reduction in the expression of the adhesion molecule Mac-1 with respect to controls. NO was significantly increased by EDTA administration and eNOS expression was higher and more diffuse in kidneys of rats treated with EDTA than in the controls. Finally, EDTA administration was able to prevent in vivo the TNFα-induced vascular leakage in the kidneys.ConclusionThis data provides evidence that EDTA treatment is able to protect rat kidneys from ischemic damage possibly through the stimulation of NO production.


Blood | 2016

HIF-1α regulates the interaction of chronic lymphocytic leukemia cells with the tumor microenvironment

Roberta Valsecchi; Nadia Coltella; Daniela Belloni; Manfredi Ponente; Elisa ten Hacken; Cristina Scielzo; Lydia Scarfò; Maria Teresa Sabrina Bertilaccio; Paola Brambilla; Elisa Lenti; Filippo Martinelli Boneschi; Andrea Brendolan; Elisabetta Ferrero; Marina Ferrarini; Paolo Ghia; Giovanni Tonon; Maurilio Ponzoni; Federico Caligaris-Cappio; Rosa Bernardi

Hypoxia-inducible transcription factors (HIFs) regulate a wide array of adaptive responses to hypoxia and are often activated in solid tumors and hematologic malignancies due to intratumoral hypoxia and emerging new layers of regulation. We found that in chronic lymphocytic leukemia (CLL), HIF-1α is a novel regulator of the interaction of CLL cells with protective leukemia microenvironments and, in turn, is regulated by this interaction in a positive feedback loop that promotes leukemia survival and propagation. Through unbiased microarray analysis, we found that in CLL cells, HIF-1α regulates the expression of important chemokine receptors and cell adhesion molecules that control the interaction of leukemic cells with bone marrow and spleen microenvironments. Inactivation of HIF-1α impairs chemotaxis and cell adhesion to stroma, reduces bone marrow and spleen colonization in xenograft and allograft CLL mouse models, and prolongs survival in mice. Of interest, we found that in CLL cells, HIF-1α is transcriptionally regulated after coculture with stromal cells. Furthermore, HIF-1α messenger RNA levels vary significantly within CLL patients and correlate with the expression of HIF-1α target genes, including CXCR4, thus further emphasizing the relevance of HIF-1α expression to CLL pathogenesis.


American Journal of Transplantation | 2010

Improved function of circulating angiogenic cells is evident in type 1 diabetic islet-transplanted patients

Alessandra Petrelli; Anna Maestroni; Gian Paolo Fadini; Daniela Belloni; Massimo Venturini; M. Albiero; Sonja Kleffel; Bechara Mfarrej; A. Del Maschio; Paola Maffi; A. Avogaro; Elisabetta Ferrero; Gianpaolo Zerbini; A. Secchi; Paolo Fiorina

Circulating angiogenic cells (CACs) are vascular‐committed bone marrow‐derived cells that are dysfunctional in type 1 diabetes (T1D). Here we studied whether restoration of normoglycemia following islet transplantation is associated with better CAC function. We carried out a cross‐sectional study of 18 T1D patients, 14 insulin‐independent islet‐transplanted patients (ITA) and 14 healthy controls (C) evaluating in vivo and in vitro CACs viability and function. We found that the percentage of CACs in vivo did not differ among the three groups while the number of CAC colonies obtained from T1D, but not from ITA, was reduced compared to C (C = 7.3 ± 1.9, T1D = 0.9 ± 0.4 and ITA = 4.7 ± 1.9; p < 0.05 T1D vs. all). In vitro CAC migration/differentiation were similar, while in vivo an improved angiogenic ability of ITA compared to T1D was shown (capillary density: C = 93.5 ± 22.1, T1D = 19.2 ± 2.8 and ITA = 44.0 ± 10.5, p < 0.05 T1D vs. all). Increased apoptosis and lesser IL‐8 secretion were evident in CACs obtained from T1D compared to C and ITA. in vitro addition of anti‐hIL‐8 reduced the number of colonies obtained from C. Finally, T1D, but not ITA, had a lower endothelial‐dependent dilatation (EDD) compared with C. These data suggest that CAC function is altered in T1D and may be improved after islet transplantation.


European Journal of Histochemistry | 2009

Productive infection of HUVEC by HHV-8 is associated with changes compatible with angiogenic transformations.

Chiara Foglieni; Scabini S; Daniela Belloni; Francesco Broccolo; Lusso P; Mauro S. Malnati; Elisabetta Ferrero

Kaposis Sarcoma (KS) is an angioproliferative disease associated with human herpesvirus 8 (HHV-8) infection. We have characterized the morphologic and phenotypic modifications of HUVEC in a model of productive HHV-8 infection. HHV-8 replication was associated with ultra-structural changes, flattened soma and a loss of marginal folds and intercellular contacts, and morphologic features, spindle cell conversion and cordon-like structures formation. Phenotypic changes observed on cordon-like structures included partial loss and redistribution of CD31/PECAM-1 and VE-cadherin, uPAR up-regulation and de novo expression of CD13/APN. Such changes demonstrate the induction, in HUVEC, of an angiogenic profile. Most of these findings are directly linked to HHV-8-encoded proteins expression, suggesting that HHV-8 itself may participate to the initial steps of the angiogenic transformation in KS.


Experimental Cell Research | 2015

Angiopoietin-2 in Bone Marrow milieu promotes Multiple Myeloma-associated angiogenesis

Daniela Belloni; Magda Marcatti; Maurilio Ponzoni; Fabio Ciceri; Lorenzo Veschini; Angelo Corti; Federico Caligaris Cappio; Marina Ferrarini; Elisabetta Ferrero

Angiopoietin-2 (Ang-2) is involved in angiogenesis in both solid and hematological malignancies. In Multiple Myeloma (MM), serum Ang-2 correlates with disease progression and response to therapy. To address the patho-physiologic role of Ang-2 in MM associated angiogenesis, we used sera from patients with active MM, which contained significantly higher levels of the molecule, compared to those from patients with smoldering MM and Monoclonal Gammopathy of Undetermined Significance. MM Bone Marrow (BM) sera with high Ang-2 concentration specifically contributed to endothelial cell (EC) activation, while Ang-1 containing sera maintained EC stabilization. The functional dichotomy of Ang-1 and Ang-2 was confirmed by the triggering of distinctive signaling pathways down-stream the common Tie-2 receptor, i.e., the Akt or the ERK- phosphorylation pathway. Notably, Ang-2 but not VEGF serum levels correlated with BM micro-vessel density, further underscoring the key role of Ang-2 in angiogenesis. Western Blot, RT-PCR and immunocytochemistry identified MMEC as the major source of Ang-2, at variance with MM cells and CD14(+) BM monocytes. These data suggest that Ang-2 produced in the BM milieu may contribute to MM angiogenesis and suggest that the molecule can be further exploited both as angiogenesis biomarker and as a potential therapeutic target.

Collaboration


Dive into the Daniela Belloni's collaboration.

Top Co-Authors

Avatar

Elisabetta Ferrero

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Marina Ferrarini

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Chiara Foglieni

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Federico Caligaris-Cappio

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Angelo Corti

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Maurilio Ponzoni

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Lorenzo Veschini

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Antonio Secchi

Vita-Salute San Raffaele University

View shared research outputs
Researchain Logo
Decentralizing Knowledge