Network


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

Hotspot


Dive into the research topics where Dania Bozzato is active.

Publication


Featured researches published by Dania Bozzato.


Clinical Chemistry and Laboratory Medicine | 2009

Pancreatic cancer biomarkers discovery by surface-enhanced laser desorption and ionization time-of-flight mass spectrometry.

Filippo Navaglia; Paola Fogar; Daniela Basso; Eliana Greco; Andrea Padoan; Loris Tonidandel; Elisa Fadi; Carlo-Federico Zambon; Dania Bozzato; Stefania Moz; Roberta Seraglia; Sergio Pedrazzoli; Mario Plebani

Abstract Background: Surface-enhanced laser desorption and ionization time-of-flight mass spectrometry (SELDI-TOF/MS), a laboratory-friendly technique, is used to identify biomarkers for cancer. The aim of the present study was to explore the application of SELDI proteomic patterns in serum for distinguishing between cases of pancreatic cancer, chronic pancreatitis, type 2 diabetes mellitus and healthy controls. Methods: Sera from 12 healthy controls, 24 patients with type 2 diabetes mellitus, 126 with pancreatic cancer, including 84 with diabetes, and 61 with chronic pancreatitis, 32 of which were diabetics, were analyzed using SELDI-TOF/MS. Spectra (IMAC-30) were clustered and classified using Biomarker Wizard and Biomarker Pattern software. Results: Two decision tree classification algorithms, one with and one without CA 19-9, were constructed. In the absence of CA 19-9, the splitting protein peaks were: m/z 1526, 1211, and 3519; when CA 19-9 was used in the analysis, it replaced the m/z 3519 splitter. The two algorithms performed equally for classifying patients. A classification tree that considered diabetic patients only was constructed; the main splitters were: 1211, CA 19-9, 7903, 3359, 1802. With this algorithm, 100% of patients with type 2 diabetes mellitus, 97% with chronic pancreatitis and 77% of patients with pancreatic cancer were correctly classified. SELDI-TOF/MS features improved the diagnostic accuracy of CA 19-9 (AUC=0.883 for CA 19-9; AUC=0.935 for CA 19-9 and SELDI-TOF/MS features combined). Conclusions: SELDI-TOF/MS allows identification of new peptides which, in addition to CA 19-9, enable the correct classification of the vast majority of patients with pancreatic cancer, which can be distinguished from patients with chronic pancreatitis or type 2 diabetes mellitus. Clin Chem Lab Med 2009;47:713–23.


PLOS ONE | 2013

Pancreatic Tumors and Immature Immunosuppressive Myeloid Cells in Blood and Spleen: Role of Inhibitory Co-Stimulatory Molecules PDL1 and CTLA4. An In Vivo and In Vitro Study

Daniela Basso; Paola Fogar; Massimo Falconi; Elisa Fadi; Cosimo Sperti; Chiara Frasson; Eliana Greco; Domenico Tamburrino; Sara Teolato; Stefania Moz; Dania Bozzato; Michela Pelloso; Andrea Padoan; Giuseppe De Franchis; Elisa Gnatta; Monica Facco; Carlo Federico Zambon; Filippo Navaglia; Claudio Pasquali; Giuseppe Basso; Gianpietro Semenzato; Sergio Pedrazzoli; Paolo Pederzoli; Mario Plebani

Background Blood and spleen expansion of immature myeloid cells (IMCs) might compromise the immune response to cancer. We studied in vivo circulating and splenic T lymphocyte and IMC subsets in patients with benign and malignant pancreatic diseases. We ascertained in vitro whether pancreatic adenocarcinoma (PDAC)-associated IMC subsets are induced by tumor-derived soluble factors and whether they are immunosuppressive focusing on the inhibitory co-stimulatory molecules PDL1 and CTLA4. Methodology and Principal Findings 103 pancreatic and/or splenic surgical patients were enrolled including 52 PDAC, 10 borderline and 10 neuroendocrine tumors (NETs). Lymphocytes and IMCs were analysed by flow cytometry in blood, in spleen and in three PDAC cell conditioned (CM) or non conditioned PBMC. PDL1 and CTLA4 were studied in 30 splenic samples, in control and conditioned PBMC. IMCs were FACS sorted and co-coltured with allogenic T lymphocytes. In PDAC a reduction was found in circulating CD8+ lymphocytes (p = 0.004) and dendritic cells (p = 0.01), which were reduced in vitro by one PDAC CM (Capan1; p = 0.03). Blood myeloid derived suppressive cells (MDSCs) CD33+CD14−HLA-DR− were increased in PDAC (p = 0.022) and were induced in vitro by BxPC3 CM. Splenic dendritic cells had a higher PDL1 expression (p = 0.007), while CD33+CD14+HLA-DR− IMCs had a lower CTLA4 expression (p = 0.029) in PDAC patients. In vitro S100A8/A9 complex, one of the possible inflammatory mediators of immune suppression in PDAC, induced PDL1 (p = 0.018) and reduced CTLA4 expression (p = 0.028) among IMCs. IMCs not expressing CTLA4 were demonstrated to be immune suppressive. Conclusion In PDAC circulating dendritic and cytotoxic T cells are reduced, while MDSCs are increased and this might favour tumoral growth and progression. The reduced CTLA4 expression found among splenic IMCs of PDAC patients was demonstrated to characterize an immune suppressive phenotype and to be consequent to the direct exposure of myeloid cells to pancreatic cancer derived products, S100A8/A9 complex in particular.


Clinica Chimica Acta | 2011

New screening tests enrich anti-transglutaminase results and support a highly sensitive two-test based strategy for celiac disease diagnosis.

Daniela Basso; Graziella Guariso; Dania Bozzato; Elisa Rossi; Matilde Pescarin; Paola Fogar; Stefania Moz; Filippo Navaglia; Michela Pelloso; Marco Gasparetto; Carlo-Federico Zambon; Andrea Padoan; Eliana Greco; Massimo Rugge; Mario Plebani

BACKGROUND The identification of specific serological algorithms allowing the diagnosis of celiac disease (CD) is a new challenge for both the clinic and the laboratory. We compared the diagnostic accuracy of three new tests proposed for CD screening with that of the well established IgA tTG, and ascertained whether any combination of these tools might enhance accuracy in diagnosing CD. METHODS In sera from 329 CD and 374 control children, the following were assayed: IgA tTG; IgA/IgG, which identify tTG-gliadin complexes (Aeskulisa Celi Check and CeliCheck IgGA); IgA/IgG, which identify deamidated gliadin peptides and tTG (QUANTA Lite(TM) h-tTG/DGP Screen). RESULTS When specificity was set at 100%, the most sensitive index of CD was IgA tTG (75.7%, cut-off=100U), followed by QUANTA Lite(TM) h-tTG/DGP Screen (65.3%, cut-off 145U), Aeskulisa Celi Check (62.6%, cut-off 909U/mL) and CeliCheck IgGA (59.6%, cut-off 977U/mL). Three algorithms were obtained by combining IgA tTG with each of the new tests. The algorithm obtained by measuring IgA tTG and QUANTA Lite(TM) h-tTG/DGP Screen allowed the correct identification of CD in 78.7% of cases (negative predictive value=97.3%). CONCLUSIONS The two-test based strategy could be used for the cost effective diagnosis of CD.


Pancreas | 2011

Pancreatic cancer alters human CD4+ T lymphocyte function: a piece in the immune evasion puzzle.

Paola Fogar; Daniela Basso; Elisa Fadi; Eliana Greco; Giorgia Pantano; Andrea Padoan; Dania Bozzato; Monica Facco; Maria Colomba Sanzari; Sara Teolato; Carlo-Federico Zambon; Filippo Navaglia; Gianpietro Semenzato; Sergio Pedrazzoli; Mario Plebani

Objectives: To verify whether the dysregulation of CD4+ T cells concurs in worsening the outcome of pancreatic cancer, we compared the effects of pancreatic cancer and other gastrointestinal cancer cell-conditioned media on the (1) proliferation, migration, and differentiation of CD4+ T cells and (2) expansion of CD4+ memory (CD45RO), naive (CD45RA), activated (CD69), and regulatory (CD25) subsets. Methods: After culture of CD4+ T cells in control, pancreatic (BxPC3, Capan1, MiaPaCa2), or gastrointestinal cancer (AGS, HepG2, HT29) cell-conditioned media, we evaluated proliferation, migration, interferon &ggr; (IFN&ggr;) production, and CD45RA, CD45RO, CD69, and CD25 membrane expression in control and conditioned CD4+ T cells. Results: Only pancreatic cancer-conditioned media (1) inhibited CD4+ T-cell proliferation (P < 0.001) and migration under human stromal cell-derived factor-&agr; chemotaxis (P < 0.001) and (2) induced CD4+ T-cell IFN&ggr; production (P < 0.05) and the expansion of the CD69-positive subset (P < 0.001) with respect to the control, with no changes being found in the CD45RA, CD45RO, and CD25 subsets. Conclusions: The in vitro findings achieved in the present study demonstrate that pancreatic cancer cells inhibit CD4+ T-cell proliferation and migration, induce IFN&ggr; production, and favor a CD69+ subset expansion, suggesting that CD4+ T cells play an important role in pancreatic cancer immune evasion.


Journal of Cellular Physiology | 2011

Altered intracellular calcium fluxes in pancreatic cancer induced diabetes mellitus: Relevance of the S100A8 N‐terminal peptide (NT‐S100A8)

Daniela Basso; Eliana Greco; Andrea Padoan; Paola Fogar; Michele Scorzeto; Elisa Fadi; Dania Bozzato; Stefania Moz; Filippo Navaglia; Carlo-Federico Zambon; Roberta Seraglia; Eugenio De Carlo; Anna Valerio; Carlo Reggiani; Sergio Pedrazzoli; Mario Plebani

After isolating NT‐S100A8 from pancreatic cancer (PC) tissue of diabetic patients, we verified whether this peptide alters PC cell growth and invasion and/or insulin release and [Ca2+]i oscillations of insulin secreting cells and/or insulin signaling. BxPC3, Capan1, MiaPaCa2, Panc1 (PC cell lines) cell growth, and invasion were assessed in the absence or presence of 50, 200, and 500 nM NT‐S100A8. In NT‐S100A8 stimulated β‐TC6 (insulinoma cell line) culture medium, insulin and [Ca2+] were measured at 2, 3, 5, 10, 15, 30, and 60 min, and [Ca2+]i oscillations were monitored (epifluorescence) for 3 min. Five hundred nanomolars NT‐S100A8 stimulated BxPC3 cell growth only and dose dependently reduced MiaPaCa2 and Panc1 invasion. Five hundred nanomolars NT‐S100A8 induced a rapid insulin release and enhanced β‐TC6 [Ca2+]i oscillations after both one (F = 6.05, P < 0.01) and 2 min (F = 7.42, P < 0.01). In the presence of NT‐S100A8, [Ca2+] in β‐TC6 culture medium significantly decreased with respect to control cells (F = 6.3, P < 0.01). NT‐S100A8 did not counteract insulin induced phosphorylation of the insulin receptor, Akt and IκB‐α, but it independently activated Akt and NF‐κB signaling in PC cells. In conclusion, NT‐S100A8 exerts a mild effect on PC cell growth, while it reduces PC cell invasion, possibly by Akt and NF‐κB signaling, NT‐S100A8 enhances [Ca2+]i oscillations and insulin release, probably by inducing Ca2+ influx from the extracellular space, but it does not interfere with insulin signaling. J. Cell. Physiol. 226: 456–468, 2011.


PLOS ONE | 2015

A Randomized Trial of Pharmacogenetic Warfarin Dosing in Naïve Patients with Non-Valvular Atrial Fibrillation

Vittorio Pengo; Carlo-Federico Zambon; Paola Fogar; Andrea Padoan; Giovanni Nante; Michela Pelloso; Stefania Moz; Anna Chiara Frigo; Francesca Groppa; Dania Bozzato; Enrico Tiso; Elisa Gnatta; Gentian Denas; Seena Padayattil Jose; Roberto Padrini; Daniela Basso; Mario Plebani

Genotype-guided warfarin dosing have been proposed to improve patient’s management. This study is aimed to determine whether a CYP2C9- VKORC1- CYP4F2-based pharmacogenetic algorithm is superior to a standard, clinically adopted, pharmacodynamic method. Two-hundred naïve patients with non-valvular atrial fibrillation were randomized to trial arms and 180 completed the study. No significant differences were found in the number of out-of-range INRs (INR<2.0 or >3.0) (p = 0.79) and in the mean percentage of time spent in the therapeutic range (TTR) after 19 days in the pharmacogenetic (51.9%) and in the control arm (53.2%, p = 0.71). The percentage of time spent at INR>4.0 was significantly lower in the pharmacogenetic (0.7%) than in the control arm (1.8%) (p = 0.02). Genotype-guided warfarin dosing is not superior in overall anticoagulation control when compared to accurate clinical standard of care. Trial Registration ClinicalTrials.gov NCT01178034


Cell Communication and Signaling | 2014

Inflammation and pancreatic cancer: molecular and functional interactions between S100A8, S100A9, NT-S100A8 and TGFβ1

Daniela Basso; Dania Bozzato; Andrea Padoan; Stefania Moz; Carlo-Federico Zambon; Paola Fogar; Eliana Greco; Michele Scorzeto; Francesca Simonato; Filippo Navaglia; Matteo Fassan; Michela Pelloso; Sirio Dupont; Sergio Pedrazzoli; Ambrogio Fassina; Mario Plebani

BackgroundIn order to gain further insight on the crosstalk between pancreatic cancer (PDAC) and stromal cells, we investigated interactions occurring between TGFβ1 and the inflammatory proteins S100A8, S100A9 and NT-S100A8, a PDAC-associated S100A8 derived peptide, in cell signaling, intracellular calcium (Cai2+) and epithelial to mesenchymal transition (EMT). NF-κB, Akt and mTOR pathways, Cai2+ and EMT were studied in well (Capan1 and BxPC3) and poorly differentiated (Panc1 and MiaPaCa2) cell lines.ResultsNT-S100A8, one of the low molecular weight N-terminal peptides from S100A8 to be released by PDAC-derived proteases, shared many effects on NF-κB, Akt and mTOR signaling with S100A8, but mainly with TGFβ1. The chief effects of S100A8, S100A9 and NT-S100A8 were to inhibit NF-κB and stimulate mTOR; the molecules inhibited Akt in Smad4-expressing, while stimulated Akt in Smad4 negative cells. By restoring Smad4 expression in BxPC3 and silencing it in MiaPaCa2, S100A8 and NT-S100A8 were shown to inhibit NF-κB and Akt in the presence of an intact TGFβ1 canonical signaling pathway. TGFβ1 counteracted S100A8, S100A9 and NT-S100A8 effects in Smad4 expressing, not in Smad4 negative cells, while it synergized with NT-S100A8 in altering Cai2+ and stimulating PDAC cell growth. The effects of TGFβ1 on both EMT (increased Twist and decreased N-Cadherin expression) and Cai2+ were antagonized by S100A9, which formed heterodimers with TGFβ1 (MALDI-TOF/MS and co-immuno-precipitation).ConclusionsThe effects of S100A8 and S100A9 on PDAC cell signaling appear to be cell-type and context dependent. NT-S100A8 mimics the effects of TGFβ1 on cell signaling, and the formation of complexes between TGFβ1 with S100A9 appears to be the molecular mechanism underlying the reciprocal antagonism of these molecules on cell signaling, Cai2+ and EMT.


Pancreas | 2010

Analogs of vitamin E epitomized by alpha-tocopheryl succinate for pancreatic cancer treatment: in vitro results induce caution for in vivo applications.

Eliana Greco; Daniela Basso; Elisa Fadi; Andrea Padoan; Paola Fogar; Carlo-Federico Zambon; Filippo Navaglia; Dania Bozzato; Stefania Moz; Sergio Pedrazzoli; Mario Plebani

Objectives: &agr;-Tocopheryl succinate (&agr;-TOS) is thought to be toxic only for cancer cells. We ascertained in vitro &agr;-TOS effects on pancreatic cancer (PC) and normal cell growth and verified whether the combination of nontoxic &agr;-TOS and 5-fluorouracil (5-FU) doses causes cancer cell death and whether &agr;-TOS effects are mediated by the proapoptotic proteins Bax/Bak and/or SMAD4/DPC4 status. Methods: Five PC cell lines, myoblasts, normal monocytes, wild-type (WT) and Bax/Bak double knockout mouse embryonic fibroblast (MEF) cells, and permanently SMAD4/DPC4-transfected PSN1 cells were cultured in 1% and 10% fetal calf serums (FCSs), without or with &agr;-TOS (5-500 &mgr;mol/L). Nontoxic 5-FU (0.0001 mmol/L) and &agr;-TOS alone or in combination were also evaluated. Results: Only PSN1 PC cell line, which had SMAD4/DPC4 homozygous deletion, was sensitive to nontoxic &agr;-TOS doses (5 &mgr;mol/L in 1% FCS and 50 &mgr;mol/L in 10% FCS). A 20-&mgr;mol/L &agr;-TOS inhibited MEF-WT, not MEF-double knockout growth. Only PSN1 cells were sensitive to nontoxic 5-FU and &agr;-TOS combination. SMAD4/DPC4 transfection restored PSN1 resistance to the effects of combined 5-FU and &agr;-TOS effects. Conclusions: Only a minority of PC cells are sensitive to the antiproliferative effects of &agr;-TOS, any sensitivity appearing to be correlated with SMAD4/DPC4 homozygous deletion and Bax/Bak expression.


Clinica Chimica Acta | 2016

Blood expression of matrix metalloproteinases 8 and 9 and of their inducers S100A8 and S100A9 supports diagnosis and prognosis of PDAC-associated diabetes mellitus.

Stefania Moz; Daniela Basso; Andrea Padoan; Dania Bozzato; Paola Fogar; Carlo-Federico Zambon; Michela Pelloso; Cosimo Sperti; Saula Vigili de Kreutzenberg; Claudio Pasquali; Sergio Pedrazzoli; Angelo Avogaro; Mario Plebani

BACKGROUND Based on the knowledge that matrix metalloproteinases (MMPs) and S100A8/A9 synergistically work in causing PDAC-associated type 2 diabetes mellitus (T2DM), we verified whether tissue and blood MMP8, MMP9, S100A8 and S100A9 expression might help in distinguishing PDAC among diabetics. METHODS Relative quantification of MMP8, MMP9, S100A8 and S100A9 mRNA was performed in tissues obtained from 8 PDAC, 4 chronic pancreatitis (ChrPa), 4 non-PDAC tumors and in PBMCs obtained from 30 controls, 43 T2DM, 41 ChrPa, 91 PDAC and 33 pancreatic-biliary tract tumors. RESULTS T2DM was observed in PDAC (66%), in pancreatic-biliary tract tumors (64%) and in ChrPa (70%). In diabetics, with or without PDAC, MMP9 tissue expression was increased (p<0.05). Both MMPs increased in PDAC and MMP9 increased also in pancreatic-biliary tract tumors PBMCs. In diabetics, MMP9 was independently associated with PDAC (p=0.025), but failed to enhance CA 19-9 discriminant efficacy. A highly reduced S100A9 expression, found in 7 PDAC, was significantly correlated with a reduced overall survival (p=0.015). CONCLUSIONS An increased expression of tissue and blood MMP9 reflects the presence of PDAC-associated diabetes mellitus. This finding fits with the hypothesized role of MMPs as part of the complex network linking cancer to diabetes.


Clinica Chimica Acta | 2013

Chemiluminescence and ELISA-based serum assays for diagnosing and monitoring celiac disease in children: A comparative study

Ada Aita; Elisa Rossi; Daniela Basso; Graziella Guariso; Dania Bozzato; Michela Pelloso; Matilde Pescarin; Carlo-Federico Zambon; Filippo Navaglia; Eliana Greco; Marco Gasparetto; Paola Fogar; Andrea Padoan; Stefania Moz; Mario Plebani

BACKGROUND Anti-transglutaminase (tTG) or anti-deamidated gliadin peptides (DGP) serum determination is the first step in diagnosing celiac disease (CD). Our aims were to: compare the performance of novel chemiluminescent tool in the detection of tTG and DGP (Q-Flash®, Inova) with that of the established ELISA (Q-Lite®, Inova) methods; identify the more reliable index for making a sound diagnosis and monitoring therapy. METHODS Using Q-Flash® and Q-Lite®, IgA and IgG class tTG and DGP were measured in the sera of 155 CD pediatric patients and 166 healthy age-matched controls. Forty-two of the patients had a follow-up one year after starting gluten free diet (GFD). RESULTS Q-Flash® IgA tTG, the more accurate (intra-assay CV for low, intermediate and high values: 2.2%, 1.6%, and 1.1%; inter-assay CV: 2.8%, 4%, and 3%), sensitive (96.1%) and specific (97%) test for diagnosing CD, was the only variable to be significantly correlated with CD at binary logistic regression analysis (r=0.263, p<0.0001, Exp(B)=1.0506, 95% CI=1.0286-1.0731). Q-Flash® IgA tTG or DGP screen were more accurate than Q-Lite® IgA tTG in monitoring CD patients on GFD (p=0.003). CONCLUSION Q- Flash® IgA tTG measurement is an extremely precise, sensitive and specific index for not only diagnosing CD, but also monitoring therapy.

Collaboration


Dive into the Dania Bozzato'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge