Gianluigi Zaza
University of Verona
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Gianluigi Zaza.
Journal of Clinical Investigation | 2005
Leo Kager; Meyling Cheok; Wenjian Yang; Gianluigi Zaza; Qing Cheng; John C. Panetta; Ching-Hon Pui; James R. Downing; Mary V. Relling; William E. Evans
The ability of leukemia cells to accumulate methotrexate polyglutamate (MTXPG) is an important determinant of the antileukemic effects of methotrexate (MTX). We measured in vivo MTXPG accumulation in leukemia cells from 101 children with acute lymphoblastic leukemia (ALL) and established that B-lineage ALL with either TEL-AML1 or E2A-PBX1 gene fusion, or T-lineage ALL, accumulates significantly lower MTXPG compared with B-lineage ALL without these genetic abnormalities or compared with hyperdiploid (fewer than 50 chromosomes) ALL. To elucidate mechanisms underlying these differences in MTXPG accumulation, we used oligonucleotide microarrays to analyze expression of 32 folate pathway genes in diagnostic leukemia cells from 197 children. This revealed ALL subtype-specific patterns of folate pathway gene expression that were significantly related to MTXPG accumulation. We found significantly lower expression of the reduced folate carrier (SLC19A1, an MTX uptake transporter) in E2A-PBX1 ALL, significantly higher expression of breast cancer resistance protein (ABCG2, an MTX efflux transporter) in TEL-AML1 ALL, and lower expression of FPGS (which catalyzes formation of MTXPG) in T-lineage ALL, consistent with lower MTXPG accumulation in these ALL subtypes. These findings reveal distinct mechanisms of subtype-specific differences in MTXPG accumulation and point to new strategies to overcome these potential causes of treatment failure in childhood ALL.
BMC Genomics | 2009
Simona Granata; Gianluigi Zaza; Simona Simone; Gaetano Villani; Dominga Latorre; Paola Pontrelli; Massimo Carella; Francesco Paolo Schena; Giuseppe Grandaliano; Giovanni Pertosa
BackgroundChronic renal disease (CKD) is characterized by complex changes in cell metabolism leading to an increased production of oxygen radicals, that, in turn has been suggested to play a key role in numerous clinical complications of this pathological condition. Several reports have focused on the identification of biological elements involved in the development of systemic biochemical alterations in CKD, but this abundant literature results fragmented and not exhaustive.ResultsTo better define the cellular machinery associated to this condition, we employed a high-throughput genomic approach based on a whole transcriptomic analysis associated with classical molecular methodologies. The genomic screening of peripheral blood mononuclear cells revealed that 44 genes were up-regulated in both CKD patients in conservative treatment (CKD, n = 9) and hemodialysis (HD, n = 17) compared to healthy subjects (HS, n = 8) (p < 0.001, FDR = 1%). Functional analysis demonstrated that 11/44 genes were involved in the oxidative phosphorylation system. Western blotting for COXI and COXIV, key constituents of the complex IV of oxidative phosphorylation system, performed on an independent testing-group (12 healthy subjects, 10 CKD and 14 HD) confirmed an higher synthesis of these subunits in CKD/HD patients compared to the control group. Only for COXI, the comparison between CKD and healthy subjects reached the statistical significance. However, complex IV activity was significantly reduced in CKD/HD patients compared to healthy subjects (p < 0.01). Finally, CKD/HD patients presented higher reactive oxygen species and 8-hydroxydeoxyguanosine levels compared to controls.ConclusionTaken together these results suggest, for the first time, that CKD/HD patients may have an impaired mitochondrial respiratory system and this condition may be both the consequence and the cause of an enhanced oxidative stress.
The FASEB Journal | 2010
Fabio Sallustio; Luca De Benedictis; Giuseppe Castellano; Gianluigi Zaza; Antonia Loverre; Vincenzo Costantino; Giuseppe Grandaliano; Francesco Paolo Schena
In the past few years, adult renal progenitor/stem cells (ARPCs) have been identified in human kidneys, and particularly in Bowmans capsule and proximal tubules. They may play an important role in the kidney regenerative processes and might prospectively be the ideal cell type for the treatment of both acute and chronic renal injury. In this study, microarray analysis identified 6 gene clusters that discriminated normal human glomerular and tubular ARPCs from renal proximal tubular epithelial cells and mesenchymal stem cells. The top‐scored pathway in the ARPC gene expression profile contained growth factor receptors and immune system‐related genes, including tolllike receptor 2 (TLR2). Stimulation of TLR2 by ligands that mime inflammatory mediators or damage associated molecular pattern molecules induced secretion of elevated amounts of monocyte chemoattractant protein‐1 (MCP‐1), IL‐6, IL‐8, and C3 via NF‐κB activation. TLR2 stimulation also increased the ARPC proliferation rate, suggesting a role for TLR2 in ARPC activation via autocrine signaling. Moreover, TLR2 stimulation improved ARPC differentiation into renal epithelial cells and was responsible of ARPC branching morphogenesis and tubule‐like structures formation. For the first time, this study provides a genomic characterization of renal multipotent progenitor cells and shows that TLR2 found on ARPCs might be responsible for their activation in the kidney, orchestrating the activation of crucial signaling networks necessary for renal repair.—Sallustio, F., De Benedictis, L., Castellano, G., Zaza, G., Loverre, A., Costantino, V., Grandaliano, G., Schena, F. P. TLR2 plays a role in the activation of human resident renal stem/progenitor cells. FASEB J. 24, 514–525 (2010). www.fasebj.org
Pharmacogenetics and Genomics | 2011
Torben S. Mikkelsen; Caroline F. Thorn; Jun Yang; Cornelia M. Ulrich; Deborah L. French; Gianluigi Zaza; Henry M. Dunnenberger; Sharon Marsh; Howard L. McLeod; Kathy Giacomini; Mara L. Becker; Roger Gaedigk; J.S. Leeder; Leo Kager; Mary V. Relling; William E. Evans; Teri E. Klein; Russ B. Altman
Methotrexate is a folate analog that is used in the treatment of cancers (e.g. acute lymphoblastic leukemia, non-Hodgkin lymphoma, osteosarcoma, and colon cancer) and autoimmune diseases (e.g. rheumatoid arthritis, Crohn’s disease, and psoriasis). In the treatment of autoimmune diseases, methotrexate is usually administrated orally or subcutaneously, whereas in the cancer treatment, it can be given orally, intramuscularly, as intrathecal injections, or as intravenous infusions (up to 12 g/m2) [1-3]. The pharmacokinetics and pharmacodynamics of methotrexate show large interpatient variability regardless of the route of administration or disease being treated [4-6]. The goal of this study is to provide an introduction to methotrexate pharmacogenomics, showing the candidate genes in the PharmGKB methotrexate pathway (Fig. 1), important variants (Tables (Tables11 and and2),2), discussing key knowledge, and pointing to more in-depth resources.
Transplantation | 2008
Paola Pontrelli; Michele Rossini; Barbara Infante; Giovanni Stallone; Antonio Schena; Antonia Loverre; Michele Ursi; Raffaella Verrienti; Annamaria Maiorano; Gianluigi Zaza; Elena Ranieri; Loreto Gesualdo; Pasquale Ditonno; Carlo Bettocchi; Francesco Paolo Schena; Giuseppe Grandaliano
Background. Chronic allograft nephropathy (CAN) is characterized by deposition of extracellular matrix (ECM) in all renal compartments. PAI-1 seems to play a pivotal role in ECM turnover in CAN. Rapamycin has been shown to improve long-term graft survival in patients with CAN. The aim of the study was to evaluate the molecular mechanisms underlying the beneficial effects of rapamycin on CAN progression at glomerular and tubulointerstitial level. Methods. After a biopsy-proven CAN diagnosis (T0), 18 patients on calcineurin inhibitors (CNI) were randomly assigned in a 2:1 ratio to continue CNI (6 patients) or to receive rapamycin (RAPA; 12 patients). After 2 years of treatment (T24), all patients underwent a second renal biopsy. Morphometric analysis was conducted at T0 and at T24. PAI-1 expression was evaluated at T0 and T24 by immunohistochemistry. We evaluated the effect of rapamycin on PAI-1 gene expression in cultured proximal tubular cells incubated with CD40L or thrombin, two potential CAN pathogenic mediators. Results. The RAPA group showed a significant regression of glomerulosclerotic lesions and only a 26% increase in interstitial fibrosis after 2 years compared to baseline, whereas the CNI group showed progression of glomerulosclerosis and 112% increase in fibrosis. Glomerular and tubulointerstitial PAI-1 expression was reduced compared to the baseline in the RAPA group, while they were unchanged in the CNI group. In vitro data showed that rapamycin significantly reduced PAI-1 gene expression induced by both CD40L and thrombin in proximal tubular epithelial cells. Conclusions. These data suggest that rapamycin may modulate ECM deposition in CAN reducing PAI-1 expression.
Kidney International | 2010
Sharon Natasha Cox; Fabio Sallustio; Grazia Serino; Paola Pontrelli; Raffaella Verrienti; Francesco Pesce; Diletta Domenica Torres; Nicola Ancona; Patrizia Stifanelli; Gianluigi Zaza; Francesco Paolo Schena
Immunoglobulin A nephropathy (IgAN) is the most common form of primary glomerulonephritis worldwide. The basic defect lies within the IgA immune system and in peripheral blood leukocytes, rather than local kidney abnormalities. To define the intracellular mechanisms leading to the disease, we conducted a microarray study to identify genes and pathways differentially modulated in peripheral blood leukocytes isolated from 12 IgAN patients and 8 healthy controls. The genes whose expression discriminated between the IgAN patients and controls were primarily involved in canonical WNT-beta-catenin and PI3K/Akt pathways. We also tested peripheral blood mononuclear cells and their subpopulations isolated from an independent group of IgAN patients and healthy controls. There were low protein levels of inversin and PTEN, key regulators of WNT-beta-catenin and PI3K/Akt, in IgAN patients, suggesting hyperactivation of these pathways. Also, there were increased phospho-Akt protein levels and nuclear beta-catenin accumulation with an enhanced peripheral blood mononuclear cell proliferation rate. Subpopulation analysis uncovered a major irregularity of WNT signaling in monocytes. Hence, hyperactivation of these pathways may provide insight into mechanisms contributing to the pathogenesis of IgAN.
Journal of Translational Medicine | 2012
Valentina Masola; Maurizio Onisto; Gianluigi Zaza; Antonio Lupo; Giovanni Gambaro
BackgroundEpithelial-mesenchymal transition of tubular cells is a widely recognized mechanism that sustains interstitial fibrosis in diabetic nephropathy (DN). The signaling of FGF-2, a growth factor involved in this mechanism, is regulated by glycosaminoglycans. Heparanase-1, an endoglycosidase that cleaves heparan sulfate, is implicated in the pathogenesis of diabetic nephropathy and is necessary to FGF-2 for the induction of tubular cells transition. Well known Heparanase-1 inhibitors are heparin(s) and sulodexide, a low-molecular weight heparin – dermatan sulphate blend, which is effective in the treatment of DN.MethodsWe have investigated the inhibition by sulodexide and its components of Heparanase-1 by an ELISA assay. We have analyzed its effect on the epithelial-mesenchymal transition of tubular cells by real time gene expression analysis, zymography and migration assay.ResultsResults show that sulodexide is an effective heparanase-1 inhibitor, exclusively in virtue to the heparin component, with an IC50 of 5 μg/ml. In FGF-2 treated tubular cells, sulodexide also prevents the over-expression of the mesenchymal markers αSMA, vimentin and fibronectin and the motility increase, i.e. the epithelial-mesenchymal transition of tubular cells. Moreover, sulodexide prevents FGF-2 induced heparanase-1 and MMP9 increase switching off the autocrine loop that FGF-2 activates to support its signal.ConclusionsThe findings highlight the capacity of sulodexide to inhibit heparanase-1 and to control tubular fibrosis triggered by epithelial-mesenchymal transition. In conclusion, these sulodexide activities support the value of this agent in controlling the progression of nephropathy to renal failure.
Transplant International | 2011
Antonia Loverre; Chiara Divella; Giuseppe Castellano; Tiziana Tataranni; Gianluigi Zaza; Michele Rossini; Pasquale Ditonno; Michele Battaglia; Silvano Palazzo; Margherita Gigante; Elena Ranieri; Francesco Paolo Schena; Giuseppe Grandaliano
Ischemia‐reperfusion injury (IRI) in kidney transplantation is the major cause of delayed graft function (DGF), an event associated with an increased risk of acute rejection. The aim of this study was to evaluate T helper (Th) cell phenotype in renal transplants with DGF. T‐bet (Th1), GATA‐3 (Th2) and IL‐17 (Th17) protein expression was investigated in pretransplant biopsies, DGF and acute tubular damage (ATD) caused by calcineurin‐inhibitor toxicity. Intracytofluorimetric analysis of IFN‐γ, IL‐4 and IL‐17 was performed to analyze Th1, Th2 and Th17 responses in peripheral blood mononuclear cells of recipients with early graft function (EGF) and DGF, before (T0) and 24 h after transplantation (T24). In pretransplant biopsies, T‐bet+, GATA‐3+ and IL‐17+ cells were barely detectable. In DGF, T‐bet+ and IL‐17+ cells were significantly increased compared with pretransplant and ATD. More than 90% of T‐bet+ and less then 5% of IL‐17+ cells were CD4+. GATA‐3+ cells were increased to a lower extent. T‐bet+/GATA‐3+ cell ratio was significantly higher in DGF. Peripheral CD4+ IFN‐γ/IL‐4 ratio was significantly decreased in DGF, while CD4+/IL‐17+ cells did not differ between T0 and T24 in DGF. Our data suggest that DGF is characterized by a prevalent Th1 phenotype within the graft. This event might represent a link between DGF and acute rejection.
PLOS ONE | 2015
Simona Granata; Valentina Masola; Elisa Zoratti; Maria Teresa Scupoli; Anna Baruzzi; Michele Messa; Fabio Sallustio; Loreto Gesualdo; Antonio Lupo; Gianluigi Zaza
To assess whether NLR pyrin domain-containing protein 3 (NLRP3) inflammasome, a multiprotein complex that mediates the activation of caspase-1 (CASP-1) and pro-inflammatory cytokines IL-18 and IL-1β, could be involved in the chronic inflammatory state observed in chronic kidney disease patients undergoing hemodialysis treatment (CKD-HD), we employed several biomolecular techniques including RT-PCR, western blot, FACS analysis, confocal microscopy and microarray. Interestingly, peripheral blood mononuclear cells from 15 CKD-HD patients showed higher mRNA levels of NLRP3, CASP-1, ASC, IL-1β, IL-18 and P2X7receptor compared to 15 healthy subjects. Western blotting analysis confirmed the above results. In particular, active forms of CASP-1, IL1-β and IL-18 resulted significantly up-regulated in CKD-HD versus controls. Additionally, elevated mitochondrial ROS level, colocalization of NLRP3/ASC/mitochondria in peripheral blood mononuclear cells from CKD-HD patients and down-regulation of CASP-1, IL1-β and IL-18 protein levels in immune-cells of CKD-HD patients stimulated with LPS/ATP in presence of mitoTEMPO, inhibitor of mitochondrial ROS production, suggested a possible role of this organelle in the aforementioned CKD-associated inflammasome activation. Then, microarray analysis confirmed, in an independent microarray study cohort, that NLRP3 and CASP-1, along with other inflammasome-related genes, were up-regulated in 17 CKD-HD patients and they were able to clearly discriminate these patients from 5 healthy subjects. All together these data showed, for the first time, that NLRP3 inflammasome was activated in uremic patients undergoing dialysis treatment and they suggested that this unphysiological condition could be possibly induced by mitochondrial dysfunction.
Clinical & Developmental Immunology | 2013
Gianluigi Zaza; Paola Tomei; Paolo Ria; Simona Granata; Luigino Boschiero; Antonio Lupo
The mammalian target of rapamycin inhibitors (mTOR-I), sirolimus and everolimus, are immunosuppressive drugs largely used in renal transplantation. The main mechanism of action of these drugs is the inhibition of the mammalian target of rapamycin (mTOR), a regulatory protein kinase involved in lymphocyte proliferation. Additionally, the inhibition of the crosstalk among mTORC1, mTORC2, and PI3K confers the antineoplastic activities of these drugs. Because of their specific pharmacological characteristics and their relative lack of nephrotoxicity, these inhibitors are valid option to calcineurine inhibitors (CNIs) for maintenance immunosuppression in renal transplant recipients with chronic allograft nephropathy. However, as other immunosuppressive drugs, mTOR-I may induce the development of several adverse effects that need to be early recognized and treated to avoid severe illness in renal transplant patients. In particular, mTOR-I may induce systemic nonnephrological side effects including pulmonary toxicity, hematological disorders, dysmetabolism, lymphedema, stomatitis, cutaneous adverse effects, and fertility/gonadic toxicity. Although most of the adverse effects are dose related, it is extremely important for clinicians to early recognize them in order to reduce dosage or discontinue mTOR-I treatment avoiding the onset and development of severe clinical complications.