Network


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

Hotspot


Dive into the research topics where V. Schinzari is active.

Publication


Featured researches published by V. Schinzari.


Journal of Hepatology | 2012

Transcriptional regulation of miR-224 upregulated in human HCCs by NFκB inflammatory pathways

C. Scisciani; Stefania Vossio; Francesca Guerrieri; V. Schinzari; Rossana De Iaco; Paolo D’Onorio de Meo; Melchiorre Cervello; Giuseppe Montalto; Teresa Pollicino; Giovanni Raimondo; Massimo Levrero; N. Pediconi

BACKGROUND & AIMS miR-224 is up-regulated in human HCCs as compared to both paired peri-tumoral cirrhotic tissues and cirrhotic livers without HCC. Here, we have cloned the miR-224 regulatory region and characterized its transcriptional regulation by the NFκB-dependent inflammatory pathways. METHODS Mature miRNA expression was evaluated by a 2 step stem-loop real-time RT-PCR. The recruitment of polymerase II and transcription factors on the pre-miR-224 promoter has been assessed by ChIPSeq and ChIP. RESULTS We found miR-224 levels strongly up-regulated in both peri-tumoral cirrhotic livers and HCC samples as compared to normal livers. In silico analysis of the putative miR-224 promoter revealed multiple NFκB sites. We showed that LTα and TNFα activate transcription from the miR-224 promoter and of endogenous miR-224 expression in HCC cell lines, whereas the expression of miR-224 target API5 was reduced. Exogenously expressed p65/RelA activates the miR-224 promoter and a dominant negative form of IκBα (IκBSR) represses it. ChIP analysis showed that p65/NFκB is recruited on the miR-224 promoter and that its binding sharply increases after exposure to LPS, TNFα, and LTα. Altogether these findings link the inflammatory signals to NFκB-mediated activation of miR-224 expression. An antago-miR specific for miR-224 blocked LPS and LTα stimulated HCC cells migration and invasion. Conversely, the IKK inhibitor BMS-345541 blocks pre-miR-224-induced cellular migration and invasion. CONCLUSIONS Our results identify p65/NFκB as a direct transcriptional regulator of miR-224 expression and link miR-224 up-regulation with the activation of the LPS, LTα, and TNFα inflammatory pathways and cell migration/invasion in HCC.


Molecular and Cellular Biology | 2009

hSirT1-dependent regulation of the PCAF-E2F1-p73 apoptotic pathway in response to DNA damage.

N. Pediconi; Francesca Guerrieri; Stefania Vossio; Tiziana Bruno; L. Belloni; V. Schinzari; C. Scisciani; Maurizio Fanciulli; Massimo Levrero

ABSTRACT The NAD+-dependent histone deacetylase hSirT1 regulates cell survival and stress responses by inhibiting p53-, NF-κB-, and E2F1-dependent transcription. Here we show that the hSirT1/PCAF interaction controls the E2F1/p73 apoptotic pathway. hSirT1 represses E2F1-dependent P1p73 promoter activity in untreated cells and inhibits its activation in response to DNA damage. hSirT1, PCAF, and E2F1 are corecruited in vivo on theP1p73 promoter. hSirT1 deacetylates PCAF in vitro and modulates PCAF acetylation in vivo. In cells exposed to apoptotic DNA damage, nuclear NAD+ levels decrease and inactivate hSirT1 without altering the hSirT1 interaction with PCAF and hSirT1 binding to the P1p73 promoter. The reactivation of hSirT1 by pyruvate that increases the [NAD+]/[NADH] ratio completely abolished the DNA damage-induced activation of TAp73 expression, thus linking the modulation of chromatin-bound hSirT1 deacetylase activity by the intracellular redox state with P1p73 promoter activity. The release of PCAF from hSirT1 repression favors the assembly of transcriptionally active PCAF/E2F1 complexes onto the P1p73 promoter and p53-independent apoptosis. Our results identify hSirT1 and PCAF as potential targets to modulate tumor cell survival and chemoresistance irrespective of p53 status.


Hepatology | 2014

Human OX40 tunes the function of regulatory T cells in tumor and nontumor areas of hepatitis C virus–infected liver tissue

Silvia Piconese; Eleonora Timperi; Ilenia Pacella; V. Schinzari; Claudio Tripodo; M. Rossi; Nicola Guglielmo; G. Mennini; Gian Luca Grazi; Simona Di Filippo; Stefania Brozzetti; Katia Fazzi; Guido Antonelli; Maria Antonietta Lozzi; Massimo Sanchez; Vincenzo Barnaba

Regulatory T cells (Tregs) can be considered as a mixed population of distinct subsets, endowed with a diverse extent and quality of adaptation to microenvironmental signals. Here, we uncovered an opposite distribution of Treg expansion, phenotype, and plasticity in different microenvironments in the same organ (liver) derived from patients with chronic hepatitis C: On the one side, cirrhotic and tumor fragments were moderately and highly infiltrated by Tregs, respectively, expressing OX40 and a T‐bethighIFN‐γ– “T‐helper (Th)1‐suppressing” phenotype; on the other side, noncirrhotic liver specimens contained low frequencies of Tregs that expressed low levels of OX40 and highly produced interferon‐gamma (IFN‐γ; T‐bet+IFN‐γ+), thus becoming “Th1‐like” cells. OX40‐expressing and Th1‐suppressing Tregs were enriched in the Helios‐positive subset, carrying highly demethylated Treg cell‐specific demethylated region that configures committed Tregs stably expressing forkhead box protein 3. OX40 ligand, mostly expressed by M2‐like monocytes and macrophages, boosted OX40+ Treg proliferation and antagonized the differentiation of Th1‐like Tregs. However, this signal is counteracted in noncirrhotic liver tissue (showing various levels of inflammation) by high availability of interleukin‐12 and IFN‐γ, ultimately leading to complete, full Th1‐like Treg differentiation. Conclusion: Our data demonstrate that Tregs can finely adapt, or even subvert, their classical inhibitory machinery in distinct microenvironments within the same organ. (Hepatology 2014;60:1494–1507)


Oncogene | 2011

p53-paralog DNp73 oncogene is repressed by IFNα/STAT2 through the recruitment of the Ezh2 polycomb group transcriptional repressor.

B. Testoni; V. Schinzari; Francesca Guerrieri; Sabine Gerbal-Chaloin; G Blandino; Massimo Levrero

The DNp73 proteins act as trans-repressors of p53 and p73-dependent transcription and exert both anti-apoptotic activity and pro-proliferative activity. DNp73s are frequently up-regulated in a variety of human cancers, including human hepatocellular carcinomas (HCCs). Increased levels of DNp73 proteins confer to HCC cells resistance to apoptosis and, irrespective to p53 status, a chemoresistant phenotype. Here, we show that interferon (IFN)α down-regulates DNp73 expression in primary human hepatocytes (PHHs) and HCC cell lines. IFNα has been used as pro-apoptotic agent in the treatment of malignancies and there is increasing evidence of IFNα effectiveness in HCC treatment and prevention of recurrence. The precise mechanisms by which class I IFNs exert their anti-proliferative and anti-tumor activity remain unclear. IFNα binding to its receptor activates multiple intracellular signaling cascades regulating the transcription of numerous direct target genes through the recruitment of a complex comprising of STAT1, STAT2 and IFN regulatory factor (IRF)9 to their promoters. We found that, in response to IFNα, the P2p73 promoter undergoes substantial chromatin remodeling. Histone deacetylases (HDACs) replace histone acetyl transferases. STAT2 is recruited onto the endogenous P2p73 promoter together with the polycomb group protein Ezh2, leading to increased H3K27 methylation and transcriptional repression. The reduction of DNp73 levels by IFNα is paralleled by an increased susceptibility to IFNα-triggered apoptosis of Huh7 hepatoma cells. Our results show, for the first time, that IFN-stimulated gene factor 3 recruitment may serve both in activating and repressing gene expression and identify the down-regulation of DNp73 as an additional mechanism to counteract the chemoresistance of liver cancer cells.


Cell Death and Disease | 2013

The sodium/iodide symporter NIS is a transcriptional target of the p53-family members in liver cancer cells

Francesca Guerrieri; Silvia Piconese; C Lacoste; V. Schinzari; Barbara Testoni; Yannick Valogne; Sabine Gerbal-Chaloin; Didier Samuel; Christian Bréchot; Jamila Faivre; Massimo Levrero

Thyroid iodide accumulation via the sodium/iodide symporter (NIS; SLC5A5) has been the basis for the longtime use of radio-iodide in the diagnosis and treatment of thyroid cancers. NIS is also expressed, but poorly functional, in some non-thyroid human cancers. In particular, it is much more strongly expressed in cholangiocarcinoma (CCA) and hepatocellular carcinoma (HCC) cell lines than in primary human hepatocytes (PHH). The transcription factors and signaling pathways that control NIS overexpression in these cancers is largely unknown. We identified two putative regulatory clusters of p53-responsive elements (p53REs) in the NIS core promoter, and investigated the regulation of NIS transcription by p53-family members in liver cancer cells. NIS promoter activity and endogenous NIS mRNA expression are stimulated by exogenously expressed p53-family members and significantly reduced by member-specific siRNAs. Chromatin immunoprecipitation analysis shows that the p53–REs clusters in the NIS promoter are differentially occupied by the p53-family members to regulate basal and DNA damage-induced NIS transcription. Doxorubicin strongly induces p53 and p73 binding to the NIS promoter, leading to an increased expression of endogenous NIS mRNA and protein in HCC and CCA cells, but not in PHH. Silencing NIS expression reduced doxorubicin-induced apoptosis in HCC cells, pointing to a possible role of a p53-family-dependent expression of NIS in apoptotic cell death. Altogether, these results indicate that the NIS gene is a direct target of the p53 family and suggests that the modulation of NIS by DNA-damaging agents is potentially exploitable to boost NIS upregulation in vivo.


Molecular Cancer Research | 2014

Dual Promoter Usage as Regulatory Mechanism of let-7c Expression in Leukemic and Solid Tumors

Andrea Pelosi; Silvia Careccia; Giulia Sagrestani; Simona Nanni; Isabella Manni; V. Schinzari; Joost H.A. Martens; Antonella Farsetti; Hendrik G. Stunnenberg; Maria Pia Gentileschi; Donatella Del Bufalo; Ruggero De Maria; Giulia Piaggio; Maria Giulia Rizzo

Let-7c, an intronic microRNA (miRNA) embedded in the long non-coding gene LINC00478, can act as a tumor suppressor by targeting oncogenes. Previous studies indicated that in acute promyelocytic leukemia (APL), a subtype of acute myelogenous leukemia (AML) bearing the leukemia promoting PML/RARα fusion protein, let-7c expression seems to be controlled by the host gene promoter, in which canonical Retinoic Acid Responsive Elements (RAREs) are bound by PML/RARα in an all transretinoic acid (ATRA)–sensitive manner. Here, let-7c transcriptional regulation was further investigated and a novel intronic promoter upstream of the pre-miRNA was identified. This new promoter has transcriptional activity strongly indicating that at least two promoters need to be considered for let-7c transcription: the distal host gene and the proximal intronic promoter. Therefore, epigenetic modifying enzymes and histone acetylation and methylation status were analyzed on both let-7c promoters. It was demonstrated that ATRA treatment leads to let-7c upregulation inducing a more open chromatin conformation of the host gene promoter, with an enrichment of epigenetic marks that correlate with a more active transcriptional state. Conversely, the epigenetic marks on the intronic promoter are not significantly affected by ATRA treatment. Interestingly, in solid tumors such as prostate and lung adenocarcinoma it was found that both host and intronic promoters are functional. These data suggest that while the host gene promoter may control let-7c expression in AML, in a nonleukemic tumor context instead the intronic promoter contributes or preferentially regulates let-7c transcription. Implications: Alternative promoter usage represents a regulatory mechanism of let-7c expression in different tissues. Mol Cancer Res; 12(6); 878–89. ©2014 AACR.


Journal of Hepatology | 2009

545 THE P53-PARALOG DNP73 ONCOGENE IS REPRESSED BY α-IFN/STAT2 THROUGH THE RECRUITMENT OF YY1 AND HDAC1 TRANSCRIPTIONAL REPRESSORS

B. Testoni; C. Scisciani; V. Schinzari; N. Pediconi; M. Levrero

ackground. Expression of the p53-paralog DNp73 oncoene increases progressively in chronic hepatitis, cirrhosis nd HCC and greatly contributes to the chemoresistant henotype of HCC cells. DNp73 isoforms do not activate ranscription and do not induce apoptosis but act as domiant negative inhibitors of both p53 and TAp73. DNp73s are xpressed from the intragenic P2p73 promoter and we have ecently shown that its activity is positively regulated in heptocytes by beta-catenin and p65/NFkB. In silico analisys of he P2p73 promoter indicates that it contains two conserved SRE elements. im. To characterize the effects of -interferon on DNp73 xpression and to evaluate the functional consequences on ell proliferation and viability. ethods. Chromatin was immunoprecipatated from ntreated and IFN treated Huh7 cells with anti-AcH3, nti-STAT2, anti-P-STAT2, anti-HDAC1, anti-p300, antiY1 and anti-Pol II antibodies. DNp73 mRNA levels were uantitated real-time PCR. esults. ChIP analysis showed that both ISRE sites are ound in vivo by Stat2 and by its phosphorilated form after h of IFN treatment. Endogenous DNp73 mRNA levels re downregulated by IFN . Immunoblot analysis showed a trong DNp73 protein downregulation after IFN-treatment. hIP analysis showed that, before the IFN treatment, H3 ysines are acetylated and p300 and Pol II are bound to the 2p73 promoter, mirroring active transcription, whereas after h treatment (when the ISGF3 complex is recruited), Pol II nd p300 binding is lost whereas HDAC1, a well-known trancription cofactor related to transcriptional repression, and he YY1 co-repressor are actively recruited. These results ndicate that the recruitment of the ISGF3 complex to the 2p73promoter has a repressive activity and suggest that this h o a a isease 41 (2009) A1–A19


Journal of Hepatology | 2011

310 CHROMATIN DYNAMICS OF GENES REPRESSION IN RESPONSE TO IFNa REVEAL NEW ROLES FOR STAT2, PHOSPHO-STAT2, HISTONE DEACETYLASES (HDACS) AND HISTONE METHYLATION MARKS

B. Testoni; C. Voellenke; F. Guerrieri; V. Schinzari; S. Gerbal; G. Blandino; M. Levrero


Archive | 2010

Genome-wide identification of HBx cellular target genes and transcriptional partners in HBV replicating HCC cells by ChIP-Seq.

Massimo Levrero; Teresa Pollicino; L. Belloni; Giovanni Blandino; Giovanni Raimondo; F. Guerrieri; V. Schinzari


Journal of Hepatology | 2010

870 EPIGENETIC REGULATION OF CELLULAR TARGET GENES BY HBX: A CHROMATIN-IMMUNOPRECIPITATION (CHIP AND CHIP-SEQ) STUDY TO DEFINE THE HBX TRANSCRIPTOME IN HBV REPLICATING HCC CELLS

F. Guerrieri; L. Belloni; V. Schinzari; C. Scisciani; Teresa Pollicino; G. Raimondo; M. Levrero

Collaboration


Dive into the V. Schinzari's collaboration.

Top Co-Authors

Avatar

Massimo Levrero

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

C. Scisciani

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

N. Pediconi

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

F. Guerrieri

Istituto Italiano di Tecnologia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

B. Testoni

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

L. Belloni

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge