Network


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

Hotspot


Dive into the research topics where Roberto Orsatti is active.

Publication


Featured researches published by Roberto Orsatti.


Oncogene | 1999

Activation and repression of the 2-5A synthetase and p21 gene promoters by IRF-1 and IRF-2

Eliana M. Coccia; Nicoletta Del Russo; Emilia Stellacci; Roberto Orsatti; Eleonora Benedetti; Giovanna Marziali; John Hiscott; Angela Battistini

The Interferon Regulatory Factors-1 and -2 (IRF-1 and IRF-2) were originally identified as transcriptional regulators of the interferon (IFN) and IFN-stimulated genes. These factors also modulate immune response and play a role in cell growth regulation. In this study we analysed the effect of the ectopic expression of IRF-1 and IRF-2 on the regulation of two potential IRF target genes involved in cell growth regulation, 2-5A synthetase and p21 (WAF/CP1), both of which contain consensus binding sites for IRF family members within their promoters. Following ectopic expression, IRF-1 transactivated 2-5A synthetase and p21 genes, an effect that was counterbalanced by concomitant ectopic expression of IRF-2. These effects were mediated by direct binding of IRF to the gene promoters. A construct expressing an IRF-2 antisense (FRI-2) was able to revert the inhibitory effect of IRF-2 on the IRF-1 transactivation. IRF-1 also induced expression of its homologous repressor IRF-2 as indicated by EMSA analysis using an IRF-E probe from the IRF-2 promoter; and by cotransfection of IRF-1 together with an IRF-2 promoter CAT construct. Therefore, the induction of IRF-1 by IFNs or other stimuli acts as a transactivator of genes involved in cell growth regulation, as well as of its own repressor IRF-2, thus providing autoinhibitory regulation of IRF-1 activated genes.


Journal of Virology | 2008

IRF-1 Is Required for Full NF-κB Transcriptional Activity at the Human Immunodeficiency Virus Type 1 Long Terminal Repeat Enhancer

Marco Sgarbanti; Anna Lisa Remoli; Giulia Marsili; Barbara Ridolfi; Alessandra Borsetti; Edvige Perrotti; Roberto Orsatti; Ramona Ilari; Leonardo Sernicola; Emilia Stellacci; Barbara Ensoli; Angela Battistini

ABSTRACT Human immunodeficiency virus type 1 (HIV-1) gene expression is controlled by a complex interplay between viral and host factors. We have previously shown that interferon-regulatory factor 1 (IRF-1) is stimulated early after HIV-1 infection and regulates promoter transcriptional activity even in the absence of the viral transactivator Tat. In this work we demonstrate that IRF-1 is also required for full NF-κB transcriptional activity. We provide evidence that IRF-1 and NF-κB form a functional complex at the long terminal repeat (LTR) κB sites, which is abolished by specific mutations in the two adjacent κB sites in the enhancer region. Silencing IRF-1 with small interfering RNA resulted in impaired NF-κB-mediated transcriptional activity and in repressed HIV-1 transcription early in de novo-infected T cells. These data indicate that in early phases of HIV-1 infection or during virus reactivation from latency, when the viral transactivator is absent or present at very low levels, IRF-1 is an additional component of the p50/p65 heterodimer binding the LTR enhancer, absolutely required for efficient HIV-1 replication.


Leukemia | 2004

Impaired myelopoiesis in mice devoid of interferon regulatory factor 1

Ugo Testa; Emilia Stellacci; Elvira Pelosi; Paola Sestili; Venditti M; Roberto Orsatti; A Fragale; Eleonora Petrucci; Luca Pasquini; Filippo Belardelli; Lucia Gabriele; Angela Battistini

Interferon regulatory factor (IRF)-1 is a transcription factor controlling the expression of several genes, which are differentially induced depending on the cell type and signal. IRF-1 modulates multiple functions, including regulation of immune responses and host defence, cell growth, cytokine signalling and hematopoietic development. Here, we investigated the role of IRF-1 in granulocytic differentiation in mice with a null mutation in the IRF-1 gene. We show that IRF-1−/− bone marrow cells exhibit an increased number of immature granulocytic precursors, associated with a decreased number of mature granulocytic elements as compared to normal mice, suggestive of a defective maturation process. Clonogenetic analyses revealed a reduced number of CFU-G, CFU-M and CFU-GM colonies in IRF-1−/− mice, while the number of BFU-E/CFU-E colonies was unchanged. At the molecular level, the expression of CAAT-enhancer-binding protein (C/EBP)-ɛ, -α and PU.1 was substantially lower in the CD11b+ cells from the bone marrow of IRF-1−/− mice as compared to cells from wild-type mice. These results, together with the fact that IRF-1 is markedly induced early during granulo-monocytic differentiation of CD34+ cells, highlight the pivotal role of IRF-1 in the early phases of myelopoiesis.


Biochemical Journal | 2004

Interferon regulatory factor-2 drives megakaryocytic differentiation.

Emilia Stellacci; Ugo Testa; Eleonora Petrucci; Eleonora Benedetti; Roberto Orsatti; Tiziana Feccia; Marit Stafsnes; Eliana M. Coccia; Giovanna Marziali; Angela Battistini

IRFs [IFN (interferon) regulatory factors] constitute a family of transcription factors involved in IFN signalling and in the development and differentiation of the immune system. IRF-2 has generally been described as an antagonist of IRF-1-mediated transcription of IFN and IFN-inducible genes; however, it has been recently identified as a transcriptional activator of some genes, such as those encoding histone H4, VCAM-1 (vascular cell adhesion molecule-1) and Fas ligand. Biologically, IRF-2 plays an important role in cell growth regulation and has been shown to be a potential oncogene. Studies in knock-out mice have also implicated IRF-2 in the differentiation and functionality of haematopoietic cells. Here we show that IRF-2 expression in a myeloid progenitor cell line leads to reprogramming of these cells towards the megakaryocytic lineage and enables them to respond to thrombopoietin, as assessed by cell morphology and expression of specific differentiation markers. Up-regulation of transcription factors involved in the development of the megakaryocytic lineage, such as GATA-1, GATA-2, FOG-1 (friend of GATA-1) and NF-E2 (nuclear factor-erythroid-2), and transcriptional stimulation of the thrombopoietin receptor were also demonstrated. Our results provide evidence for a key role for IRF-2 in the induction of a programme of megakaryocytic differentiation, and reveal a remarkable functional diversity of this transcription factor in the regulation of cellular responses.


Journal of Immunology | 2011

Critical Role of IRF-8 in Negative Regulation of TLR3 Expression by Src Homology 2 Domain-Containing Protein Tyrosine Phosphatase-2 Activity in Human Myeloid Dendritic Cells

Alessandra Fragale; Emilia Stellacci; Ramona Ilari; Anna Lisa Remoli; Angela Lanciotti; Edvige Perrotti; Iart Luca Shytaj; Roberto Orsatti; Harshani R. Lawrence; Nicholas J. Lawrence; Jerry Wu; Michael Rehli; Keiko Ozato; Angela Battistini

Despite extensive studies that unraveled ligands and signal transduction pathways triggered by TLRs, little is known about the regulation of TLR gene expression. TLR3 plays a crucial role in the recognition of viral pathogens and induction of immune responses by myeloid DCs. IFN regulatory factor (IRF)-8, a member of the IRF family, is a transcriptional regulator that plays essential roles in the development and function of myeloid lineage, affecting different subsets of myeloid DCs. In this study, we show that IRF-8 negatively controls TLR3 gene expression by suppressing IRF-1– and/or polyinosinic-polycytidylic acid-stimulated TLR3 expression in primary human monocyte-derived DCs (MDDCs). MDDCs expressed TLR3 increasingly during their differentiation from monocytes to DCs with a peak at day 5, when TLR3 expression was further enhanced upon stimulation with polyinosinic-polycytidylic acid and then was promptly downregulated. We found that both IRF-1 and IRF-8 bind the human TLR3 promoter during MDDC differentiation in vitro and in vivo but with different kinetic and functional effects. We demonstrate that IRF-8–induced repression of TLR3 is specifically mediated by ligand-activated Src homology 2 domain-containing protein tyrosine phosphatase association. Indeed, Src homology 2 domain-containing protein tyrosine phosphatase–dephosphorylated IRF-8 bound to the human TLR3 promoter competing with IRF-1 and quashing its activity by recruitment of histone deacetylase 3. Our findings identify IRF-8 as a key player in the control of intracellular viral dsRNA-induced responses and highlight a new mechanism for negative regulation of TLR3 expression that can be exploited to block excessive TLR activation.


Annals of the New York Academy of Sciences | 2007

IRF-7: new role in the regulation of genes involved in adaptive immunity.

Marco Sgarbanti; Giulia Marsili; Anna Lisa Remoli; Roberto Orsatti; Angela Battistini

Abstract:  The interferon regulatory factor 7 (IRF‐7), a member of the IRF family of transcription factors, is a key player in the innate immune response against viral infections. Constitutive expression of IRF‐7 is limited to peripheral blood lymphocytes and dendritic cells while in most cell types its expression can be induced by type I interferon (INF). IRF‐7 is sequestered in the cytoplasm of uninfected cells and following viral infection, double‐stranded RNA (dsRNA), or toll‐like receptor (TLR) signaling, it becomes phosphorylated by TBK and IKK‐i kinases. Phosphorylated IRF‐7 migrates in the nucleus where it can activate IFN type I genes and other interferon‐stimulated genes (ISGs). Here we report that the overexpression of a constitutively active form of IRF‐7 binds and positively regulates the transcriptional activity of the promotor of IRF‐1 and low molecular mass polypeptide‐2 (LMP‐2), two proteins that play a key role in adaptive immunity. The so far unrecognized role of IRF‐7 in LMP‐2 stimulation points to IRF‐7 as a transcriptional regulator that bridges innate and adaptive immunity.


Molecular and Cellular Biology | 2014

IκB kinase ε targets interferon regulatory factor 1 in activated T lymphocytes.

Marco Sgarbanti; Giulia Marsili; Anna Lisa Remoli; Emilia Stellacci; Antonello Mai; Dante Rotili; Edvige Perrotti; Chiara Acchioni; Roberto Orsatti; Nunzio Iraci; Mathieu Ferrari; Alessandra Borsetti; John Hiscott; Angela Battistini

ABSTRACT IκB kinase ε (IKK-ε) has an essential role as a regulator of innate immunity, functioning downstream of pattern recognition receptors to modulate NF-κB and interferon (IFN) signaling. In the present study, we investigated IKK-ε activation following T cell receptor (TCR)/CD28 stimulation of primary CD4+ T cells and its role in the stimulation of a type I IFN response. IKK-ε was activated following TCR/CD28 stimulation of primary CD4+ T cells; however, in T cells treated with poly(I·C), TCR/CD28 costimulation blocked induction of IFN-β transcription. We demonstrated that IKK-ε phosphorylated the transcription factor IFN regulatory factor 1 (IRF-1) at amino acid (aa) 215/219/221 in primary CD4+ T cells and blocked its transcriptional activity. At the mechanistic level, IRF-1 phosphorylation impaired the physical interaction between IRF-1 and the NF-κB RelA subunit and interfered with PCAF-mediated acetylation of NF-κB RelA. These results demonstrate that TCR/CD28 stimulation of primary T cells stimulates IKK-ε activation, which in turn contributes to suppression of IFN-β production.


Archive | 1996

Iron Regulation of Transferrin Receptor and Ferritin Expression in Differentiating Friend Leukemia Cells

Eliana M. Coccia; Emilia Stellacci; Giovanna Marziali; Roberto Orsatti; Edvige Perrotti; Nicoletta Del Russo; Ugo Testa; Angela Battistini

Transferrin receptor (TfR) and ferritin expression has been investigated in Friend erythroleukemia cells (FLCs) induced to differentiate by dimethylsulfoxide (Me2SO). In differentiating FLCs, administration of hemin increases the ferritin content approximately 20–25 fold; conversely, iron salts have only mild stimulatory effects on ferritin accumulation and iron chelators only slightly inhibit the stimulatory effect exerted by hemin. Moreover, in Me2SO-induced FLC, the negative feedback reported in a variety of other cell types for the regulation of TfR expression by heme is not operative. We conclude that in FLCs induced to differentiate, hemin acts synergistically with the differentiation inducer increasing ferritin expression and is not able to down modulate the TfR number. These effects appear characteristic of differentiating erythroid cells since they are not observed in other cell types (i.e. fibroblastic cell lines).


Viruses | 2018

Development and Validation of a Novel Dual Luciferase Reporter Gene Assay to Quantify Ebola Virus VP24 Inhibition of IFN Signaling

Elisa Fanunza; Aldo Frau; Marco Sgarbanti; Roberto Orsatti; Angela Corona; Enzo Tramontano

The interferon (IFN) system is the first line of defense against viral infections. Evasion of IFN signaling by Ebola viral protein 24 (VP24) is a critical event in the pathogenesis of the infection and, hence, VP24 is a potential target for drug development. Since no drugs target VP24, the identification of molecules able to inhibit VP24, restoring and possibly enhancing the IFN response, is a goal of concern. Accordingly, we developed a dual signal firefly and Renilla luciferase cell-based drug screening assay able to quantify IFN-mediated induction of Interferon Stimulated Genes (ISGs) and its inhibition by VP24. Human Embryonic Kidney 293T (HEK293T) cells were transiently transfected with a luciferase reporter gene construct driven by the promoter of ISGs, Interferon-Stimulated Response Element (ISRE). Stimulation of cells with IFN-α activated the IFN cascade leading to the expression of ISRE. Cotransfection of cells with a plasmid expressing VP24 cloned from a virus isolated during the last 2014 outbreak led to the inhibition of ISRE transcription, quantified by a luminescent signal. To adapt this system to test a large number of compounds, we performed it in 96-well plates; optimized the assay analyzing different parameters; and validated the system by calculating the Z′- and Z-factor, which showed values of 0.62 and 0.53 for IFN-α stimulation assay and VP24 inhibition assay, respectively, indicative of robust assay performance.


Virology | 1990

A full-length murine 2-5A synthetase cDNA transfected in NIH-3T3 cells impairs EMCV but not VSV replication

Eliana M. Coccia; Giovanna Romeo; Ahuva Nissim; Giovanna Marziali; Roberto Albertini; Elisabetta Affabris; Angela Battistini; Gianna Fiorucci; Roberto Orsatti; Giovanni B. Rossi; Judith Chebath

Collaboration


Dive into the Roberto Orsatti's collaboration.

Top Co-Authors

Avatar

Angela Battistini

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Emilia Stellacci

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Edvige Perrotti

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Eliana M. Coccia

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Giovanna Marziali

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Ugo Testa

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Marco Sgarbanti

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Alessandra Borsetti

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Anna Lisa Remoli

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Ramona Ilari

Istituto Superiore di Sanità

View shared research outputs
Researchain Logo
Decentralizing Knowledge