Network


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

Hotspot


Dive into the research topics where Ramona Ilari is active.

Publication


Featured researches published by Ramona Ilari.


Molecular and Cellular Biology | 1997

Transcriptional regulation of the ferritin heavy-chain gene: the activity of the CCAAT binding factor NF-Y is modulated in heme-treated Friend leukemia cells and during monocyte-to-macrophage differentiation.

Giovanna Marziali; Edvige Perrotti; Ramona Ilari; Ugo Testa; Eliana M. Coccia; Angela Battistini

The ferritin H-chain gene promoter regulation was analyzed in heme-treated Friend leukemia cells (FLCs) and during monocyte-to-macrophage differentiation. In the majority of cell lines studied, the regulation of ferritin expression was exerted mostly at the translational level. However, in differentiating erythroid cells, which must incorporate high levels of iron to sustain hemoglobin synthesis, and in macrophages, which are involved in iron storage, transcriptional regulation seemed to be a relevant mechanism. We show here that the minimum region of the ferritin H-gene promoter that is able to confer transcriptional regulation by heme in FLCs to a reporter gene is 77 nucleotides upstream of the TATA box. This cis element binds a protein complex referred to as HRF (heme-responsive factor), which is greatly enhanced both in heme-treated FLCs and during monocyte-to-macrophage differentiation. The CCAAT element present in reverse orientation in this promoter region of the ferritin H-chain gene is necessary for binding and for gene activity, since a single point mutation is able to abolish the binding of HRF and the transcriptional activity in transfected cells. By competition experiments and supershift assays, we identified the induced HRF as containing at least the ubiquitous transcription factor NF-Y. NF-Y is formed by three subunits, A, B, and C, all of which are necessary for DNA binding. Cotransfection with a transdominant negative mutant of the NF-YA subunit abolishes the transcriptional activation by heme, indicating that NF-Y plays an essential role in this activation. We have also observed a differential expression of the NF-YA subunit in heme-treated and control FLCs and during monocyte-to-macrophage differentiation.


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.


Journal of Immunology | 2008

IFN Regulatory Factor-1 Negatively Regulates CD4 + CD25 + Regulatory T Cell Differentiation by Repressing Foxp3 Expression

Alessandra Fragale; Lucia Gabriele; Emilia Stellacci; Paola Borghi; Edvige Perrotti; Ramona Ilari; Angela Lanciotti; Anna Lisa Remoli; Massimo Venditti; Filippo Belardelli; Angela Battistini

Regulatory T (Treg) cells are critical in inducing and maintaining tolerance. Despite progress in understanding the basis of immune tolerance, mechanisms and molecules involved in the generation of Treg cells remain poorly understood. IFN regulatory factor (IRF)-1 is a pleiotropic transcription factor implicated in the regulation of various immune processes. In this study, we report that IRF-1 negatively regulates CD4+CD25+ Treg cell development and function by specifically repressing Foxp3 expression. IRF-1-deficient (IRF-1−/−) mice showed a selective and marked increase of highly activated and differentiated CD4+CD25+Foxp3+ Treg cells in thymus and in all peripheral lymphoid organs. Furthermore, IRF-1−/− CD4+CD25− T cells showed extremely high bent to differentiate into CD4+CD25+Foxp3+ Treg cells, whereas restoring IRF-1 expression in IRF-1−/− CD4+CD25− T cells impaired their differentiation into CD25+Foxp3+ cells. Functionally, both isolated and TGF-β-induced CD4+CD25+ Treg cells from IRF-1−/− mice exhibited more increased suppressive activity than wild-type Treg cells. Such phenotype and functional characteristics were explained at a mechanistic level by the finding that IRF-1 binds a highly conserved IRF consensus element sequence (IRF-E) in the foxp3 gene promoter in vivo and negatively regulates its transcriptional activity. We conclude that IRF-1 is a key negative regulator of CD4+CD25+ Treg cells through direct repression of Foxp3 expression.


Biochemical Journal | 2006

Intracellular HIV-1 Tat protein represses constitutive LMP2 transcription increasing proteasome activity by interfering with the binding of IRF-1 to STAT1

Anna Lisa Remoli; Giulia Marsili; Edvige Perrotti; Eleonora Gallerani; Ramona Ilari; Filomena Nappi; Aurelio Cafaro; Barbara Ensoli; Riccardo Gavioli; Angela Battistini

The Tat protein is the transcriptional activator of HIV-1 gene expression, which is not only essential for viral replication, but also important in the complex HIV-induced pathogenesis of AIDS, as both an intracellular and an extracellular released protein. Accordingly, Tat is able to profoundly affect cellular gene expression, regulating several cellular functions, also in non-infected cells. We showed recently that Tat induces modification of immunoproteasomes in that it up-regulates LMP7 (low-molecular-mass polypeptide 7) and MECL1 (multicatalytic endopeptidase complex-like 1) subunits and down-modulates the LMP2 subunit, resulting in a change in the generation and presentation of epitopes in the context of MHC class I. In particular, Tat increases presentation of subdominant and cryptic epitopes. In the present study, we investigated the molecular mechanism responsible for the Tat-induced LMP2 down-regulation and show that intracellular Tat represses transcription of the LMP2 gene by competing with STAT1 (signal transducer and activator of transcription 1) for binding to IRF-1 (interferon-regulatory factor-1) on the overlapping ICS-2 (interferon consensus sequence-2)-GAS (gamma-interferon-activated sequence) present in the LMP2 promoter. This element is constitutively occupied in vivo by the unphosphorylated STAT1-IRF-1 complex, which is responsible for the basal transcription of the gene. Sequestration of IRF-1 by intracellular Tat impairs the formation of the complex resulting in lower LMP2 gene transcription and LMP2 protein expression, which is associated with increased proteolytic activity. On the other hand, extracellular Tat induces the expression of LMP2. These effects of Tat provide another effective mechanism by which HIV-1 affects antigen presentation in the context of the MHC class I complex and may have important implications in the use of Tat for vaccination strategies.


Oncogene | 2002

Role of Ets-1 in transcriptional regulation of transferrin receptor and erythroid differentiation.

Giovanna Marziali; Edvige Perrotti; Ramona Ilari; Valentina Lulli; Eliana M. Coccia; Rémy Moret; Lukas C. Kühn; Ugo Testa; Angela Battistini

High expression of transferrin receptor (TfR) on the membrane of erythroid cells accounts for the high level of iron required to sustain heme synthesis. Several studies indicate that during erythroid differentiation TfR expression is highly dependent on transcriptional regulation. In this study we characterized the minimal region able to confer transcriptional regulation during erythroid differentiation in Friend leukemia cells (FLC). This region of 120 bp, upstream the transcription start site, contains an overlapping consensus recognition sequence for AP1/CREB/ATF transcription factors and for proteins of the Ets family and a GC rich region. Here, we report that both the Ets and the Ap1/CRE like sites are essential for promoter activity during erythroid differentiation. We showed that Ets-1 binds to the EBS-TfR and its binding activity decreases in FLC induced to differentiate and during normal erythroid differentiation. Consistent with this, FLC constitutively expressing Ets-1 show a decrease in TfR gene expression, globin mRNA and hemoglobin synthesis. We conclude that Ets-1 binding activity is modulated during erythroid maturation and that a deregulated expression of this transcription factor interferes with terminal erythroid differentiation.


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.


Oncotarget | 2015

miR-135b suppresses tumorigenesis in glioblastoma stem-like cells impairing proliferation, migration and self-renewal

Valentina Lulli; Mariachiara Buccarelli; Maurizio Martini; Michele Signore; Mauro Biffoni; Stefano Giannetti; Liliana Morgante; Giovanna Marziali; Ramona Ilari; Alfredo Pagliuca; Luigi Maria Larocca; Ruggero De Maria; Roberto Pallini; Lucia Ricci-Vitiani

Glioblastoma multiforme (GBM) is the most common and fatal malignant adult primary brain tumor. Currently, the overall prognosis for GBM patients remains poor despite advances in neurosurgery and adjuvant treatments. MicroRNAs (miRNAs) contribute to the pathogenesis of various types of tumor, including GBM. In this study we analyzed the expression of a panel of miRNAs, which are known to be differentially expressed by the brain and GBM tumor, in a collection of patient-derived GBM stem-like cells (GSCs). Notably, the average expression level of miR-135b, was the most downregulated compared to its normal counterpart, suggesting a potential role as anti-oncogene. Restoration of miR-135b in GSCs significantly decreased proliferation, migration and clonogenic abilities. More importantly, miR-135b restoration was able to significantly reduce brain infiltration in mouse models of GBM obtained by intracerebral injection of GSC lines. We identified ADAM12 and confirmed SMAD5 and GSK3β as miR-135b targets and potential mediators of its effects. The whole transcriptome analysis ascertained that the expression of miR-135b downmodulated additional genes driving key pathways in GBM survival and infiltration capabilities. Our results identify a critical role of miR-135b in the regulation of GBM development, suggesting that miR-135b might act as a tumor-suppressor factor and thus providing a potential candidate for the treatment of GBM patients.


Molecular Oncology | 2017

A three-microRNA signature identifies two subtypes of glioblastoma patients with different clinical outcomes

Giovanna Marziali; Mariachiara Buccarelli; Ramona Ilari; Sveva Grande; Alessandra Palma; Quintino Giorgio D'Alessandris; Maurizio Martini; Mauro Biffoni; Roberto Pallini; Lucia Ricci-Vitiani

Glioblastoma multiforme (GBM) is the most common and malignant primary brain tumor in adults, characterized by aggressive growth, limited response to therapy, and inexorable recurrence. Because of the extremely unfavorable prognosis of GBM, it is important to develop more effective diagnostic and therapeutic strategies based on biologically and clinically relevant patient stratification systems. Analyzing a collection of patient‐derived GBM stem‐like cells (GSCs) by gene expression profiling, nuclear magnetic resonance spectroscopy, and signal transduction pathway activation, we identified two GSC clusters characterized by different clinical features. Due to the widely documented role played by microRNAs (miRNAs) in the tumorigenesis process, in this study we explored whether these two GBM patient subtypes could also be discriminated by different miRNA signatures. Global miRNA expression pattern was analyzed by oblique principal component analysis and principal component analysis. By a combined inferential strategy on PCA results, we identified a reduced set of three miRNAs – miR‐23a, miR‐27a, and miR‐9* (miR‐9‐3p) – able to discriminate the proneural‐ and mesenchymal‐like GSC phenotypes as well as mesenchymal and proneural subtypes of primary GBM included in The Cancer Genome Atlas (TCGA) data set. Kaplan–Meier analysis showed a significant correlation between the selected miRNAs and overall survival in 429 GBM specimens from TCGA‐identifying patients who had an unfavorable outcome. The survival prognostic capability of the three‐miRNA signatures could have important implications for the understanding of the biology of GBM subtypes and could be useful in patient stratification to facilitate interpretation of results from clinical trials.


Journal of Hematology & Oncology | 2016

PML-RAR alpha induces the downmodulation of HHEX: a key event responsible for the induction of an angiogenetic response

Ernestina Saulle; Alessia Petronelli; Elvira Pelosi; Elena Coppotelli; Luca Pasquini; Ramona Ilari; Francesco Lo-Coco; Ugo Testa

BackgroundRecent studies indicate that angiogenesis is important in the pathogenesis of acute myeloid leukemias (AMLs). Among the various AMLs, the bone marrow angiogenetic response is particularly pronounced in acute promyelocytic leukemia (APL). However, the molecular mechanisms responsible for this angiogenetic response are largely unknown. In the present study, we have explored the role of HHEX, a homeodomain transcription factor, as a possible mediator of the pro-angiogenetic response observed in APL. This transcription factor seems to represent an ideal candidate for this biologic function because it is targeted by PML-RARα, is capable of interaction with PML and PML-RARα, and acts as a regulator of the angiogenetic response.MethodsWe used various cellular systems of APL, including primary APL cells and leukemic cells engineered to express PML-RARα, to explore the role of the PML-RARα fusion protein on HHEX expression. Molecular and biochemical techniques have been used to investigate the mechanisms through which PML-RARα downmodulates HHEX and the functional consequences of this downmodulation at the level of the expression of various angiogenetic genes, cell proliferation and differentiation.ResultsOur results show that HHEX expression is clearly downmodulated in APL and that this effect is directly mediated by a repressive targeting of the HHEX gene promoter by PML-RARα. Studies carried out in primary APL cells and in a cell line model of APL with inducible PML-RARα expression directly support the view that this fusion protein through HHEX downmodulation stimulates the expression of various genes involved in angiogenesis and inhibits cell differentiation.ConclusionsOur data suggest that HHEX downmodulation by PML-RARα is a key event during APL pathogenesis.


Blood Cells Molecules and Diseases | 2015

SCF-mediated γ-globin gene expression in adult human erythroid cells is associated with KLF1, BCL11A and SOX6 down-regulation

Valentina Lulli; Paolo Romania; Ornella Morsilli; Ramona Ilari; Marco Gabbianelli; Ugo Testa; Giovanna Marziali

In humans, switching from fetal to adult hemoglobin occurs during the perinatal period and requires developmental stage-specific changes in chromatin remodeling complexes and transcription factors to progressively lead to β-globin gene activation and repression of γ-globin gene expression. Variable levels of HbF persist into adulthood. This variability depends on many genetic loci: three major loci – Xmn1HBG2 single nucleotide polymorphism, HBS1L-MYB intergenic region on chromosome 6q and a region of 14 kb in intron 2 of the BCL11A gene – have been identified as the main regulators of HbF level in humans [1]. The principle that elevated HbF ameliorates the severity of the β-hemoglobin disorders has been the driving force behind efforts to stimulate fetal hemoglobin production. Several pharmacologic agents (hypomethylating agents, histone deacetylase inhibitors, short-chain fatty acids) have been shown to reactivate and augment HbF expression. In addition to chemical inducers, HbF synthesis may be stimulated by hematopoietic growth factors, such as stem cell factor (SCF) [2]. SCF receptor and SCF play a central role in hematopoiesis and, particularly, in the control of erythropoiesis [2–4]. Although much is known regarding trans-acting regulators and cis-acting elements within the core promoter and distal enhancer of γ-globin, the transcription factors through which SCF stimulates HbF synthesis in adult erythroid cells are largely unknown. To analyze the transcription factors through which SCF may stimulate HbF synthesis in human erythroid cells we employed erythroid unilineage cultures (E), derived from peripheral blood (PB) CD34hematopoietic progenitor cells (HPCs) obtained from6 independent volunteers, using experimental conditions previously reported [2]. In these serum-free liquid suspension cultures, purified HPCs are induced to selective E growth by very low dosages of IL-3, GM-CSF and saturating erythropoietin (Epo) level. These cultureswere supplemented or not with 100 ng/ml SCF. Cell growth is associated with a gradual decrease of CD34 marker, followed by a progressively increasing expression of specific erythroid markers, such as glycophorin A (data not shown). Consistently, cell morphological analysis showed a gradual wave of maturation along the erythroid lineage up to terminal mature erythroid cells (Fig. 1A). In line with previously published results [3,4], SCF sharply stimulated erythroid cell proliferation, as indicated by a 2to 3-log rise of cell output over control values, while delaying by about one week terminal differentiation/maturation (Fig. 1A). PB E cultures, grown in the absence of exogenous SCF, showed low γ-globin mRNA levels; low γ-globin-protein levels were observed both by Western Blot analysis and HPLC assay (Fig. 1B). Particularly, in these cultures, γ / β + γ globin ratio is consistently less than 3%. In PB E cultures supplemented with SCF, a remarkable increase of HbF synthesis was observed, as consistently supported by mRNA and γ-globin protein evaluations (Fig. 1B). In these cultures, the γ / β + γ globin ratio is

Collaboration


Dive into the Ramona Ilari's collaboration.

Top Co-Authors

Avatar

Edvige Perrotti

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Angela Battistini

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

Emilia Stellacci

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

Eliana M. Coccia

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Alessandra Fragale

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Angela Lanciotti

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Roberto Orsatti

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Valentina Lulli

Istituto Superiore di Sanità

View shared research outputs
Researchain Logo
Decentralizing Knowledge