Network


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

Hotspot


Dive into the research topics where Paola Sestili is active.

Publication


Featured researches published by Paola Sestili.


Journal of Experimental Medicine | 2002

ICSBP is essential for the development of mouse type I interferon-producing cells and for the generation and activation of CD8α+ dendritic cells

Giovanna Schiavoni; Fabrizio Mattei; Paola Sestili; Paola Borghi; Massimo Venditti; Herbert C. Morse; Filippo Belardelli; Lucia Gabriele

Interferon (IFN) consensus sequence-binding protein (ICSBP) is a transcription factor playing a critical role in the regulation of lineage commitment, especially in myeloid cell differentiation. In this study, we have characterized the phenotype and activation pattern of subsets of dendritic cells (DCs) in ICSBP−/− mice. Remarkably, the recently identified mouse IFN-producing cells (mIPCs) were absent in all lymphoid organs from ICSBP−/− mice, as revealed by lack of CD11clowB220+Ly6C+CD11b− cells. In parallel, CD11c+ cells isolated from ICSBP−/− spleens were unable to produce type I IFNs in response to viral stimulation. ICSBP−/− mice also displayed a marked reduction of the DC subset expressing the CD8α marker (CD8α+ DCs) in spleen, lymph nodes, and thymus. Moreover, ICSBP−/− CD8α+ DCs exhibited a markedly impaired phenotype when compared with WT DCs. They expressed very low levels of costimulatory molecules (intercellular adhesion molecule [ICAM]-1, CD40, CD80, CD86) and of the T cell area-homing chemokine receptor CCR7, whereas they showed higher levels of CCR2 and CCR6, as revealed by reverse transcription PCR. In addition, these cells were unable to undergo full phenotypic activation upon in vitro culture in presence of maturation stimuli such as lipopolysaccharide or poly (I:C), which paralleled with lack of Toll-like receptor (TLR)3 mRNA expression. Finally, cytokine expression pattern was also altered in ICSBP−/− DCs, as they did not express interleukin (IL)-12p40 or IL-15, but they displayed detectable IL-4 mRNA levels. On the whole, these results indicate that ICSBP is a crucial factor in the regulation of two possibly linked processes: (a) the development and activity of mIPCs, whose lack in ICSBP−/− mice may explain their high susceptibility to virus infections; (b) the generation and activation of CD8α+ DCs, whose impairment in ICSBP−/− mice can be responsible for the defective generation of a Th1 type of immune response.


Journal of Immunology | 2002

Type I IFN as a Natural Adjuvant for a Protective Immune Response: Lessons from the Influenza Vaccine Model

Enrico Proietti; Laura Bracci; Simona Puzelli; Tiziana Di Pucchio; Paola Sestili; Enrico De Vincenzi; Massimo Venditti; Imerio Capone; Isabelle Seif; Edward De Maeyer; David F. Tough; Isabella Donatelli; Filippo Belardelli

The identification of natural adjuvants capable of selectively promoting an efficient immune response against infectious agents would represent an important advance in immunology, with direct implications for vaccine development, whose progress is generally hampered by the difficulties in defining powerful synthetic adjuvants suitable for clinical use. Here, we demonstrate that endogenous type I IFN is necessary for the Th1 type of immune response induced by typical adjuvants in mice and that IFN itself is an unexpectedly powerful adjuvant when administered with the human influenza vaccine, for inducing IgG2a and IgA production and conferring protection from virus challenge. The finding that these cytokines, currently used in patients, are necessary for full expression of adjuvant activity and are sufficient for the generation of a protective immune response opens new perspectives in understanding the basis of immunity and in vaccine development.


Clinical Cancer Research | 2007

Cyclophosphamide Enhances the Antitumor Efficacy of Adoptively Transferred Immune Cells through the Induction of Cytokine Expression, B-Cell and T-Cell Homeostatic Proliferation, and Specific Tumor Infiltration

Laura Bracci; Federica Moschella; Paola Sestili; Valentina La Sorsa; Mara Valentini; Irene Canini; Sara Baccarini; Sonia Maccari; Carlo Ramoni; Filippo Belardelli; Enrico Proietti

Purpose: Immunotherapy is a promising antitumor strategy, which can be successfully combined with current anticancer treatments, as suggested by recent studies showing the paradoxical chemotherapy-induced enhancement of the immune response. The purpose of the present work is to dissect the biological events induced by chemotherapy that cooperate with immunotherapy in the success of the combined treatment against cancer. In particular, we focused on the following: (a) cyclophosphamide-induced modulation of several cytokines, (b) homeostatic proliferation of adoptively transferred lymphocytes, and (c) homing of transferred lymphocytes to secondary lymphoid organs and tumor mass. Experimental Design: Here, we used the adoptive transfer of tumor-immune cells after cyclophosphamide treatment of tumor-bearing mice as a model to elucidate the mechanisms by which cyclophosphamide can render the immune lymphocytes competent to induce tumor rejection. Results: The transfer of antitumor immunity was found to be dependent on CD4+ T cells and on the cooperation of adoptively transferred cells with the host immune system. Of note, tumor-immune lymphocytes migrated specifically to the tumor only in mice pretreated with cyclophosphamide. Cyclophosphamide treatment also promoted homeostatic proliferation/activation of transferred B and T lymphocytes. Optimal therapeutic responses to the transfer of immune cells were associated with the cyclophosphamide-mediated induction of a “cytokine storm” [including granulocyte macrophage colony-stimulating factor, interleukin (IL)-1β, IL-7, IL-15, IL-2, IL-21, and IFN-γ], occurring during the “rebound phase” after drug-induced lymphodepletion. Conclusions: The ensemble of these data provides a new rationale for combining immunotherapy and chemotherapy to induce an effective antitumor response in cancer patients.


Gene Therapy | 2000

Type I consensus interferon (CIFN) gene transfer into human melanoma cells up-regulates p53 and enhances cisplatin-induced apoptosis: implications for new therapeutic strategies with IFN-alpha

Monica Mecchia; Paola Matarrese; Walter Malorni; Giuseppina D'Agostino; Paola Sestili; Stefano M. Santini; M C Gauzzi; Massimo Venditti; A Mazzocchi; G Parmiani; Filippo Belardelli; Maria Ferrantini

In this study, we describe the effects produced by the retroviral transduction of human type I consensus IFN (CIFN) coding sequence into the 8863 and 1B6 human melanoma cell lines, derived from a metastatic and a primary human melanoma, respectively. Melanoma cell lines producing approximately 103 IU/ml of IFN were obtained. Interestingly, cisplatin treatment of IFN-producing 8863 and 1B6 melanoma cells resulted in a three- to four-fold increase in the percentage of apoptotic cells with respect to similarly treated parental or control-transduced cell cultures. A similar effect, although less intense, was caused by cultivation of parental melanoma cells in the presence of exogenous CIFN. The increased susceptibility of the IFN-producing melanoma cell lines to cisplatin-induced apoptosis was associated with an IFN-dependent accumulation of p53, which also correlated with a decrease in Bcl-2 expression. Addition of exogenous CIFN to parental melanoma cells resulted in similar although weaker modulations of p53 and Bcl-2 expression. Cisplatin administration to nude mice bearing 3-day-old IFN-producing 8863 tumors resulted in complete tumor regression, while only a partial tumor inhibition was observed upon cisplatin treatment of mice bearing parental or control-transduced 8863 tumors. Starting the cisplatin treatment 7 days after tumor cell injection still resulted in a stronger inhibition of tumor growth in the mice bearing IFN-producing 8863 tumors as compared with parental tumor-bearing mice. A comparable therapeutic effect was obtained after repeated peritumoral administration of 103 IU of exogenous CIFN and cisplatin treatment. Interestingly, a spontaneous tumor regression was observed in nude mice injected with IFN-producing 1B6 cells, in contrast to the progressive tumor growth occurring in mice receiving a similar inoculum of the parental or control-transduced 1B6 melanoma cells. Repeated peritumoral administration of 103 IU of exogenous CIFN to mice bearing parental 1B6 tumors caused only a transient inhibition of tumor growth. These results indicate that type I IFN gene transfer is an effective approach for suppressing the tumorigenic phenotype of human melanoma cells and for increasing the efficacy of anticancer drugs. These observations, together with our previous findings showing the importance of IFN-α–T cell interactions in the generation of an antitumor response in mouse models, underline the interest of using type I IFN in gene therapy strategies for the treatment of human melanoma.


Journal of Leukocyte Biology | 2006

IRF-1 deficiency skews the differentiation of dendritic cells toward plasmacytoid and tolerogenic features

Lucia Gabriele; Alessandra Fragale; Paola Borghi; Paola Sestili; Emilia Stellacci; Massimo Venditti; Giovanna Schiavoni; Massimo Sanchez; Filippo Belardelli; Angela Battistini

Members of the IFN regulatory factors (IRFs) family are transcriptional regulators that play essential roles in the homeostasis and function of the immune system. Recent studies indicate a direct involvement of some members of the family in the development of different subsets of dendritic cells (DC). Here, we report that IRF‐1 is a potent modulator of the development and functional maturation of DC. IRF‐1‐deficient mice (IRF‐1−/−) exhibited a predominance of plasmacytoid DC and a selective reduction of conventional DC, especially the CD8α+ subset. IRF‐1−/− splenic DC were markedly impaired in their ability to produce proinflammatory cytokines such as IL‐12. By contrast, they expressed high levels of IL‐10, TGF‐β, and the tolerogenic enzyme indoleamine 2,3 dioxygenase. As a consequence, IRF‐1−/− DC were unable to undergo full maturation and retained plasmacytoid and tolerogenic characteristics following virus infection ex vivo and in vivo. Accordingly, DC from IRF‐1−/− mice were less efficient in stimulating the proliferation of allogeneic T cells and instead, induced an IL‐10‐mediated, suppressive activity in allogeneic CD4+CD25+ regulatory T cells. Together, these results indicate that IRF‐1 is a key regulator of DC differentiation and maturation, exerting a variety of effects on the functional activation and tolerogenic potential of these cells.


AIDS | 2001

Vaginal transmission of HIV-1 in hu-SCID mice: a new model for the evaluation of vaginal microbicides.

Simonetta Di Fabio; Giacomo Giannini; Caterina Lapenta; Massimo Spada; Andrea Binelli; Elena Germinario; Paola Sestili; Filippo Belardelli; Enrico Proietti; Stefano Vella

ObjectiveTo develop an animal model of vaginal transmission of HIV-1 for the evaluation of vaginal microbicides. DesignVaginal infection was performed in SCID mice reconstituted with 4 × 107 human peripheral blood lymphocytes (hu-PBL) by non-invasive vaginal administration. The hu-PBL were previously infected in vitro with a non-syncytium (NSI) strain of HIV-1 (SF162) (hu-PBL-SCID). Lymphocyte migration in vivo was examined using fluorescently labelled human lymphocytes. MethodsThe percentage of CD4 T cells, plasma viral load and p24 antigen were evaluated using fluorescent activated cell sorting (FACS), the Amplicor HIV-1 monitor kit and enzyme-linked immunosorbent assay, respectively. Polymerase chain reaction (PCR) analysis was performed on DNA extracted from spleen and lymph nodes. For in vivo migration of labelled lymphocytes, the mice were sacrified after 4, 24 and 48 h; vaginae and local lymph nodes were removed, snap frozen with OCT, sectioned and examined by fluorescent microscopy and FACS. ResultsHIV transmission was established using virus-infected cells inoculated vaginally, as shown by FACS, HIV viral load, p24 and PCR results. Labelled cells were easily located within the vaginal tissues after 4 h. However, few or no cells could be identified after 24 or 48 h at the vaginal level, whereas labelled cells could be detected at the level of regional lymph nodes. ConclusionsBecause of its simplicity and practical features compared with other animal models, the vaginal HIV-infected hu-SCID mouse model may prove useful to test the activity of compounds against cell-associated HIV and, possibly, other sexually transmitted diseases.


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.


Journal of Immunotoxicology | 2014

A multidisciplinary study using in vivo tumor models and microfluidic cell-on-chip approach to explore the cross-talk between cancer and immune cells

Fabrizio Mattei; Giovanna Schiavoni; Adele De Ninno; Valeria Lucarini; Paola Sestili; Antonella Sistigu; Alessandra Fragale; Massimo Sanchez; Massimo Spada; Annamaria Gerardino; Filippo Belardelli; Luca Businaro; Lucia Gabriele

Abstract A full elucidation of events occurring inside the cancer microenvironment is fundamental for the optimization of more effective therapies. In the present study, the cross-talk between cancer and immune cells was examined by employing mice deficient (KO) in interferon regulatory factor (IRF)-8, a transcription factor essential for induction of competent immune responses. The in vivo results showed that IRF-8 KO mice were highly permissive to B16.F10 melanoma growth and metastasis due to failure of their immune cells to exert proper immunosurveillance. These events were found to be dependent on soluble factors released by cells of the immune system capable of shaping the malignant phenotype of melanoma cells. An on-chip model was then generated to further explore the reciprocal interactions between the B16.F10 and immune cells. B16.F10 and immune cells were co-cultured in a microfluidic device composed of three culturing chambers suitably inter-connected by an array of microchannels; mutual interactions were then followed using time-lapse microscopy. It was observed that WT immune cells migrated through the microchannels towards the B16.F10 cells, establishing tight interactions that in turn limited tumor spread. In contrast, IRF-8 KO immune cells poorly interacted with the melanoma cells, resulting in a more invasive behavior of the B16.F10 cells. These results suggest that IRF-8 expression plays a key role in the cross-talk between melanoma and immune cells, and under-score the value of cell-on-chip approaches as useful in vitro tools to reconstruct complex in vivo microenvironments on a microscale level to explore cell interactions such as those occurring within a cancer immunoenvironment.


Transplantation | 1995

The SCID mouse reaction to human peripheral blood mononuclear leukocyte engraftment. Neutrophil recruitment induced expression of a wide spectrum of murine cytokines and mouse leukopoiesis, including thymic differentiation.

Stefano M. Santini; Paola Rizza; Maria Antonia Logozzi; Paola Sestili; Giovanni Gherardi; Roberto Lande; Caterina Lapenta; Filippo Belardelli; Stefano Fais

In this study, we describe the kinetics of host immune reactions occurring in mice with severe combined immunodeficiency (SCID) at different times after the intraperitoneal injection of human peripheral blood mononuclear leukocytes (huPBL). At 24 hr, a massive neutrophil recruitment and an induced expression of a wide spectrum of murine cytokine mRNA (i.e., interleukin [IL]-10, IL-4, IL-6, IL-10, IL-12, tumor necrosis factor [TNF]-α and interferon [IFN]-γ) occurred in the huPBL-SCID mouse peritoneal cavity. By using ELISAs specific for mouse cytokines, large amounts of IL-1-α, TNF-α, IL-6, and IFN-γ were detected in the peritoneal washings of huPBL-SCID mice 1 day after intraperitoneal injection. IL-6 and IFN-γ production persisted for up to 2 weeks after PBL transplantation. Medullary and extramedullary expansion of the SCID mouse hematopoietic cells also occurred in the chimeras as early as 1 week after injection, together with a marked thymic differentiation (murine CD4+/CD8+ cells) at 10–12 weeks after transplantation. On the whole, these results indicate that, after huPBL injection, SCID mice mount a complex multistage immune response. These host reactions should be taken into consideration for any accurate interpretation of results obtained using the huPBL-SCD3 model. The control of responses (by means of specific antibodies to murine cytokines and to granulocytes or through the use of anti-inflammatory drugs) may be helpful in improving the engraftment of huPBL in SCID mice and in furthering our knowledge of the T and B cell-independent natural immune reactions.


Journal of Interferon and Cytokine Research | 1999

Type I consensus IFN (IFN-con1) Gene Transfer into KSHV/HHV-8-Infected BCBL-1 Cells Causes Inhibition of Viral Lytic Cycle Activation via Induction of Apoptosis and Abrogates Tumorigenicity in SCID Mice

Giuseppina D'Agostino; Eleonora Aricò; Laura Santodonato; Massimo Venditti; Paola Sestili; Laura Masuelli; Anna Coletti; Andrea Modesti; Gaston Picchio; Donald E. Mosier; Maria Ferrantini; Filippo Belardelli

In this study, we investigated the effects of human type I consensus interferon (IFN-con1) (Amgen) gene transfer into body cavity-based lymphomas (BCBL)-1 cells, which are latently infected with Kaposis sarcoma-associated herpesvirus (KSHV) human herpesvirus-8 (HHV-8). Both the basal and 12-O-tetradecanoyl phorbol-13-acetate (TPA)-stimulated production of KSHV/HHV-8 mature virions was strongly inhibited in genetically modified IFN-producing BCBL-1 cells as compared with parental or control transduced counterparts. A similar inhibition was obtained on treatment of parental BCBL-1 cells with exogenous IFN-con1. The reduction in KSHV/HHV-8 production was associated with a decrease in the basal and TPA-stimulated intracellular amount of the linear form of the viral genome. Interestingly, 25%40% of the IFN-producing BCBL-1 cell population underwent spontaneous apoptosis in vitro. TPA treatment, which did not significantly affect the viability of the parental and control BCBL-1 cells, resulted in the apoptotic death of up to 70% of the IFN-producing cell population. Addition of exogenous IFN-con1 to parental BCBL-1 cells produced similar effects, although less intense. Injection of either parental or control-transduced BCBL-1 cells into SCID mice resulted in progressively growing tumors characterized by an unusually high level of tumor angiogenesis. In contrast, complete tumor regression was observed in all the mice injected either subcutaneously (s.c.) or intraperitoneally (i.p.) with the IFN-producing BCBL-1 cells. These results represent the first evidence that type I IFN can counteract the activation of a productive herpesvirus infection in latently infected tumor cells by the induction of apoptosis, providing an interesting link between the antiviral and antitumor activities of this cytokine. These data suggest the possible advantages of strategies of type I IFN gene transfer (with respect to the use of the exogenous cytokine) for the treatment of patients with some HHV-8-induced malignancies.

Collaboration


Dive into the Paola Sestili's collaboration.

Top Co-Authors

Avatar

Filippo Belardelli

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Enrico Proietti

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Giovanna Schiavoni

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Massimo Venditti

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Fabrizio Mattei

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Lucia Gabriele

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Laura Bracci

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Massimo Spada

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Paola Borghi

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Antonella Sistigu

Catholic University of the Sacred Heart

View shared research outputs
Researchain Logo
Decentralizing Knowledge