Network


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

Hotspot


Dive into the research topics where Luca Battistini is active.

Publication


Featured researches published by Luca Battistini.


Nature Neuroscience | 2011

Caspase-3 triggers early synaptic dysfunction in a mouse model of Alzheimer's disease

Marcello D'Amelio; Virve Cavallucci; Silvia Middei; Cristina Marchetti; Simone Pacioni; Alberto Ferri; Adamo Diamantini; Daniela De Zio; Paolo Carrara; Luca Battistini; Sandra Moreno; Alberto Bacci; Martine Ammassari-Teule; Hélène Marie; Francesco Cecconi

Synaptic loss is the best pathological correlate of the cognitive decline in Alzheimers disease; however, the molecular mechanisms underlying synaptic failure are unknown. We found a non-apoptotic baseline caspase-3 activity in hippocampal dendritic spines and an enhancement of this activity at the onset of memory decline in the Tg2576-APPswe mouse model of Alzheimers disease. In spines, caspase-3 activated calcineurin, which in turn triggered dephosphorylation and removal of the GluR1 subunit of AMPA-type receptor from postsynaptic sites. These molecular modifications led to alterations of glutamatergic synaptic transmission and plasticity and correlated with spine degeneration and a deficit in hippocampal-dependent memory. Notably, pharmacological inhibition of caspase-3 activity in Tg2576 mice rescued the observed Alzheimer-like phenotypes. Our results identify a previously unknown caspase-3–dependent mechanism that drives synaptic failure and contributes to cognitive dysfunction in Alzheimers disease. These findings indicate that caspase-3 is a potential target for pharmacological therapy during early disease stages.


Journal of Experimental Medicine | 2005

Boosting antitumor responses of T lymphocytes infiltrating human prostate cancers

Vincenzo Bronte; Tihana Kasic; Giorgia Gri; Keti Gallana; Giovanna Borsellino; Ilaria Marigo; Luca Battistini; Massimo Iafrate; Tommaso Prayer-Galetti; Francesco Pagano; Antonella Viola

Immunotherapy may provide valid alternative therapy for patients with hormone-refractory metastatic prostate cancer. However, if the tumor environment exerts a suppressive action on antigen-specific tumor-infiltrating lymphocytes (TIL), immunotherapy will achieve little, if any, success. In this study, we analyzed the modulation of TIL responses by the tumor environment using collagen gel matrix–supported organ cultures of human prostate carcinomas. Our results indicate that human prostatic adenocarcinomas are infiltrated by terminally differentiated cytotoxic T lymphocytes that are, however, in an unresponsive status. We demonstrate the presence of high levels of nitrotyrosines in prostatic TIL, suggesting a local production of peroxynitrites. By inhibiting the activity of arginase and nitric oxide synthase, key enzymes of L-arginine metabolism that are highly expressed in malignant but not in normal prostates, reduced tyrosine nitration and restoration of TIL responsiveness to tumor were achieved. The metabolic control exerted by the tumor on TIL function was confirmed in a transgenic mouse prostate model, which exhibits similarities with human prostate cancer. These results identify a novel and dominant mechanism by which cancers induce immunosuppression in situ and suggest novel strategies for tumor immunotherapy.


Circulation Research | 2005

Exogenous High-Mobility Group Box 1 Protein Induces Myocardial Regeneration After Infarction via Enhanced Cardiac C-Kit+ Cell Proliferation and Differentiation

Federica Limana; Antonia Germani; Antonella Zacheo; Jan Kajstura; Anna Di Carlo; Giovanna Borsellino; Omar Leoni; Roberta Palumbo; Luca Battistini; Raffaella Rastaldo; Susanne Müller; Giulio Pompilio; Piero Anversa; Marco Bianchi; Maurizio C. Capogrossi

High-mobility group box 1 protein (HMGB1) is a chromatin protein that is released by inflammatory and necrotic cells. Extracellular HMGB1 signals tissue damage, stimulates the secretion of proinflammatory cytokines and chemokines, and modulates stem cell function. The present study examined exogenous HMGB1 effect on mouse left-ventricular function and myocyte regeneration after infarction. Myocardial infarction was induced in C57BL/6 mice by permanent coronary artery ligation. After 4 hours animals were reoperated and 200 ng of purified HMGB1 was administered in the peri-infarcted left ventricle. This intervention resulted in the formation of new myocytes within the infarcted portion of the wall. The regenerative process involved the proliferation and differentiation of endogenous cardiac c-kit+ progenitor cells. Circulating c-kit+ cells did not significantly contribute to HMGB1-mediated cardiac regeneration. Echocardiographic and hemodynamic parameters at 1, 2, and 4 weeks demonstrated a significant recovery of cardiac performance in HMGB1-treated mice. These effects were not observed in infarcted hearts treated either with the unrelated protein glutathione S-transferase or a truncated form of HMGB1. Thus, HMGB1 appears to be a potent inducer of myocardial regeneration following myocardial infarction.


The Journal of Neuroscience | 2009

Inflammation Triggers Synaptic Alteration and Degeneration in Experimental Autoimmune Encephalomyelitis

Diego Centonze; Luca Muzio; Silvia Rossi; Francesca Cavasinni; Valentina De Chiara; Alessandra Bergami; Alessandra Musella; Marcello D'Amelio; Virve Cavallucci; Alessandro Martorana; Andrea Bergamaschi; Maria Teresa Cencioni; Adamo Diamantini; Erica Butti; Giancarlo Comi; Giorgio Bernardi; Francesco Cecconi; Luca Battistini; Roberto Furlan; Gianvito Martino

Neurodegeneration is the irremediable pathological event occurring during chronic inflammatory diseases of the CNS. Here we show that, in experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis, inflammation is capable in enhancing glutamate transmission in the striatum and in promoting synaptic degeneration and dendritic spine loss. These alterations occur early in the disease course, are independent of demyelination, and are strongly associated with massive release of tumor necrosis factor-α from activated microglia. CNS invasion by myelin-specific blood-borne immune cells is the triggering event, and the downregulation of the early gene Arc/Arg3.1, leading to the abnormal expression and phosphorylation of AMPA receptors, represents a culminating step in this cascade of neurodegenerative events. Accordingly, EAE-induced synaptopathy subsided during pharmacological blockade of AMPA receptors. Our data establish a link between neuroinflammation and synaptic degeneration and calls for early neuroprotective therapies in chronic inflammatory diseases of the CNS.


The FASEB Journal | 2007

Local expression of IGF-1 accelerates muscle regeneration by rapidly modulating inflammatory cytokines and chemokines

Laura Pelosi; Cristina Giacinti; Chiara Nardis; Giovanna Borsellino; Emanuele Rizzuto; Carmine Nicoletti; Francesca Wannenes; Luca Battistini; Nadia Rosenthal; Mario Molinaro; Antonio Musarò

Muscle regeneration following injury is characterized by myonecrosis accompanied by local inflammation, activation of satellite cells, and repair of injured fibers. The resolution of the inflammatory response is necessary to proceed toward muscle repair, since persistence of inflammation often renders the damaged muscle incapable of sustaining efficient muscle regeneration. Here, we show that local expression of a muscle‐restricted insulin‐like growth factor (IGF)‐1 (mIGF‐1) transgene accelerates the regenerative process of injured skeletal muscle, modulating the inflammatory response, and limiting fibrosis. At the molecular level, mIGF‐1 expression significantly down‐regulated proinflammatory cytokines, such as tumor necrosis factor (TNF)‐alpha and interleukin (IL)‐1beta, and modulated the expression of CC chemokines involved in the recruitment of monocytes/macrophages. Analysis of the underlying molecular mechanisms revealed that mIGF‐1 expression modulated key players of inflammatory response, such as macrophage migration inhibitory factor (MIF), high mobility group protein‐1 (HMGB1), and transcription NF‐KB. The rapid restoration of injured mIGF‐1 transgenic muscle was also associated with connective tissue remodeling and a rapid recovery of functional properties. By modulating the inflammatory response and reducing fibrosis, supplemental mIGF‐1 creates a qualitatively different environment for sustaining more efficient muscle regeneration and repair.—Pelosi, L., Giacinti, C., Nardis, C., Borsellino, G., Rizzuto, E., Nicoletti, C., Wannenes, F., Battistini, L., Rosenthal, N., Molinaro, M., Musaro, A. Local expression of IGF‐1 accelerates muscle regeneration by rapidly modulating inflammatory cytokines and chemokines. FASEB J. 21, 1393–1402 (2007)


Circulation Research | 2007

Identification of Myocardial and Vascular Precursor Cells in Human and Mouse Epicardium

Federica Limana; Antonella Zacheo; David Mocini; Antonella Mangoni; Giovanna Borsellino; Adamo Diamantini; Roberta De Mori; Luca Battistini; Elisa Vigna; Massimo Santini; Vincenzo Loiaconi; Giulio Pompilio; Antonia Germani; Maurizio C. Capogrossi

During cardiac development, the epicardium is the source of multipotent mesenchymal cells, which give rise to endothelial and smooth muscle cells in coronary vessels and also, possibly, to cardiomyocytes. The aim of the present study was to determine whether stem cells are retained in the adult human and murine epicardium and to investigate the regenerative potential of these cells following acute myocardial infarction. We show that c-kit+ and CD34+ cells can indeed be detected in human fetal and adult epicardium and that they represent 2 distinct populations. Both subsets of cells were negative for CD45, a cell surface marker that identifies the hematopoietic cell lineage. Immunofluorescence revealed that freshly isolated c-kit+ and CD34+ cells expressed early and late cardiac transcription factors and could acquire an endothelial phenotype in vitro. In the murine model of myocardial infarction, there was an increase in the absolute number and proliferation of epicardial c-kit+ cells 3 days after coronary ligation; at this time point, epicardial c-kit+ cells were identified in the subepicardial space and expressed GATA4. Furthermore, 1 week after myocardial infarction, cells coexpressing c-kit+, together with endothelial or smooth muscle cell markers, were identified in the wall of subepicardial blood vessels. In summary, the postnatal epicardium contains a cell population with stem cell characteristics that retains the ability to give rise to myocardial precursors and vascular cells. These cells may play a role in the regenerative response to cardiac damage.


Developmental Neuroscience | 1994

Reactive Nitrogen Intermediates in Human Neuropathology: An Overview

Celia F. Brosnan; Luca Battistini; Cedric S. Raine; Dennis W. Dickson; Arturo Casadevall; Sunhee C. Lee

Nitric oxide (NO) is a recently recognized messenger molecule that has been shown to possess pleiotropic properties, including vasodilation, neurotransmission, cytotoxicity and antimicrobial activity. Constitutive and inducible forms of NO synthase (NOS) have been identified. Activation of cNOS releases relatively low levels of NO for short periods of time whereas induction of iNOS releases high levels of NO for extended periods of time. In rodents, iNOS is predominantly found in cells of the monocyte/macrophage series, including microglia, where it is induced by a combination of bacterial products and cytokines. cNOS and iNOS have also been reported in rodent astrocytes. Activation of iNOS in the CNS could be toxic to many different cell types, including neurons and oligodendrocytes. iNOS, however, has been difficult to demonstrate in human peripheral blood cells, suggesting that the regulation of expression of this enzyme in humans is different from that found in rodents. In this overview, we show that in human glial cells cultured in vitro, astrocytes, but not microglia, can be induced by cytokines to express NO-like activity. Bacterial products are without effect, but a combination of IL-1 and TNF alpha or IFN gamma is a potent stimulus. NO production by astrocytes inhibits Cryptococcus neoformans growth in vitro. In vivo, we show in acute multiple sclerosis lesions, intense NADPH-diaphorase activity is present in hypertrophic astrocytes in the lesion center and at the lesion edge, whereas microglia are nonreactive. Increased NADPH-diaphorase activity colocalizes with immunoreactivity for IL-1 and TNF. These results suggests that the induction of reactive nitrogen intermediates in humans differs from that found in rodents, and supports the conclusion that hypertrophic astrocytes are the major source of NO-like activity in the inflamed CNS.


Journal of Neuroimmunology | 2010

Altered miRNA expression in T regulatory cells in course of multiple sclerosis.

Giuseppe De Santis; Manuela Ferracin; Andrea Biondani; Luisa Caniatti; Maria Rosaria Tola; Massimiliano Castellazzi; Barbara Zagatti; Luca Battistini; Giovanna Borsellino; Enrico Fainardi; Riccardo Gavioli; Massimo Negrini; Roberto Furlan; Enrico Granieri

OBJECTIVES Multiple sclerosis (MS) is a chronic inflammatory response against constituents of the central nervous system. It is known that regulatory T cells (Tregs) play a key role in the autoimmune balance and their improper function may facilitate the expansion of autoaggressive T cell clones. Recently, microRNAs (miRNAs) have been involved in autoimmune disorders and their loss-of-function in immune cells was shown to facilitate systemic autoimmune disorders. Here, we analyzed the miRNA expression profile in Tregs from MS-RR. METHODS We assessed miRNA genome-wide expression profile by microarray analysis on CD4(+)CD25(+high) T cells from 12 MS relapsing-remitting patients in stable condition and 14 healthy controls. Since CD4(+)CD25(+high) T cells comprise both T regulatory cells (CD4(+)CD25(+high)CD127(dim/-)) and T effector cells (CD4(+)CD25(+high)CD127(+)), we performed a quantitative RT-PCR on CD4(+)CD25(+high)CD127(dim/-) and CD4(+)CD25(+high)CD127(+) cells isolated from the same blood sample. RESULTS We found 23 human miRNAs differentially expressed between CD4(+)CD25(high)bona fide Treg cells from MS patients vs. healthy donors, but, conversely, among the deregulated miRNAs, members of the miR-106b-25 were found down-regulated in MS patients when compared to healthy donors in CD4(+)CD25(high)CD127(dim/-) T regulatory cells. More interesting, the ratio between Treg/Teff showed an enrichment of these microRNA in T regulatory cells derived from patients if compared to healthy controls. CONCLUSION miR-106b and miR-25 were previously shown to modulate the TGF-β signaling pathway through their action on CDKN1A/p21 and BCL2L11/Bim. TGF-β is involved in T regulatory cells differentiation and maturation. Therefore, the deregulation of this miRNA cluster may alter Treg cells activity in course of MS, by altering TGF-β biological functions.


Journal of Leukocyte Biology | 2003

Phenotypic and functional analysis of T cells homing into the CSF of subjects with inflammatory diseases of the CNS.

Debora Giunti; Giovanna Borsellino; Roberto Benelli; Monica Marchese; Elisabetta Capello; M. T. Valle; Enrico Pedemonte; Douglas M. Noonan; Adriana Albini; Giorgio Bernardi; Giovanni Luigi Mancardi; Luca Battistini; Antonio Uccelli

The recruitment of lymphocytes across the blood brain barrier (BBB) is mediated by adhesion molecules and chemokines. The expression of activation markers and of chemokine receptors on T cells homing to the nervous system (NS) may help define their functional state. In the cerebrospinal fluid (CSF) of subjects with inflammatory neurological diseases (IND), including multiple sclerosis, we observed an increased number of T cells coexpressing CXCR3 and CCR5 as well as T cells with a CD45RO+ CCR7+ CD27+ memory phenotype. A subset of CCR7+ T cells coexpressed CXCR3 and CCR5. We also detected an increased number of interferon‐γ‐producing T cells in the CSF compared with peripheral blood, mostly but not exclusively in the CD45RO+ CCR7− CD27− compartment. T helper 1 (Th1) clones, established from the CSF of individuals with IND and from a healthy subject, similarly migrated to CXCL10, CXCL12, and CCL5. CXCL10, CXCL12, and CCL19 were increased in the CSF of individuals with neuroinflammation. These findings suggest that CSF is enriched in Th1‐polarized memory T cells capable of differentiating into effector cells upon antigen encounter. These cells are recruited into the CSF by inducible chemokines. Thus, CSF represents a transitional station for T cells trafficking to and from the NS.


PLOS ONE | 2010

Anandamide Suppresses Proliferation and Cytokine Release from Primary Human T-Lymphocytes Mainly via CB2 Receptors

Maria Teresa Cencioni; Valerio Chiurchiù; Giuseppina Catanzaro; Giovanna Borsellino; Giorgio Bernardi; Luca Battistini; Mauro Maccarrone

Background Anandamide (AEA) is an endogenous lipid mediator that exerts several effects in the brain as well as in peripheral tissues. These effects are mediated mainly by two types of cannabinoid receptors, named CB1R and CB2R, making AEA a prominent member of the “endocannabinoid” family. Also immune cells express CB1 and CB2 receptors, and possess the whole machinery responsible for endocannabinoid metabolism. Not surprisingly, evidence has been accumulated showing manifold roles of endocannabinoids in the modulation of the immune system. However, details of such a modulation have not yet been disclosed in primary human T-cells. Methodology/Significance In this investigation we used flow cytometry and ELISA tests, in order to show that AEA suppresses proliferation and release of cytokines like IL-2, TNF-α and INF-γ from activated human peripheral T-lymphocytes. However, AEA did not exert any cytotoxic effect on T-cells. The immunosuppression induced by AEA was mainly dependent on CB2R, since it could be mimicked by the CB2R selective agonist JWH-015, and could be blocked by the specific CB2R antagonist SR144528. Instead the selective CB1R agonist ACEA, or the selective CB1R antagonist SR141716, were ineffective. Furthermore, we demonstrated an unprecedented immunosuppressive effect of AEA on IL-17 production, a typical cytokine that is released from the unique CD4+ T-cell subset T-helper 17. Conclusions/Significance Overall, our study investigates for the first time the effects of the endocannabinoid AEA on primary human T-lymphocytes, demonstrating that it is a powerful modulator of immune cell functions. In particular, not only we clarify that CB2R mediates the immunosuppressive activity of AEA, but we are the first to describe such an immunosuppressive effect on the newly identified Th-17 cells. These findings might be of crucial importance for the rational design of new endocannabinoid-based immunotherapeutic approaches.

Collaboration


Dive into the Luca Battistini's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marco Salvetti

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Claudio Gasperini

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Giorgio Bernardi

Stazione Zoologica Anton Dohrn

View shared research outputs
Top Co-Authors

Avatar

Giovanni Ristori

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Celia F. Brosnan

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Adamo Diamantini

Necker-Enfants Malades Hospital

View shared research outputs
Top Co-Authors

Avatar

Fabrizio Poccia

University of Rome Tor Vergata

View shared research outputs
Researchain Logo
Decentralizing Knowledge