Network


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

Hotspot


Dive into the research topics where Agostina Longo is active.

Publication


Featured researches published by Agostina Longo.


PLOS ONE | 2009

Paracrine Diffusion of PrPC and Propagation of Prion Infectivity by Plasma Membrane-Derived Microvesicles

Vincenzo Mattei; Maria Grazia Barenco; Vincenzo Tasciotti; Tina Garofalo; Agostina Longo; Klaus Boller; Johannes Löwer; Roberta Misasi; Fabio Montrasio; Maurizio Sorice

Cellular prion protein (PrPc) is a physiological constituent of eukaryotic cells. The cellular pathways underlying prions spread from the sites of prions infection/peripheral replication to the central nervous system are still not elucidated. Membrane-derived microvesicles (MVs) are submicron (0.1–1 µm) particles, that are released by cells during plasma membrane shedding processes. They are usually liberated from different cell types, mainly upon activation as well as apoptosis, in this case, one of their hallmarks is the exposure of phosphatidylserine in the outer leaflet of the membrane. MVs are also characterized by the presence of adhesion molecules, MHC I molecules, as well as of membrane antigens typical of their cell of origin. Evidence exists that MVs shedding provide vehicles to transfer molecules among cells, and that MVs are important modulators of cell-to-cell communication. In this study we therefore analyzed the potential role of membrane-derived MVs in the mechanism(s) of PrPC diffusion and prion infectivity transmission. We first identified PrPC in association with the lipid raft components Fyn, flotillin-2, GM1 and GM3 in MVs from plasma of healthy human donors. Similar findings were found in MVs from cell culture supernatants of murine neuronal cells. Furthermore we demonstrated that PrPSc is released from infected murine neuronal cells in association with plasma membrane-derived MVs and that PrPSc-bearing MVs are infectious both in vitro and in vivo. The data suggest that MVs may contribute both to the intercellular mechanism(s) of PrPC diffusion and signaling as well as to the process of prion spread and neuroinvasion.


FEBS Letters | 2001

Evidence for cell surface association between CXCR4 and ganglioside GM3 after gp120 binding in SupT1 lymphoblastoid cells

Maurizio Sorice; Tina Garofalo; Roberta Misasi; Agostina Longo; Vincenzo Mattei; Patrizio Sale; Vincenza Dolo; Roberto Gradini; Antonio Pavan

CXCR4 (fusin) is a chemokine receptor which is involved as a coreceptor in gp120 binding to the cell surface. In this study we provide evidence that binding of gp120 triggers CXCR4 recruitment to glycosphingolipid‐enriched microdomains. Scanning confocal microscopy showed a nearly complete localization of CXCR4 within GM3‐enriched plasma membrane domains of SupT1 cells and coimmunoprecipitation experiments revealed that CXCR4 was immunoprecipitated by IgG anti‐GM3 after gp120 pretreatment. These findings reveal that gp120 binding induces a strict association between CXCR4 and ganglioside GM3, supporting the view that GM3 and CXCR4 are components of a functional multimolecular complex critical for HIV‐1 entry.


Glycoconjugate Journal | 2003

Role of GM3-enriched microdomains in signal transduction regulation in T lymphocytes

Maurizio Sorice; Agostina Longo; Tina Garofalo; Vincenzo Mattei; Roberta Misasi; Antonio Pavan

Gangliosides, sialic acid containing glycosphigolipids, are ubiquitous constituents of cell plasma membranes. Each cell type shows a peculiar ganglioside expression pattern. In human T lymphocytes monosialoganglioside GM3 represents the main ganglioside constituent of cell plasma membrane where it is concentrated in glycosphingolipid-enriched microdomains (GEM). The presence of tyrosine kinase receptors, mono- (Ras, Rap) and heterotrimeric G proteins, Src-like tyrosine kinases (lck, lyn, fyn), PKC isozymes, glycosylphosphatidylinositol (GPI)-anchored proteins and, after T cell activation, the Syk-family kinase Zap-70, prompts these portions of the plasma membrane to be considered as “glycosignaling domains.” In particular, during T cell activation and/or other dynamic functions of the cell, such as apoptosis, key signaling molecules are recruited to these microdomains, where they strictly interact with GM3. The association of transducer proteins with GM3 in microdomains suggests that this ganglioside is the main marker of GEM in human lymphocytes and is a component of a cell plasma membrane multimolecular signaling complex involved in cell-cell interaction, signal transduction, and cell activation. Published in 2004.


Blood | 2012

Autoantibodies specific to a peptide of β2-glycoprotein I cross-react with TLR4, inducing a proinflammatory phenotype in endothelial cells and monocytes

Tania Colasanti; Cristiano Alessandri; Antonella Capozzi; Maurizio Sorice; Federica Delunardo; Agostina Longo; Marina Pierdominici; Fabrizio Conti; S. Truglia; Alessandra Siracusano; Guido Valesini; Elena Ortona; Paola Margutti

β(2)-glycoprotein I (β(2)GPI) is the major antigenic target for antiphospholipid Abs. Anti-β(2)GPI Abs are a heterogeneous population of Igs targeting all domains of the molecule. Abs specific to β(2)GPI domain I are strongly associated with thrombosis and obstetric complications. In the present study, we sought to understand the possible pathogenic mechanism for this subset of anti-β(2)GPI Abs, investigating their potential cross-reactivity with other self-proteins involved in inflammatory or coagulant events. We compared the amino acid sequence of the β(2)GPI domain I with human proteins in a protein databank and identified a peptide sharing 88% identity with an epitope of human TLR4. A high percentage of patients with antiphospholipid syndrome (41%) and systemic lupus erythematosus (50%) presented serum IgG specific to this peptide. Anti-β(2)GPI peptide Abs binding the TLR4 were able to induce NF-κB activation in HEK293 cells that were stably transfected with the TLR4 gene. Anti-β(2)GPI peptide Abs induced activation of TLR4 and triggered interleukin-1 receptor-associated kinase phosphorylation and NF-κB translocation, promoting VCAM expression on endothelial cells and TNF-α release by monocytes. In conclusion, our observations suggest a novel pathogenic mechanism in the TLR4 stimulation by anti-β(2)GPI peptide Abs that links adaptive immune responses with innate immunity in antiphospholipid syndrome and systemic lupus erythematosus.


Clinical & Developmental Immunology | 2014

The mosaic of "seronegative" antiphospholipid syndrome.

Fabrizio Conti; Antonella Capozzi; S. Truglia; Emanuela Lococo; Agostina Longo; Roberta Misasi; Cristiano Alessandri; Guido Valesini; Maurizio Sorice

In the clinical practice it is possible to find patients with clinical signs suggestive of antiphospholipid syndrome (APS), who are persistently negative for the laboratory criteria of APS, that is, anti-cardiolipin antibodies (aCL), anti-β 2-GPI antibodies and lupus anticoagulant. Therefore, it was proposed for these cases the term of seronegative APS (SN-APS). In order to detect autoantibodies with different methodological approaches, sera from 24 patients with SN-APS were analysed for anti-phospholipid antibodies using TLC immunostaining, for anti-vimentin/cardiolipin antibodies by enzyme-linked immunosorbent assay (ELISA), and for anti-annexin V and anti-prothrombin antibodies by ELISA and dot blot. Control groups of our study were 25 patients with APS, 18 with systemic lupus erythematosus (SLE), and 32 healthy controls. Results revealed that 13/24 (54.2%) SN-APS sera were positive for aCL (9 of whom were also positive for lysobisphosphatidic acid) by TLC immunostaining, 11/24 (45.8%) for anti-vimentin/cardiolipin antibodies, 3/24 (12.5%) for anti-prothrombin antibodies, and 1/24 (4.2%) for anti-annexin V antibodies. These findings suggest that in sera from patients with SN-APS, antibodies may be detected using “new” antigenic targets (mainly vimentin/cardiolipin) or methodological approaches different from traditional techniques (mainly TLC immunostaining). Thus, SN-APS represents a mosaic, in which antibodies against different antigenic targets may be detected.


Clinical and Experimental Immunology | 2012

Thin-layer chromatography immunostaining in detecting anti-phospholipid antibodies in seronegative anti-phospholipid syndrome

Fabrizio Conti; Cristiano Alessandri; Maurizio Sorice; Antonella Capozzi; Agostina Longo; Tina Garofalo; Roberta Misasi; D. Bompane; G. R. V. Hughes; Munther A. Khamashta; Guido Valesini

In clinical practice it is possible to find patients with clinical signs suggestive of anti‐phospholipid syndrome (APS) who are persistently negative for the routinely used anti‐phospholipid antibodies (aPL). Therefore, the term proposed for these cases was seronegative APS (SN‐APS). We investigated the clinical usefulness of thin‐layer chromatography (TLC) immunostaining in detecting serum aPL in patients presenting clinical features of SN‐APS. Sera from 36 patients with SN‐APS, 19 patients with APS, 18 patients with systemic lupus erythematosus (SLE), 20 anti‐hepatitis C virus (HCV)‐positive subjects and 32 healthy controls were examined for aPL using TLC immunostaining. Anti‐β2‐glycoprotein‐I, anti‐annexin II, anti‐annexin V and anti‐prothrombin antibodies were tested by enzyme‐linked immunosorbent assays (ELISA). Eahy926, a human‐derived endothelial cell line, was incubated with immunoglobulin (Ig)G fraction from SN‐APS patients and analysis of phospho‐interleukin (IL)‐1 receptor‐associated kinase (IRAK) and phospho‐nuclear factor (NF)‐κB was performed by Western blot, vascular cell adhesion molecule 1 (VCAM‐1) expression by cytofluorimetric analysis and supernatants tissue factor (TF) levels by ELISA. TLC immunostaining showed aPL in 58·3% of SN‐APS patients: anti‐cardiolipin in 47·2%, anti‐lyso(bis)phosphatidic acid in 41·7% and anti‐phosphatidylethanolamine in 30·5%. Six of 36 patients showed anti‐annexin II. Incubation of Eahy926 cells with IgG from SN‐APS induced IRAK phosphorylation, NF‐κB activation, VCAM‐1 surface expression and TF cell release. TLC immunostaining could identify the presence of aPL in patients with SN‐APS. Moreover, the results suggest the proinflammatory and procoagulant effects in vitro of these antibodies.


Clinical & Developmental Immunology | 2015

New antigenic targets and methodological approaches for refining laboratory diagnosis of antiphospholipid syndrome.

Roberta Misasi; Antonella Capozzi; Agostina Longo; Serena Recalchi; Emanuela Lococo; Cristiano Alessandri; Fabrizio Conti; Guido Valesini; Maurizio Sorice

Antiphospholipid antibodies (aPLs) are a heterogeneous group of antibodies directed against phospholipids or protein/phospholipid complexes. Currently, aPLs are assessed using either “solid-phase” assays that identify anticardiolipin antibodies and anti-β2-glycoprotein I antibodies or “liquid-phase” assay that identifies lupus anticoagulant. However, in the last few years, “new” antigenic targets and methodological approaches have been employed for refining laboratory diagnosis of antiphospholipid syndrome (APS). In this review the potential diagnostic value of antibodies to domains of β2-GPI, prothrombin/phosphatidylserine, vimentin/cardiolipin, protein S, protein C, annexin A2, annexin A5, and phospholipid antigens is discussed. Moreover, new technical approaches, including chemiluminescence, multiline dot assay, and thin layer chromatography (TLC) immunostaining, which utilize different supports for detection of aPL, have been developed. A special focus has been dedicated on “seronegative” APS, that is, those patients with a clinical profile suggestive of APS (thromboses, recurrent miscarriages, or foetal loss), who are persistently negative for the routinely used aPL. Recent findings suggest that, in sera from patients with SN-APS, antibodies may be detected using “new” antigenic targets (mainly vimentin/cardiolipin) or methodological approaches different from traditional techniques (TLC immunostaining). Thus, APS represents a mosaic, in which antibodies against different antigenic targets may be detected thanks to the continuously evolving new technologies.


Glycoconjugate Journal | 2000

Association between GM3 and CD4-Ick complex in human peripheral blood lymphocytes.

Maurizio Sorice; Tina Garofalo; Roberta Misasi; Agostina Longo; Joanna Mikulak; Vincenza Dolo; Giuseppe Pontieri; Antonio Pavan

The aim of this study was to further elucidate our previous observation on molecular interaction of GM3, CD4 and p56lck in microdomains of human peripheral blood lymphocytes (PBL). We analyzed GM3 distribution by immunoelectron microscopy and the association between GM3 and CD4-p56lck complex by scanning confocal microscopy and co-immunoprecipitation experiments. Scanning confocal microscopy analysis showed an uneven signal distribution of GM3 molecules over the surface of human lymphocytes. Nearly complete colocalization areas indicated that CD4 molecules were distributed in GM3-enriched plasma membrane domains. Co-immunoprecipitation experiments revealed that CD4 and p56lck were immunoprecipitated by IgG anti-GM3, demonstrating that GM3 tightly binds to the CD4-p56lck complex in human PBL. In order to verify whether GM3 association with CD4 molecules may depend on the presence of p56lck, we analyzed this association in U937, a CD4+and p56lck negative cell line. The immunoprecipitation with anti-GM3 revealed the presence of a 58[emsp4 ]kDa band immunostained with anti-CD4 Ab, suggesting that the GM3-CD4 interaction does not require its association with p56lck. These findings support the view that GM3 enriched-domains may represent a functional multimolecular complex involved in signal transduction and cell activation.


Apoptosis | 2017

Changes in membrane lipids drive increased endocytosis following Fas ligation

Mauro Degli Esposti; Paola Matarrese; Antonella Tinari; Agostina Longo; Serena Recalchi; Roya Khosravi-Far; Walter Malorni; Roberta Misasi; Tina Garofalo; Maurizio Sorice

Once activated, some surface receptors promote membrane movements that open new portals of endocytosis, in part to facilitate the internalization of their activated complexes. The prototypic death receptor Fas (CD95/Apo1) promotes a wave of enhanced endocytosis that induces a transient intermixing of endosomes with mitochondria in cells that require mitochondria to amplify death signaling. This initiates a global alteration in membrane traffic that originates from changes in key membrane lipids occurring in the endoplasmic reticulum (ER). We have focused the current study on specific lipid changes occurring early after Fas ligation. We analyzed the interaction between endosomes and mitochondria in Jurkat T cells by nanospray-Time-of-flight (ToF) Mass Spectrometry. Immediately after Fas ligation, we found a transient wave of lipid changes that drives a subpopulation of early endosomes to merge with mitochondria. The earliest event appears to be a decrease of phosphatidylcholine (PC), linked to a metabolic switch enhancing phosphatidylinositol (PI) and phosphoinositides, which are crucial for the formation of vacuolar membranes and endocytosis. Lipid changes occur independently of caspase activation and appear to be exacerbated by caspase inhibition. Conversely, inhibition or compensation of PC deficiency attenuates endocytosis, endosome-mitochondria mixing and the induction of cell death. Deficiency of receptor interacting protein, RIP, also limits the specific changes in membrane lipids that are induced by Fas activation, with parallel reduction of endocytosis. Thus, Fas activation rapidly changes the interconversion of PC and PI, which then drives enhanced endocytosis, thus likely propagating death signaling from the cell surface to mitochondria and other organelles.


Immunobiology | 2000

C3 Synthesis and CRs Expression during DiHerentiation of a Murine Stem Cell Line

Stefania Mardente; Agostina Longo; Luisa Lenti; Giovanna De Capua; Wolfgang M. Prodinger; Ida Silvestri; Giuseppe Pontieri; Marcella Lipari

C3 production, release and CRs expression during the neutrophilic differentiation of a murine non tumorigenic cell line is investigated. The murine non tumorigenic cell line 32DCl3(G) which undergoes terminal differentiation into polymorphonuclear granulocytes when cultured in presence of G-CSF was selected as a suitable in vitro model for this study. The results show that as the cells progress into the differentiation program, levels of C3 mRNA increase, accompanied by increased C3 production. As differentiation progresses the cells gradually express CRs on their surface; these are undetectable on the surface of undifferentiated cells. As a consequence of CRs appearance, cells become able to bind C3 through receptorial binding. Differences were found in the modality of C3 secretion: differentiated cells tend to store C3 in their intracellular compartments rather than secrete it continuously into the medium and they respond to membrane stimulation with increased secretion of C3. Treatment of 32DCl3(G) with TNF-alpha increased C3 production in a time- and dose-dependent fashion. Cell response to this stimulus progressively increases during the differentiation process suggesting that they acquire functionality in the signal transduction mechanisms.

Collaboration


Dive into the Agostina Longo's collaboration.

Top Co-Authors

Avatar

Maurizio Sorice

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Roberta Misasi

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Tina Garofalo

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Guido Valesini

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Antonella Capozzi

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fabrizio Conti

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Vincenzo Mattei

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Giuseppe Pontieri

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge