Network


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

Hotspot


Dive into the research topics where Alessio Biagioni is active.

Publication


Featured researches published by Alessio Biagioni.


Journal of Molecular Medicine | 2015

Inhibition of uPAR-TGFβ crosstalk blocks MSC-dependent EMT in melanoma cells

Anna Laurenzana; Alessio Biagioni; Francesca Bianchini; Silvia Peppicelli; Anastasia Chillà; Francesca Margheri; Cristina Luciani; Nicola Pimpinelli; Mario Del Rosso; Lido Calorini; Gabriella Fibbi

The capacity of cancer cells to undergo epithelial-to-mesenchymal transition (EMT) is now considered a hallmark of tumor progression, and it is known that interactions between cancer cells and mesenchymal stem cells (MSCs) of tumor microenvironment may promote this program. Herein, we demonstrate that MSC-conditioned medium (MSC-CM) is a potent inducer of EMT in melanoma cells. The EMT profile acquired by MSC-CM-exposed melanoma cells is characterized by an enhanced level of mesenchymal markers, including TGFβ/TGFβ-receptors system upregulation, by increased invasiveness and uPAR expression, and in vivo tumor growth. Silencing TGFβ in MSC is found to abrogate ability of MSC to promote EMT characteristics in melanoma cells, together with uPAR expression, and this finding is strengthened using an antagonist peptide of TGFβRIII, the so-called P17. Finally, we demonstrate that the uPAR antisense oligonucleotide (uPAR aODN) may inhibit EMT of melanoma cells either stimulated by exogenous TGFβ or MSC-CM. Thus, uPAR upregulation in melanoma cells exposed to MSC-medium drives TGFβ-mediated EMT. On the whole, TGFβ/uPAR dangerous liaison in cancer cell/MSC interactions may disclose a new strategy to abrogate melanoma progression.Key messageMesenchymal stem cell (MSC)-conditioned medium induces EMT-like profile in melanoma.MSC-derived TGFβ promotes uPAR and TGFβ/TGFβ-receptor upregulation in melanoma.TGFβ gene silencing in MSCs downregulates uPAR expression and EMT in melanoma.uPAR downregulation prevents MSC-induced EMT-like profile in melanoma cells.Inhibition of the dangerous TGFβ/uPAR relationship might abrogate melanoma progression.


Current Molecular Medicine | 2015

Endothelial Progenitor Cells in Sprouting Angiogenesis: Proteases Pave the Way.

Anna Laurenzana; Gabriella Fibbi; Francesca Margheri; Alessio Biagioni; Cristina Luciani; M. Del Rosso; Anastasia Chillà

Sprouting angiogenesis consists of the expansion and remodelling of existing vessels, where the vascular sprouts connect each other to form new vascular loops. Endothelial Progenitor Cells (EPCs) are a subtype of stem cells, with high proliferative potential, able to differentiate into mature Endothelial Cells (ECs) during the neovascularization process. In addition to this direct structural role EPCs improve neovascularization, also secreting numerous pro-angiogenic factors able to enhance the proliferation, survival and function of mature ECs, and other surrounding progenitor cells. While sprouting angiogenesis by mature ECs involves resident ECs, the vasculogenic contribution of EPCs is a high hurdle race. Bone marrowmobilized EPCs have to detach from the stem cell niche, intravasate into bone marrow vessels, reach the hypoxic area or tumour site, extravasate and incorporate into the new vessel lumen, thus complementing the resident mature ECs in sprouting angiogenesis. The goal of this review is to highlight the role of the main protease systems able to control each of these steps. The pivotal protease systems here described, involved in vascular patterning in sprouting angiogenesis, are the matrix-metalloproteinases (MMPs), the serineproteinases urokinase-type plasminogen activator (uPA) associated with its receptor (uPAR) and receptorassociated plasminogen/plasmin, the neutrophil elastase and the cathepsins. Since angiogenesis plays a critical role not only in physiological but also in pathological processes, such as in tumours, controlling the contribution of EPCs to the angiogenic process, through the regulation of the protease systems involved, could yield new opportunities for the therapeutic prospect of efficient control of pathological angiogenesis.


Journal of Experimental & Clinical Cancer Research | 2017

Type II CRISPR/Cas9 approach in the oncological therapy

Alessio Biagioni; Anastasia Chillà; Elena Andreucci; Anna Laurenzana; Francesca Margheri; Silvia Peppicelli; M. Del Rosso; Gabriella Fibbi

CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) is a prokaryotic adaptable immune mechanism used by many bacteria and archaea to protect themselves from foreign nucleic acids. This complex system can recognize and cut non-self DNA in order to provide the prokaryotic organisms a strong defense against foreign viral or plasmid attacks and make the cell immune from further assaults. Today, it has been adapted to be used in vitro and in vivo in eukaryotic cells to perform a complete and highly selective gene knockout or a specific gene editing. The ease of use and the low cost are only two features that have made it very popular among the scientific community and the possibility to be used as a clinical treatment in several genetic derived pathologies has rapidly spread its fame worldwide. However, CRISPR is still not fully understood and many efforts need to be done in order to make it a real power tool for the human clinical treatment especially for oncological patients. Indeed, since cancer originates from non-lethal genetic disorders, CRISPR discovery fuels the hope to strike tumors on their roots. More than 4000 papers regarding CRISPR were published in the last ten years and only few of them take in count the possible applications in oncology. The purpose of this review is to clarify many problematics on the CRISPR usage and highlight its potential in oncological therapy.


Journal of Cellular and Molecular Medicine | 2015

Differential uPAR recruitment in caveolar-lipid rafts by GM1 and GM3 gangliosides regulates endothelial progenitor cells angiogenesis

Francesca Margheri; Laura Papucci; Nicola Schiavone; Riccardo D'Agostino; Silvana Trigari; Simona Serratì; Anna Laurenzana; Alessio Biagioni; Cristina Luciani; Anastasia Chillà; Elena Andreucci; Tommaso Del Rosso; Giancarlo Margheri; Mario Del Rosso; Gabriella Fibbi

Gangliosides and the urokinase plasminogen activator receptor (uPAR) tipically partition in specialized membrane microdomains called lipid‐rafts. uPAR becomes functionally important in fostering angiogenesis in endothelial progenitor cells (EPCs) upon recruitment in caveolar‐lipid rafts. Moreover, cell membrane enrichment with exogenous GM1 ganglioside is pro‐angiogenic and opposite to the activity of GM3 ganglioside. On these basis, we first checked the interaction of uPAR with membrane models enriched with GM1 or GM3, relying on the adoption of solid‐supported mobile bilayer lipid membranes with raft‐like composition formed onto solid hydrophilic surfaces, and evaluated by surface plasmon resonance (SPR) the extent of uPAR recruitment. We estimated the apparent dissociation constants of uPAR‐GM1/GM3 complexes. These preliminary observations, indicating that uPAR binds preferentially to GM1‐enriched biomimetic membranes, were validated by identifying a pro‐angiogenic activity of GM1‐enriched EPCs, based on GM1‐dependent uPAR recruitment in caveolar rafts. We have observed that addition of GM1 to EPCs culture medium promotes matrigel invasion and capillary morphogenesis, as opposed to the anti‐angiogenesis activity of GM3. Moreover, GM1 also stimulates MAPKinases signalling pathways, typically associated with an angiogenesis program. Caveolar‐raft isolation and Western blotting of uPAR showed that GM1 promotes caveolar‐raft partitioning of uPAR, as opposed to control and GM3‐challenged EPCs. By confocal microscopy, we have shown that in EPCs uPAR is present on the surface in at least three compartments, respectively, associated to GM1, GM3 and caveolar rafts. Following GM1 exogenous addition, the GM3 compartment is depleted of uPAR which is recruited within caveolar rafts thereby triggering angiogenesis.


International Journal of Cancer | 2017

uPA/uPAR system activation drives a glycolytic phenotype in melanoma cells

Anna Laurenzana; Anastasia Chillà; Cristina Luciani; Silvia Peppicelli; Alessio Biagioni; Francesca Bianchini; Elena Tenedini; Eugenio Torre; Alessandra Mocali; Lido Calorini; Francesca Margheri; Gabriella Fibbi; Mario Del Rosso

In this manuscript, we show the involvement of the uPA/uPAR system in the regulation of aerobic glycolysis of melanoma cells. uPAR over‐expression in human melanoma cells controls an invasive and glycolytic phenotype in normoxic conditions. uPAR down‐regulation by siRNA or its uncoupling from integrins, and hence from integrin‐linked tyrosine kinase receptors (IL‐TKRs), by an antagonist peptide induced a striking inhibition of the PI3K/AKT/mTOR/HIF1α pathway, resulting into impairment of glucose uptake, decrease of several glycolytic enzymes and of PKM2, a checkpoint that controls metabolism of cancer cells. Further, binding of uPA to uPAR regulates expression of molecules that govern cell invasion, including extracellular matrix metallo‐proteinases inducer (EMPPRIN) and enolase, a glycolytyc enzyme that also serves as a plasminogen receptor, thus providing a common denominator between tumor metabolism and phenotypic invasive features. Such effects depend on the α5β1‐integrin‐mediated uPAR connection with EGFR in melanoma cells with engagement of the PI3K‐mTOR‐HIFα pathway. HIF‐1α trans‐activates genes whose products mediate tumor invasion and glycolysis, thus providing the common denominator between melanoma metabolism and its invasive features. These findings unveil a unrecognized interaction between the invasion‐related uPAR and IL‐TKRs in the control of glycolysis and disclose a new pharmacological target (i.e., uPAR/IL‐TKRs axis) for the therapy of melanoma.


Journal of Experimental & Clinical Cancer Research | 2018

Mature and progenitor endothelial cells perform angiogenesis also under protease inhibition: the amoeboid angiogenesis

Anastasia Chillà; Francesca Margheri; Alessio Biagioni; Mario Del Rosso; Gabriella Fibbi; Anna Laurenzana

BackgroundControlling vascular growth is a challenging aim for the inhibition of tumor growth and metastasis. The amoeboid and mesenchymal types of invasiveness are two modes of migration interchangeable in cancer cells: the Rac-dependent mesenchymal migration requires the activity of proteases; the Rho-ROCK-dependent amoeboid motility is protease-independent and has never been described in endothelial cells.MethodsA cocktail of physiologic inhibitors (Ph-C) of serine-proteases, metallo-proteases and cysteine-proteases, mimicking the physiological environment that cells encounter during their migration within the angiogenesis sites was used to induce amoeboid style migration of Endothelial colony forming cells (ECFCs) and mature endothelial cells (ECs). To evaluate the mesenchymal-ameboid transition RhoA and Rac1 activation assays were performed along with immunofluorescence analysis of proteins involved in cytoskeleton organization. Cell invasion was studied in Boyden chambers and Matrigel plug assay for the in vivo angiogenesis.ResultsIn the present study we showed in both ECFCs and ECs, a decrease of activated Rac1 and an increase of activated RhoA upon shifting of cells to the amoeboid conditions. In presence of Ph-C inhibitors both cell lines acquired a round morphology and Matrigel invasion was greatly enhanced with respect to that observed in the absence of protease inhibition. We also observed that the urokinase-plasminogen-activator (uPAR) receptor silencing and uPAR-integrin uncoupling with the M25 peptide abolished both mesenchymal and amoeboid angiogenesis of ECFCs and ECs in vitro and in vivo, indicating a role of the uPAR-integrin-actin axis in the regulation of amoeboid angiogenesis. Furthermore, under amoeboid conditions endothelial cells seem to be indifferent to VEGF stimulation, which induces an amoeboid signaling pattern also in mesenchymal conditions.ConclusionHere we first provide a data set disclosing that endothelial cells can move and differentiate into vascular structures in vitro and in vivo also in the absence of proteases activity, performing a new type of neovascularization: the “amoeboid angiogenesis”. uPAR is indispensable for ECs and ECFCs to perform an efficient amoeboid angiogenesis. Therefore, uPAR silencing or the block of its integrin-interaction, together with standard treatment against VEGF, could be a possible solution for angiogenesis inhibition.


Molecular and Cellular Biochemistry | 2017

Triazole RGD antagonist reverts TGFβ1-induced endothelial-to-mesenchymal transition in endothelial precursor cells

Francesca Bianchini; Silvia Peppicelli; Pierangelo Fabbrizzi; Alessio Biagioni; Benedetta Mazzanti; Gloria Menchi; Lido Calorini; Alberto Pupi; Andrea Trabocchi

Fibrosis is the dramatic consequence of a dysregulated reparative process in which activated fibroblasts (myofibroblasts) and Transforming Growth Factor β1 (TGFβ1) play a central role. When exposed to TGFβ1, fibroblast and epithelial cells differentiate in myofibroblasts; in addition, endothelial cells may undergo endothelial-to-mesenchymal transition (EndoMT) and actively participate to the progression of fibrosis. Recently, the role of αv integrins, which recognize the Arg-Gly-Asp (RGD) tripeptide, in the release and signal transduction activation of TGFβ1 became evident. In this study, we present a class of triazole-derived RGD antagonists that interact with αvβ3 integrin. Above different compounds, the RGD-2 specifically interferes with integrin-dependent TGFβ1 EndoMT in Endothelial Colony-Forming Cells (ECPCs) derived from circulating Endothelial Precursor Cells (ECPCs). The RGD-2 decreases the amount of membrane-associated TGFβ1, and reduces both ALK5/TGFβ1 type I receptor expression and Smad2 phosphorylation in ECPCs. We found that RGD-2 antagonist reverts EndoMT, reducing α-smooth muscle actin (α-SMA) and vimentin expression in differentiated ECPCs. Our results outline the critical role of integrin in fibrosis progression and account for the opportunity of using integrins as target for anti-fibrotic therapeutic treatment.


Oncotarget | 2014

The receptor for urokinase-plasminogen activator (uPAR) controls plasticity of cancer cell movement in mesenchymal and amoeboid migration style

Francesca Margheri; Cristina Luciani; Maria Letizia Taddei; Elisa Giannoni; Anna Laurenzana; Alessio Biagioni; Anastasia Chillà; Paola Chiarugi; Gabriella Fibbi; Mario Del Rosso


Oncotarget | 2014

Melanoma cell therapy: Endothelial progenitor cells as shuttle of the MMP12 uPAR-degrading enzyme.

Anna Laurenzana; Alessio Biagioni; Silvia D’Alessio; Francesca Bianchini; Anastasia Chillà; Francesca Margheri; Cristina Luciani; Benedetta Mazzanti; Nicola Pimpinelli; Eugenio Torre; Silvio Danese; Lido Calorini; Mario Del Rosso; Gabriella Fibbi


Oncotarget | 2016

Tumor-tropic endothelial colony forming cells (ECFCs) loaded with near-infrared sensitive Au nanoparticles: A “cellular stove” approach to the photoablation of melanoma

Giancarlo Margheri; Angela Zoppi; Roberto Olmi; Silvana Trigari; Rita Traversi; Mirko Severi; Daniele Bani; Francesca Bianchini; Eugenio Torre; Francesca Margheri; Anastasia Chillà; Alessio Biagioni; Lido Calorini; Anna Laurenzana; Gabriella Fibbi; Mario Del Rosso

Collaboration


Dive into the Alessio Biagioni's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge