Network


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

Hotspot


Dive into the research topics where Massimo Resnati is active.

Publication


Featured researches published by Massimo Resnati.


The EMBO Journal | 1996

Proteolytic cleavage of the urokinase receptor substitutes for the agonist-induced chemotactic effect.

Massimo Resnati; M Guttinger; S Valcamonica; Nicolai Sidenius; Francesco Blasi; Francesca Fazioli

Physiological concentrations of urokinase plasminogen activator (uPA) stimulated a chemotactic response in human monocytic THP‐1 through binding to the urokinase receptor (uPAR). The effect did not require the protease moiety of uPA, as stimulation was achieved also with the N‐terminal fragment (ATF), while the 33 kDa low molecular weight uPA was ineffective. Co‐immunoprecipitation experiments showed association of uPAR with intracellular kinase(s), as demonstrated by in vitro kinase assays. Use of specific antibodies identified p56/p59hck as a kinase associated with uPAR in THP‐1 cell extracts. Upon addition of ATF, p56/p59hck activity was stimulated within 2 min and returned to normal after 30 min. Since uPAR lacks an intracellular domain capable of interacting with intracellular kinase, activation of p56/p59hck must require a transmembrane adaptor. Evidence for this was strongly supported by the finding that a soluble form of uPAR (suPAR) was capable of inducing chemotaxis not only in THP‐1 cells but also in cells lacking endogenous uPAR (IC50, 5 pM). However, activity of suPAR require chymotrypsin cleavage between the N‐terminal domain D1 and D2 + D3. Chymotrypsin‐cleaved suPAR also induced activation of p56/p59hck in THP‐1 cells, with a time course comparable with ATF. Our data show that uPA‐induced signal transduction takes place via uPAR, involves activation of intracellular tyrosine kinase(s) and requires an as yet undefined adaptor capable of connecting the extracellular ligand binding uPAR to intracellular transducer(s).


The EMBO Journal | 1997

A urokinase-sensitive region of the human urokinase receptor is responsible for its chemotactic activity.

Francesca Fazioli; Massimo Resnati; Nicolai Sidenius; Yuichiro Higashimoto; Ettore Appella; Francesco Blasi

The role of urokinase‐type plasminogen activator (uPA) and its receptor (uPAR/CD87) in cell migration and invasion is well substantiated. Recently, uPA has been shown to be essential in cell migration, since uPA−/− mice are greatly impaired in inflammatory cell recruitment. We have shown previously that the uPA‐induced chemotaxis requires interaction with and modification of uPAR/CD87, which is the true chemoattracting molecule acting through an unidentified cell surface component which mediates this cell surface chemokine activity. By expressing and testing several uPAR/CD87 variants, we have located and functionally characterized a potent uPAR/CD87 epitope that mimics the effects of the uPA–uPAR interaction. The chemotactic activity lies in the region linking domains 1 and 2, the only protease‐sensitive region of uPAR/CD87, efficiently cleaved by uPA at physiological concentrations. Synthetic peptides carrying this epitope promote chemotaxis and activate p56/p59hck tyrosine kinase. Both chemotaxis and kinase activation are pertussis toxin sensitive, involving a Gi/o protein in the pathway.


Oncogene | 2001

Urokinase/urokinase receptor and vitronectin/αvβ3 integrin induce chemotaxis and cytoskeleton reorganization through different signaling pathways

Bernard Degryse; Simone Orlando; Massimo Resnati; Shafaat Ahmed Rabbani; Francesco Blasi

Vitronectin (VN) and pro-urokinase (pro-uPA) stimulated migration of rat smooth muscle cells in a dose-dependent and additive way, and induced motile-type changes in cell morphology together with a complete reorganization of the actin filaments and of the microtubules. All these effects were inhibited by pertussis toxin, or by antibodies directed against the urokinase receptor (uPAR) or against the VN receptor αvβ3 suggesting that an association between the two receptors is required to mediate both signals. Investigation of the signaling pathways showed that increasing the intracellular cAMP resulted in a selective inhibition of VN-induced cell migration. On the other hand, PD 98059, an inhibitor of MEK, differentially inhibited the pro-uPA- but not the VN-induced cell migration. Phosphorylation and nuclear translocation of Erk by pro-uPA was directly observed. We conclude that the signaling pathways of pro-uPA and VN must be at least in part different.


FEBS Letters | 2001

PAI-1 inhibits urokinase-induced chemotaxis by internalizing the urokinase receptor

Bernard Degryse; Cornelis F. M. Sier; Massimo Resnati; Massimo Conese; Francesco Blasi

PAI‐1 (plasminogen activator inhibitor‐1) binds the urokinase‐type plasminogen activator (uPA) and causes its degradation via its receptor uPAR and low‐density lipoprotein receptor‐related protein (LRP). While both uPA and PAI‐1 are chemoattractants, we find that a preformed uPA–PAI‐1 complex has no chemotactic activity and that PAI‐1 inhibits uPA‐induced chemotaxis. The inhibitory effect of PAI‐1 on uPA‐dependent chemotaxis is reversed when uPAR internalization is inhibited by the 39 kDa receptor‐associated protein or by anti‐LRP antibodies. Under the same conditions, the uPA–PAI‐1 complex is turned into a chemoattractant causing cytoskeleton reorganization and extracellular‐regulated kinase/mitogen‐activated protein kinases activation. Thus, uPAR internalization by PAI‐1 regulates cell migration.


Journal of Cell Science | 2004

The soluble D2D388-274 fragment of the urokinase receptor inhibits monocyte chemotaxis and integrin-dependent cell adhesion

Federico Furlan; Simone Orlando; Carlo Laudanna; Massimo Resnati; Veronica Basso; Francesco Blasi; Anna Mondino

We have previously shown that chymotrypsin-cleaved soluble uPAR (D2D388-274) elicits migration of monocytic cells through interaction with FPRL-1, a G protein-coupled receptor that is homologous to the fMLP receptor. Here, we report that D2D388-274 also modulates the ability of monocytes to migrate in response to other chemokines. Pretreatment of monocytes with increasing amounts of D2D388-274 prevents cell migration in response to MCP-1, RANTES and fMLP. We demonstrate that D2D388-274 does not inhibit MCP-1 receptor binding, elicit CCR2 internalization and prevent MCP-1-induced intracellular Ca2+ increase. Thus, CCR2 receptor desensitization cannot account for D2D388-274-mediated inhibition of MCP-1-induced cell migration. Rather, we show that pretreatment of monocytes with D2D388-274 dramatically decreases chemokine-induced integrin-dependent rapid cell adhesion by interacting with FPRL-1. Together, our results indicate that chemokine-dependent cell migration can be regulated not only by homologous and heterologous receptor desensitization, but also by inhibition of integrin-dependent cell adhesion, an important step in cell transmigration.


Fibrinolysis and Proteolysis | 1994

The urokinase receptor: Structure, regulation and inhibitor-mediated internalization

Francesco Blasi; Massimo Conese; L.B. Møller; N. Pedersen; U. Cavallaro; M.V. Cubellis; Francesca Fazioli; L. Hemandez-Marrero; Paola Limongi; P. Munoz-Canoves; Massimo Resnati; Leena Riittinen; Nicolai Sidenius; E. Soravia; M.R. Soria; M.P. Stoppelli; Daniela Talarico; Tambet Teesalu; S. Valcamonica

Abstract The receptor for urokinase plasminogen activator (uPAR) acts as an anchorage site for uPA on the cell surface where it stimulates pro-uPA activation, allows the internalization of uPA:inhibitor and other complexes and sends directly or indirectly signals into the cell that may promote migration, adhesion and growth. It is a GPI-anchored, three-domain protein that belongs to the Ly6 family and is present at the focal and cell-to-cell contacts, where it concentrates uPA activity. Its activity appears to be important to regulate the invasiveness of human cancer cells both in vitro and in vivo, and its inhibition is now a target for antimetastatic therapy.


Journal of Immunology | 2000

uPA/uPAR System Is Active in Immature Dendritic Cells Derived from CD14+CD34+ Precursors and Is Down-Regulated upon Maturation

Elisabetta Ferrero; Katuscia Vettoretto; Attilio Bondanza; Antonello Villa; Massimo Resnati; Alessandro Poggi; Maria Raffaella Zocchi

We recently described a subset of peripheral CD14+CD34+ cells able to migrate across endothelial cell monolayers and differentiate into immunostimulatory dendritic cells (DC). In this paper we show that immature DC derived from CD14+CD34+ precursors are also capable of reverse transendothelial migration and extracellular matrix (ECM) invasion using the urokinase plasminogen activator receptor (uPAR). We found that these cells respond to macrophage-inflammatory protein (MIP)-1α, enhancing their ability to invade ECM and supporting the idea that immature DC are selectively recruited at the site of inflammation to expand the pool of APCs. Interestingly, MIP-1α was also capable of preventing the decreased matrix invasion observed by blocking uPAR, suggesting that the uPA/uPAR system and MIP-1α cooperate in driving immature DC migration through the subendothelial matrix. Upon exposure to maturating stimuli, such as TNF-α, CD14+CD34+-derived DC enhance their APC function and decrease the capacity of invading ECM; these changes are accompanied by altered expression and function of uPAR. Moreover, mature DC shift their sensitivity from MIP-1α to MIP-3β, enhancing their transendothelial migration capability in response to the latter chemokine. Our data support the hypothesis that bloodborne DC can move through ECM toward the site of pathogen entry where they differentiate into fully mature APCs with their motility and function regulated by microenvironmental stimuli, including MIP-1α, MIP-3β, and TNF-α.


Autophagy | 2015

A plastic SQSTM1/p62-dependent autophagic reserve maintains proteostasis and determines proteasome inhibitor susceptibility in multiple myeloma cells.

Enrico Milan; Tommaso Perini; Massimo Resnati; Ugo Orfanelli; Laura Oliva; Andrea Raimondi; Paolo Cascio; Angela Bachi; Magda Marcatti; Fabio Ciceri; Simone Cenci

Multiple myeloma (MM) is the paradigmatic proteasome inhibitor (PI) responsive cancer, but many patients fail to respond. An attractive target to enhance sensitivity is (macro)autophagy, recently found essential to bone marrow plasma cells, the normal counterpart of MM. Here, integrating proteomics with hypothesis-driven strategies, we identified the autophagic cargo receptor and adapter protein, SQSTM1/p62 as an essential component of an autophagic reserve that not only synergizes with the proteasome to maintain proteostasis, but also mediates a plastic adaptive response to PIs, and faithfully reports on inherent PI sensitivity. Lentiviral engineering revealed that SQSTM1 is essential for MM cell survival and affords specific PI protection. Under basal conditions, SQSTM1-dependent autophagy alleviates the degradative burden on the proteasome by constitutively disposing of substantial amounts of ubiquitinated proteins. Indeed, its inhibition or stimulation greatly sensitized to, or protected from, PI-induced protein aggregation and cell death. Moreover, under proteasome stress, myeloma cells selectively enhanced SQSTM1 de novo expression and reset its vast endogenous interactome, diverting SQSTM1 from signaling partners to maximize its association with ubiquitinated proteins. Saturation of such autophagic reserve, as indicated by intracellular accumulation of undigested SQSTM1-positive aggregates, specifically discriminated patient-derived myelomas inherently susceptible to PIs from primarily resistant ones. These aggregates correlated with accumulation of the endoplasmic reticulum, which comparative proteomics identified as the main cell compartment targeted by autophagy in MM. Altogether, the data integrate autophagy into our previously established proteasome load-versus-capacity model, and reveal SQSTM1 aggregation as a faithful marker of defective proteostasis, defining a novel prognostic and therapeutic framework for MM.


FEBS Letters | 1995

Biosynthesis and apical localization of the urokinase receptor in polarized MDCK epithelial cells.

Paola Limongi; Massimo Resnati; Luciano Hernandez-Marrero; Ottavio Cremona; Francesco Blasi; Francesca Fazioli

The biosynthesis and the surface localization of the urokinase plasminogen activator receptor (uPAR) were analysed in MDCK epithelial cells and in unpolarized fibroblasts. No differences were observed with respect to rate of synthesis, nature of precursors and time of surface appearance. uPAR was localized particularly at the focal and cell‐cell contacts when expressed in fibroblasts. On the contrary, in MDCK cells uPAR was found mostly on the apical surface; in agreement with its localization, down‐regulation of uPAR by the uPA‐PAI‐1 complex was observed only from the apical membrane.


Blood | 2017

The amyloidogenic light chain is a stressor that sensitizes plasma cells to proteasome inhibitor toxicity

Laura Oliva; Ugo Orfanelli; Massimo Resnati; Andrea Raimondi; Andrea Orsi; Enrico Milan; Giovanni Palladini; Paolo Milani; Fulvia Cerruti; Paolo Cascio; Simona Casarini; Paola Rognoni; Thierry Touvier; Magda Marcatti; Fabio Ciceri; Silvia Mangiacavalli; Alessandro Corso; Giampaolo Merlini; Simone Cenci

Systemic light chain (AL) amyloidosis is caused by the clonal production of an unstable immunoglobulin light chain (LC), which affects organ function systemically. Although pathogenic LCs have been characterized biochemically, little is known about the biology of amyloidogenic plasma cells (PCs). Intrigued by the unique response rates of AL amyloidosis patients to the first-in-class proteasome inhibitor (PI) bortezomib, we purified and investigated patient-derived AL PCs, in comparison with primary multiple myeloma (MM) PCs, the prototypical PI-responsive cells. Functional, biochemical, and morphological characterization revealed an unprecedented intrinsic sensitivity of AL PCs to PIs, even higher than that of MM PCs, associated with distinctive organellar features and expression patterns indicative of cellular stress. These consisted of expanded endoplasmic reticulum (ER), perinuclear mitochondria, and a higher abundance of stress-related transcripts, and were consistent with reduced autophagic control of organelle homeostasis. To test whether PI sensitivity stems from AL LC production, we engineered PC lines that can be induced to express amyloidogenic and nonamyloidogenic LCs, and found that AL LC expression alters cell growth and proteostasis and confers PI sensitivity. Our study discloses amyloidogenic LC production as an intrinsic PC stressor, and identifies stress-responsive pathways as novel potential therapeutic targets. Moreover, we contribute a cellular disease model to dissect the biology of AL PCs.

Collaboration


Dive into the Massimo Resnati's collaboration.

Top Co-Authors

Avatar

Francesco Blasi

Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico

View shared research outputs
Top Co-Authors

Avatar

Nicolai Sidenius

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anna Mondino

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Francesco Blasi

Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico

View shared research outputs
Top Co-Authors

Avatar

Guido Poli

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Massimo Alfano

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Bernard Degryse

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Enrico Milan

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Antonello Villa

University of Milano-Bicocca

View shared research outputs
Researchain Logo
Decentralizing Knowledge