Network


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

Hotspot


Dive into the research topics where Matteo Stravalaci is active.

Publication


Featured researches published by Matteo Stravalaci.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Synthetic amyloid-β oligomers impair long-term memory independently of cellular prion protein

Claudia Balducci; Marten Beeg; Matteo Stravalaci; Antonio Bastone; Alessandra Sclip; Emiliano Biasini; Laura Colombo; Claudia Manzoni; Tiziana Borsello; Roberto Chiesa; Marco Gobbi; Mario Salmona; Gianluigi Forloni

Inability to form new memories is an early clinical sign of Alzheimer’s disease (AD). There is ample evidence that the amyloid-β (Aβ) peptide plays a key role in the pathogenesis of this disorder. Soluble, bio-derived oligomers of Aβ are proposed as the key mediators of synaptic and cognitive dysfunction, but more tractable models of Aβ−mediated cognitive impairment are needed. Here we report that, in mice, acute intracerebroventricular injections of synthetic Aβ1–42 oligomers impaired consolidation of the long-term recognition memory, whereas mature Aβ1–42 fibrils and freshly dissolved peptide did not. The deficit induced by oligomers was reversible and was prevented by an anti-Aβ antibody. It has been suggested that the cellular prion protein (PrPC) mediates the impairment of synaptic plasticity induced by Aβ. We confirmed that Aβ1–42 oligomers interact with PrPC, with nanomolar affinity. However, PrP-expressing and PrP knock-out mice were equally susceptible to this impairment. These data suggest that Aβ1–42 oligomers are responsible for cognitive impairment in AD and that PrPC is not required.


Journal of Immunology | 2011

Alternative Pathway Activation of Complement by Shiga Toxin Promotes Exuberant C3a Formation That Triggers Microvascular Thrombosis

Marina Morigi; Miriam Galbusera; Sara Gastoldi; Monica Locatelli; Simona Buelli; Anna Pezzotta; Chiara Pagani; Marina Noris; Marco Gobbi; Matteo Stravalaci; Daniela Rottoli; Francesco Tedesco; Giuseppe Remuzzi; Carlamaria Zoja

Shiga toxin (Stx)-producing E.coli O157:H7 has become a global threat to public health; it is a primary cause of diarrhea-associated hemolytic uremic syndrome (HUS), a disorder of thrombocytopenia, microangiopathic hemolytic anemia, and acute renal failure with thrombi occluding renal microcirculation. In this study, we explored whether Stx triggers complement-dependent microvascular thrombosis in in vitro and in vivo experimental settings of HUS. Stx induced on human microvascular endothelial cell surface the expression of P-selectin, which bound and activated C3 via the alternative pathway, leading to thrombus formation under flow. In the search for mechanisms linking complement activation and thrombosis, we found that exuberant complement activation in response to Stx generated an increased amount of C3a that caused further endothelial P-selectin expression, thrombomodulin (TM) loss, and thrombus formation. In a murine model of HUS obtained by coinjection of Stx2 and LPS and characterized by thrombocytopenia and renal dysfunction, upregulation of glomerular endothelial P-selectin was associated with C3 and fibrin(ogen) deposits, platelet clumps, and reduced TM expression. Treatment with anti–P-selectin Ab limited glomerular C3 accumulation. Factor B-deficient mice after Stx2/LPS exhibited less thrombocytopenia and were protected against glomerular abnormalities and renal function impairment, indicating the involvement of complement activation via the alternative pathway in the glomerular thrombotic process in HUS mice. The functional role of C3a was documented by data showing that glomerular fibrin(ogen), platelet clumps, and TM loss were markedly decreased in HUS mice receiving C3aR antagonist. These results identify Stx-induced complement activation, via P-selectin, as a key mechanism of C3a-dependent microvascular thrombosis in diarrhea-associated HUS.


Journal of Biological Chemistry | 2013

An N-terminal Fragment of the Prion Protein Binds to Amyloid-β Oligomers and Inhibits Their Neurotoxicity in Vivo

Brian R. Fluharty; Emiliano Biasini; Matteo Stravalaci; Alessandra Sclip; Luisa Diomede; Claudia Balducci; Pietro La Vitola; Massimo Messa; Laura Colombo; Gianluigi Forloni; Tiziana Borsello; Marco Gobbi; David A. Harris

Background: The cellular prion protein (PrPC) could be a toxicity-transducing receptor for amyloid-β (Aβ) oligomers. Results: N1, a naturally occurring fragment of PrPC, binds Aβ oligomers, inhibits their polymerization into fibrils, and suppresses their neurotoxic effects in vitro and in vivo. Conclusion: N1 binds tightly to Aβ oligomers and blocks their neurotoxicity. Significance: Administration of exogenous N1 or related peptides may represent an effective therapy for Alzheimer disease. A hallmark of Alzheimer disease (AD) is the accumulation of the amyloid-β (Aβ) peptide in the brain. Considerable evidence suggests that soluble Aβ oligomers are responsible for the synaptic dysfunction and cognitive deficit observed in AD. However, the mechanism by which these oligomers exert their neurotoxic effect remains unknown. Recently, it was reported that Aβ oligomers bind to the cellular prion protein with high affinity. Here, we show that N1, the main physiological cleavage fragment of the cellular prion protein, is necessary and sufficient for binding early oligomeric intermediates during Aβ polymerization into amyloid fibrils. The ability of N1 to bind Aβ oligomers is influenced by positively charged residues in two sites (positions 23–31 and 95–105) and is dependent on the length of the sequence between them. Importantly, we also show that N1 strongly suppresses Aβ oligomer toxicity in cultured murine hippocampal neurons, in a Caenorhabditis elegans-based assay, and in vivo in a mouse model of Aβ-induced memory dysfunction. These data suggest that N1, or small peptides derived from it, could be potent inhibitors of Aβ oligomer toxicity and represent an entirely new class of therapeutic agents for AD.


Annals of Neurology | 2009

Recombinant C1 inhibitor in brain ischemic injury.

Raffaella Gesuete; Claudio Storini; Alessandro Fantin; Matteo Stravalaci; Elisa R. Zanier; Franca Orsini; Helene Vietsch; Maurice Mannesse; Bertjan Ziere; Marco Gobbi; Maria Grazia De Simoni

C1 inhibitor (C1‐INH) is an endogenous inhibitor of complement and kinin systems. We have explored the efficacy and the therapeutic window of the recently available human recombinant (rh) C1‐INH on ischemic brain injury and investigated its mechanism of action in comparison with that of plasma‐derived (pd) C1‐INH.


Circulation | 2012

Targeting Mannose-Binding Lectin Confers Long-Lasting Protection With a Surprisingly Wide Therapeutic Window in Cerebral Ischemia

Franca Orsini; Pia Villa; Sara Parrella; Rosalia Zangari; Elisa R. Zanier; Raffaella Gesuete; Matteo Stravalaci; Stefano Fumagalli; Roberta Ottria; José J. Reina; Alessandra Paladini; Edoardo Micotti; Renato Ribeiro-Viana; Javier Rojo; Vasile I. Pavlov; Gregory L. Stahl; Anna Bernardi; Marco Gobbi; Maria Grazia De Simoni

Background— The involvement of the complement system in brain injury has been scarcely investigated. Here, we document the pivotal role of mannose-binding lectin (MBL), one of the recognition molecules of the lectin complement pathway, in brain ischemic injury. Methods and Results— Focal cerebral ischemia was induced in mice (by permanent or transient middle cerebral artery occlusion) and rats (by 3-vessel occlusion). We first observed that MBL is deposited on ischemic vessels up to 48 hours after injury and that functional MBL/MBL-associated serine protease 2 complexes are increased. Next, we demonstrated that (1) MBL−/− mice are protected from both transient and permanent ischemic injury; (2) Polyman2, the newly synthesized mannosylated molecule selected for its binding to MBL, improves neurological deficits and infarct volume when given up to 24 hours after ischemia in mice; (3) anti-MBL-A antibody improves neurological deficits and infarct volume when given up to 18 hours after ischemia, as assessed after 28 days in rats. Conclusions— Our data show an important role for MBL in the pathogenesis of brain ischemic injury and provide a strong support to the concept that MBL inhibition may be a relevant therapeutic target in humans, one with a wide therapeutic window of application.


Journal of Biological Chemistry | 2010

Non-peptidic thrombospondin-1-mimics as fibroblast growth factor-2 inhibitors: an integrated strategy for the development of new antiangiogenic compounds

Giorgio Colombo; Barbara Margosio; Laura Ragona; Marco A. C. Neves; Silvia Bonifacio; Douglas S. Annis; Matteo Stravalaci; Simona Tomaselli; Raffaella Giavazzi; Marco Rusnati; Marco Presta; Lucia Zetta; Deane F. Mosher; Domenico Ribatti; Marco Gobbi; Giulia Taraboletti

Endogenous inhibitors of angiogenesis, such as thrombospondin-1 (TSP-1), are promising sources of therapeutic agents to treat angiogenesis-driven diseases, including cancer. TSP-1 regulates angiogenesis through different mechanisms, including binding and sequestration of the angiogenic factor fibroblast growth factor-2 (FGF-2), through a site located in the calcium binding type III repeats. We hypothesized that the FGF-2 binding sequence of TSP-1 might serve as a template for the development of inhibitors of angiogenesis. Using a peptide array approach followed by binding assays with synthetic peptides and recombinant proteins, we identified a FGF-2 binding sequence of TSP-1 in the 15-mer sequence DDDDDNDKIPDDRDN. Molecular dynamics simulations, taking the full flexibility of the ligand and receptor into account, and nuclear magnetic resonance identified the relevant residues and conformational determinants for the peptide-FGF interaction. This information was translated into a pharmacophore model used to screen the NCI2003 small molecule databases, leading to the identification of three small molecules that bound FGF-2 with affinity in the submicromolar range. The lead compounds inhibited FGF-2-induced endothelial cell proliferation in vitro and affected angiogenesis induced by FGF-2 in the chicken chorioallantoic membrane assay. These small molecules, therefore, represent promising leads for the development of antiangiogenic agents. Altogether, this study demonstrates that new biological insights obtained by integrated multidisciplinary approaches can be used to develop small molecule mimics of endogenous proteins as therapeutic agents.


Journal of Biological Chemistry | 2012

Specific recognition of biologically active amyloid-β oligomers by a new Surface Plasmon Resonance-based immunoassay and an in vivo assay in Caenorhabditis elegans

Matteo Stravalaci; Antonio Bastone; Marten Beeg; Alfredo Cagnotto; Laura Colombo; Giuseppe Di Fede; Fabrizio Tagliavini; Laura Cantù; Elena Del Favero; Michele Mazzanti; Roberto Chiesa; Mario Salmona; Luisa Diomede; Marco Gobbi

Background: Aβ oligomers are major players in Alzheimer disease, but the tools for their detection are not satisfactory. Results: We developed a SPR-based immunoassay and a test in C. elegans, specifically identifying toxic oligomers. Conclusion: These methods allow study of the effects of mutations or drugs on Aβ oligomerization. Significance: The SPR-based immunoassay provides new opportunities for the detection of toxic oligomers in biological samples. Soluble oligomers of the amyloid-β (Aβ) peptide play a key role in the pathogenesis of Alzheimers disease, but their elusive nature makes their detection challenging. Here we describe a novel immunoassay based on surface plasmon resonance (SPR) that specifically recognizes biologically active Aβ oligomers. As a capturing agent, we immobilized on the sensor chip the monoclonal antibody 4G8, which targets a central hydrophobic region of Aβ. This SPR assay allows specific recognition of oligomeric intermediates that rapidly appear and disappear during the incubation of synthetic Aβ1–42, discriminating them from monomers and higher order aggregates. The species recognized by SPR generate ionic currents in artificial lipid bilayers and inhibit the physiological pharyngeal contractions in Caenorhabditis elegans, a new method for testing the toxic potential of Aβ oligomers. With these assays we found that the formation of biologically relevant Aβ oligomers is inhibited by epigallocatechin gallate and increased by the A2V mutation, previously reported to induce early onset dementia. The SPR-based immunoassay provides new opportunities for detection of toxic Aβ oligomers in biological samples and could be adapted to study misfolding proteins in other neurodegenerative disorders.


Analytical Biochemistry | 2011

A modified protocol to prepare seed-free starting solutions of amyloid-β (Aβ)1–40 and Aβ1–42 from the corresponding depsipeptides

Marten Beeg; Matteo Stravalaci; Antonio Bastone; Mario Salmona; Marco Gobbi

Preparing reliable, seed-free stock solutions of the highly amyloidogenic peptides amyloid-β (Aβ) is difficult. Besides the formation of aggregates during synthesis and storage, dissolution of the peptide is a critical step because vortexing can induce aggregation. To overcome this, synthesis of the more water-soluble depsi-Aβ(1-42) peptide, from which the native sequence is easily obtained, has been suggested. We further refined this technique, including a cutoff filtration step and switching the depsipeptide in basic conditions, to stabilize the formed native peptide. The obtained solutions of native Aβ(1-40) and Aβ(1-42) peptides were homogeneous and aggregate free, as indicated by thioflavin T and circular dichroism analysis.


Sensors | 2012

Applications of Surface Plasmon Resonance (SPR) for the Characterization of Nanoparticles Developed for Biomedical Purposes

Mara Canovi; Jacopo Lucchetti; Matteo Stravalaci; Francesca Re; Davide Moscatelli; Paolo Bigini; Mario Salmona; Marco Gobbi

Great interest is currently being devoted to the development of nanoparticles (NPs) for biomedical purposes, designed to improve the pharmacokinetic profile of their cargos (either imaging probes or drugs) and to enhance the specific targeting at the disease site. Recent works suggest that Surface Plasmon Resonance (SPR), widely used for the analysis of biomolecular interactions, represents a technique of choice for rapid and quantitative analyses of the interaction between NPs—functionalized with specific ligands—and their putative biological targets. Moreover, SPR can provide important details on the formation and the role of the protein “corona”, i.e., the protein layer which coats NPs once they come into contact with biological fluids. These novel applications of SPR sensors may be very useful to characterize, screen and develop nanodevices for biomedical purposes.


PLOS ONE | 2009

Immunopurification of Pathological Prion Protein Aggregates

Emiliano Biasini; Susanna Mantovani; Matteo Stravalaci; Marco Gobbi; David A. Harris; Roberto Chiesa

Background Prion diseases are fatal neurodegenerative disorders that can arise sporadically, be genetically inherited or acquired through infection. The key event in these diseases is misfolding of the cellular prion protein (PrPC) into a pathogenic isoform that is rich in β-sheet structure. This conformational change may result in the formation of PrPSc, the prion isoform of PrP, which propagates itself by imprinting its aberrant conformation onto PrPC molecules. A great deal of effort has been devoted to developing protocols for purifying PrPSc for structural studies, and testing its biological properties. Most procedures rely on protease digestion, allowing efficient purification of PrP27-30, the protease-resistant core of PrPSc. However, protease treatment cannot be used to isolate abnormal forms of PrP lacking conventional protease resistance, such as those found in several genetic and atypical sporadic cases. Principal Findings We developed a method for purifying pathological PrP molecules based on sequential centrifugation and immunoprecipitation with a monoclonal antibody selective for aggregated PrP. With this procedure we purified full-length PrPSc and mutant PrP aggregates at electrophoretic homogeneity. PrPSc purified from prion-infected mice was able to seed misfolding of PrPC in a protein misfolding cyclic amplification reaction, and mutant PrP aggregates from transgenic mice were toxic to cultured neurons. Significance The immunopurification protocol described here isolates biologically active forms of aggregated PrP. These preparations may be useful for investigating the structural and chemico-physical properties of infectious and neurotoxic PrP aggregates.

Collaboration


Dive into the Matteo Stravalaci's collaboration.

Top Co-Authors

Avatar

Marco Gobbi

University of Strasbourg

View shared research outputs
Top Co-Authors

Avatar

Mario Salmona

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Marten Beeg

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Luisa Diomede

Australian National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Antonio Bastone

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Franca Orsini

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Alfredo Cagnotto

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge