Network


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

Hotspot


Dive into the research topics where Laura Colombo is active.

Publication


Featured researches published by Laura Colombo.


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.


Science | 2009

A Recessive Mutation in the APP Gene with Dominant-Negative Effect on Amyloidogenesis

Giuseppe Di Fede; Marcella Catania; Michela Morbin; Giacomina Rossi; Silvia Suardi; Giulia Mazzoleni; Marco Merlin; Anna Rita Giovagnoli; Sara Prioni; Alessandra Erbetta; Chiara Falcone; Marco Gobbi; Laura Colombo; Antonio Bastone; Marten Beeg; Claudia Manzoni; Bruna Francescucci; Alberto Spagnoli; Laura Cantù; Elena Del Favero; Efrat Levy; Mario Salmona; Fabrizio Tagliavini

β-Amyloid precursor protein (APP) mutations cause familial Alzheimers disease with nearly complete penetrance. We found an APP mutation [alanine-673→valine-673 (A673V)] that causes disease only in the homozygous state, whereas heterozygous carriers were unaffected, consistent with a recessive Mendelian trait of inheritance. The A673V mutation affected APP processing, resulting in enhanced β-amyloid (Aβ) production and formation of amyloid fibrils in vitro. Co-incubation of mutated and wild-type peptides conferred instability on Aβ aggregates and inhibited amyloidogenesis and neurotoxicity. The highly amyloidogenic effect of the A673V mutation in the homozygous state and its anti-amyloidogenic effect in the heterozygous state account for the autosomal recessive pattern of inheritance and have implications for genetic screening and the potential treatment of Alzheimers disease.


FEBS Letters | 2001

Anti-amyloidogenic activity of tetracyclines: Studies in vitro

Gianluigi Forloni; Laura Colombo; Laura Girola; Fabrizio Tagliavini; Mario Salmona

Cerebral deposition of β‐amyloid is a major neuropathological feature in Alzheimers disease. Here we show that tetracyclines, tetracycline and doxycycline, classical antibiotics, exhibit anti‐amyloidogenic activity. This capacity was determined by the exposure of β 1‐42 amyloid peptide to the drugs followed by the electron microscopy examination of the amyloid fibrils spontaneously formed and quantified with thioflavine T binding assay. The drugs reduced also the resistance of β 1‐42 amyloid fibrils to trypsin digestion. Tetracyclines not only inhibited the β‐amyloid aggregates formation but also disassembled the pre‐formed fibrils. The results indicate that drugs with a well‐known clinical profile, including activity in the central nervous system, are potentially useful for Alzheimers therapy.


Journal of Virology | 2003

Evaluation of Quinacrine Treatment for Prion Diseases

A. Barret; Fabrizio Tagliavini; Gianluigi Forloni; Clive Bate; Mario Salmona; Laura Colombo; A. De Luigi; Lucia Limido; S. Suardi; Giacomina Rossi; Frédéric Auvré; K. T. Adjou; Nicole Salès; Alun Williams; Corinne Ida Lasmézas; Jean-Philippe Deslys

ABSTRACT Based on in vitro observations in scrapie-infected neuroblastoma cells, quinacrine has recently been proposed as a treatment for Creutzfeldt-Jakob disease (CJD), including a new variant CJD which is linked to contamination of food by the bovine spongiform encephalopathy (BSE) agent. The present study investigated possible mechanisms of action of quinacrine on prions. The ability of quinacrine to interact with and to reduce the protease resistance of PrP peptide aggregates and PrPres of human and animal origin were analyzed, together with its ability to inhibit the in vitro conversion of the normal prion protein (PrPc) to the abnormal form (PrPres). Furthermore, the efficiencies of quinacrine and chlorpromazine, another tricyclic compound, were examined in different in vitro models and in an experimental murine model of BSE. Quinacrine efficiently hampered de novo generation of fibrillogenic prion protein and PrPres accumulation in ScN2a cells. However, it was unable to affect the protease resistance of preexisting PrP fibrils and PrPres from brain homogenates, and a “curing” effect was obtained in ScGT1 cells only after lengthy treatment. In vivo, no detectable effect was observed in the animal model used, consistent with other recent studies and preliminary observations in humans. Despite its ability to cross the blood-brain barrier, the use of quinacrine for the treatment of CJD is questionable, at least as a monotherapy. The multistep experimental approach employed here could be used to test new therapeutic regimes before their use in human trials.


Journal of Neurochemistry | 2009

The SIRT1 activator resveratrol protects SK‐N‐BE cells from oxidative stress and against toxicity caused by α‐synuclein or amyloid‐β (1‐42) peptide

Diego Albani; Letizia Polito; Sara Batelli; Stefania De Mauro; Claudia Fracasso; Giuliana Martelli; Laura Colombo; Claudia Manzoni; Mario Salmona; Silvio Caccia; Alessandro Negro; Gianluigi Forloni

Human sirtuins are a family of seven conserved proteins (SIRT1‐7). The most investigated is the silent mating type information regulation‐2 homolog (SIRT1, NM_012238), which was associated with neuroprotection in models of polyglutamine toxicity or Alzheimer’s disease (AD) and whose activation by the phytocompound resveratrol (RES) has been described. We have examined the neuroprotective role of RES in a cellular model of oxidative stress, a common feature of neurodegeneration. RES prevented toxicity triggered by hydrogen peroxide or 6‐hydroxydopamine (6‐OHDA). This action was likely mediated by SIRT1 activation, as the protection was lost in the presence of the SIRT1 inhibitor sirtinol and when SIRT1 expression was down‐regulated by siRNA approach. RES was also able to protect SK‐N‐BE from the toxicity arising from two aggregation‐prone proteins, the AD‐involved amyloid‐β (1‐42) peptide (Aβ42) and the familiar Parkinson’s disease linked α‐synuclein(A30P) [α‐syn(A30P)]. Alpha‐syn(A30P) toxicity was restored by sirtinol addition, while a partial RES protective effect against Aβ42 was found even in presence of sirtinol, thus suggesting a direct RES effect on Aβ42 fibrils. We conclude that SIRT1 activation by RES can prevent in our neuroblastoma model the deleterious effects triggered by oxidative stress or α‐syn(A30P) aggregation, while RES displayed a SIRT1‐independent protective action against Aβ42.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Tetracyclines affect prion infectivity

Gianluigi Forloni; Selina Iussich; Tazeen Awan; Laura Colombo; Nadia Angeretti; Laura Girola; Ilaria Bertani; Giorgio Poli; Maria Caramelli; Maria Grazia Bruzzone; Laura Farina; Lucia Limido; Giacomina Rossi; Giorgio Giaccone; James Ironside; Orso Bugiani; Mario Salmona; Fabrizio Tagliavini

Prion diseases are transmissible neurodegenerative disorders of humans and animals for which no effective treatment is available. Conformationally altered, protease-resistant forms of the prion protein (PrP) termed PrPSc are critical for disease transmissibility and pathogenesis, thus representing a primary target for therapeutic strategies. Based on previous findings that tetracyclines revert abnormal physicochemical properties and abolish neurotoxicity of PrP peptides in vitro, we tested the ability of these compounds to interact with PrPSc from patients with the new variant of Creutzfeldt–Jakob disease (vCJD) and cattle with bovine spongiform encephalopathy (BSE). The incubation with tetracycline hydrochloride or doxycycline hyclate at concentrations ranging from 10 μM to 1 mM resulted in a dose-dependent decrease in protease resistance of PrPSc. This finding prompted us to investigate whether tetracyclines affect prion infectivity by using an animal model of disease. Syrian hamsters were injected intracerebrally with 263K scrapie-infected brain homogenate that was coincubated with 1 mM tetracycline hydrochloride, 1 mM doxycycline hyclate, or vehicle solution before inoculation. Hamsters injected with tetracycline-treated inoculum showed a significant delay in the onset of clinical signs of disease and prolonged survival time. These effects were paralleled by a delay in the appearance of magnetic-resonance abnormalities in the thalamus, neuropathological changes, and PrPSc accumulation. When tetracycline was preincubated with highly diluted scrapie-infected inoculum, one third of hamsters did not develop disease. Our data suggest that these well characterized antibiotics reduce prion infectivity through a direct interaction with PrPSc and are potentially useful for inactivation of BSE- or vCJD-contaminated products and prevention strategies.


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.


The Journal of Neuroscience | 2011

Calcitonin Gene-Related Peptide-Mediated Enhancement of Purinergic Neuron/Glia Communication by the Algogenic Factor Bradykinin in Mouse Trigeminal Ganglia from Wild-Type and R192Q Cav2.1 Knock-In Mice: Implications for Basic Mechanisms of Migraine Pain

Stefania Ceruti; Giovanni Villa; Marta Fumagalli; Laura Colombo; Giulia Magni; M. Zanardelli; Elsa Fabbretti; Claudia Verderio; Arn M. J. M. van den Maagdenberg; A. Nistri; Maria P. Abbracchio

Within the trigeminal ganglion, crosstalk between neurons and satellite glial cells (SGCs) contributes to neuronal sensitization and transduction of painful stimuli, including migraine pain, at least partly through activation of purinergic receptor mechanisms. We previously showed that the algogenic mediator bradykinin (BK) potentiates purinergic P2Y receptors on SGCs in primary trigeminal cultures. Our present study investigated the molecular basis of this effect in wild-type (WT) mice and CaV2.1 α1 R192Q mutant knock-in (KI) mice expressing a human mutation causing familial hemiplegic migraine type 1. Single-cell calcium imaging of WT cultures revealed functional BK receptors in neurons only, suggesting a paracrine action by BK to release a soluble mediator responsible for its effects on SGCs. We identified this mediator as the neuropeptide calcitonin gene-related peptide (CGRP), whose levels were markedly increased by BK, while the CGRP antagonist CGRP8-37 and the anti-migraine drug sumatriptan inhibited BK actions. Unlike CGRP, BK was ineffective in neuron-free SGC cultures, confirming the CGRP neuronal source. P2Y receptor potentiation induced by CGRP in SGCs was mediated via activation of the extracellular signal-regulated kinase 1/2 pathways, and after exposure to CGRP, a significant release of several cytokines was detected. Interestingly, both basal and BK-stimulated CGRP release was higher in KI mouse cultures, where BK significantly upregulated the number of SGCs showing functional UTP-sensitive P2Y receptors. Our findings suggest that P2Y receptors on glial cells might be considered as novel players in the cellular processes underlying migraine pathophysiology and might represent new targets for the development of innovative therapeutic agents against migraine pain.


PLOS ONE | 2011

The Molecular Assembly of Amyloid Aβ Controls Its Neurotoxicity and Binding to Cellular Proteins

Claudia Manzoni; Laura Colombo; Paolo Bigini; Valentina Diana; Alfredo Cagnotto; Massimo Messa; Monica Lupi; Valentina Bonetto; Mauro Pignataro; Cristina Airoldi; Erika Sironi; Alun Williams; Mario Salmona

Accumulation of β-sheet-rich peptide (Aβ) is strongly associated with Alzheimers disease, characterized by reduction in synapse density, structural alterations of dendritic spines, modification of synaptic protein expression, loss of long-term potentiation and neuronal cell death. Aβ species are potent neurotoxins, however the molecular mechanism responsible for Aβ toxicity is still unknown. Numerous mechanisms of toxicity were proposed, although there is no agreement about their relative importance in disease pathogenesis. Here, the toxicity of Aβ 1–40 and Aβ 1–42 monomers, oligomers or fibrils, was evaluated using the N2a cell line. A structure-function relationship between peptide aggregation state and toxic properties was established. Moreover, we demonstrated that Aβ toxic species cross the plasma membrane, accumulate in cells and bind to a variety of internal proteins, especially on the cytoskeleton and in the endoplasmatic reticulum (ER). Based on these data we suggest that numerous proteins act as Aβ receptors in N2a cells, triggering a multi factorial toxicity.


PLOS ONE | 2008

The Efficacy of Tetracyclines in Peripheral and Intracerebral Prion Infection

Ada De Luigi; Laura Colombo; Luisa Diomede; Raffaella Capobianco; Michela Mangieri; Claudia Miccolo; Lucia Limido; Gianluigi Forloni; Fabrizio Tagliavini; Mario Salmona

We have previously shown that tetracyclines interact with and reverse the protease resistance of pathological prion protein extracted from scrapie-infected animals and patients with all forms of Creutzfeldt-Jakob disease, lowering the prion titre and prolonging survival of cerebrally infected animals. To investigate the effectiveness of these drugs as anti-prion agents Syrian hamsters were inoculated intramuscularly or subcutaneously with 263K scrapie strain at a 10−4 dilution. Tetracyclines were injected intramuscularly or intraperitoneally at the dose of 10 mg/kg. A single intramuscular dose of doxycycline one hour after infection in the same site of inoculation prolonged median survival by 64%. Intraperitoneal doses of tetracyclines every two days for 40 or 44 days increased survival time by 25% (doxycycline), 32% (tetracycline); and 81% (minocycline) after intramuscular infection, and 35% (doxycycline) after subcutaneous infection. To extend the therapeutic potential of tetracyclines, we investigated the efficacy of direct infusion of tetracyclines in advanced infection. Since intracerebroventricular infusion of tetracycline solutions can cause overt acute toxicity in animals, we entrapped the drugs in liposomes. Animals were inoculated intracerebrally with a 10−4 dilution of the 263K scrapie strain. A single intracerebroventricular infusion of 25 µg/ 20 µl of doxycycline or minocycline entrapped in liposomes was administered 60 days after inoculation, when 50% of animals showed initial symptoms of the disease. Median survival increased of 8.1% with doxycycline and 10% with minocycline. These data suggest that tetracyclines might have therapeutic potential for humans.

Collaboration


Dive into the Laura Colombo's collaboration.

Top Co-Authors

Avatar

Mario Salmona

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Gianluigi Forloni

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Fabrizio Tagliavini

Carlo Besta Neurological Institute

View shared research outputs
Top Co-Authors

Avatar

Massimo Messa

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Cristina Airoldi

University of Milano-Bicocca

View shared research outputs
Top Co-Authors

Avatar

E. Mussini

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Michela Morbin

Carlo Besta Neurological Institute

View shared research outputs
Top Co-Authors

Avatar

Alfredo Cagnotto

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Luisa Diomede

Australian National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge