Network


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

Hotspot


Dive into the research topics where Michela Tantucci is active.

Publication


Featured researches published by Michela Tantucci.


The Journal of Neuroscience | 2011

The distinct role of medium spiny neurons and cholinergic interneurons in the D₂/A₂A receptor interaction in the striatum: implications for Parkinson's disease

Alessandro Tozzi; Massimiliano Di Filippo; Michela Tantucci; Cinzia Costa; Franco Borsini; Veronica Ghiglieri; Carmen Giampà; Francesca Fusco; Barbara Picconi; Paolo Calabresi

A2A adenosine receptor antagonists are currently under investigation as potential therapeutic agents for Parkinsons disease (PD). However, the molecular mechanisms underlying this therapeutic effect is still unclear. A functional antagonism exists between A2A adenosine and D2 dopamine (DA) receptors that are coexpressed in striatal medium spiny neurons (MSNs) of the indirect pathway. Since this interaction could also occur in other neuronal subtypes, we have analyzed the pharmacological modulation of this relationship in murine MSNs of the direct and indirect pathways as well in striatal cholinergic interneurons. Under physiological conditions, endogenous cannabinoids (eCBs) play a major role in the inhibitory effect on striatal glutamatergic transmission exerted by the concomitant activation of D2 DA receptors and blockade of A2A receptors in both D2- and D1-expressing striatal MSNs. In experimental models of PD, the inhibition of striatal glutamatergic activity exerted by D2 receptor activation did not require the concomitant inhibition of A2A receptors, while it was still dependent on the activation of CB1 receptors in both D2- and D1-expressing MSNs. Interestingly, the antagonism of M1 muscarinic receptors blocked the effects of D2/A2A receptor modulation on MSNs. Moreover, in cholinergic interneurons we found coexpression of D2 and A2A receptors and a reduction of the firing frequency exerted by the same pharmacological agents that reduced excitatory transmission in MSNs. This evidence supports the hypothesis that striatal cholinergic interneurons, projecting to virtually all MSN subtypes, are involved in the D2/A2A and endocannabinoid-mediated effects observed on both subpopulations of MSNs in physiological conditions and in experimental PD.


Neuropharmacology | 2008

Plasticity and repair in the post-ischemic brain.

Massimiliano Di Filippo; Alessandro Tozzi; Cinzia Costa; Vincenzo Belcastro; Michela Tantucci; Barbara Picconi; Paolo Calabresi

Stroke is the second commonest cause of death and the principal cause of adult disability in the world. In most cases ischemic injuries have been reported to induce mild to severe permanent deficits. Nevertheless, recovery is often dynamic, reflecting the ability of the injured neuronal networks to adapt. Plastic phenomena occurring in the cerebral cortex and in subcortical structures after ischemic injuries have been documented at the synaptic, cellular, and network level and several findings suggest that they may play a key role during neurorehabilitation in human stroke survivors. In particular, in vitro studies have demonstrated that oxygen and glucose deprivation (in vitro ischemia) exerts long-term effects on the efficacy of synaptic transmission via the induction of a post-ischemic long-term potentiation (i-LTP). i-LTP may deeply influence the plastic reorganization of cortical representational maps occurring after cerebral ischemia, inducing a functional connection of previously non-interacting neurons. On the other hand, there is evidence that i-LTP may exert a detrimental effect in the peri-infarct area, facilitating excitotoxic processes via the sustained, long-term enhancement of glutamate mediated neurotransmission. In the present work we will review the molecular and synaptic mechanisms underlying ischemia-induced synaptic plastic changes taking into account their potential adaptive and/or detrimental effects on the neuronal network in which they occur. Thereafter, we will consider the implications of brain plastic phenomena in the post-stroke recovery phase as well as during the rehabilitative and therapeutic intervention in human subjects.


Behavioural Brain Research | 2009

Short-term and long-term plasticity at corticostriatal synapses: Implications for learning and memory

Massimiliano Di Filippo; Barbara Picconi; Michela Tantucci; Veronica Ghiglieri; Vincenza Bagetta; Carmelo Sgobio; Alessandro Tozzi; Lucilla Parnetti; Paolo Calabresi

The striatum is the major division of the basal ganglia, representing the input station of the circuit and arguably the principal site within the basal ganglia where information processing occurs. Striatal activity is critically involved in motor control and learning. Many parts of the striatum are involved in reward processing and in various forms of learning and memory, such as reward-association learning. Moreover, the striatum appears to be a brain center for habit formation and is likely to be involved in advanced stages of addiction. The critical role played by the striatum in learning and cognitive processes is thought to be based on changes in neuronal activity when specific behavioral tasks are being learned. Accordingly, excitatory corticostriatal synapses onto both striatal projecting spiny neurons and interneurons are able to undergo the main forms of synaptic plasticity, including long-term potentiation, long-term depression, short-term forms of intrinsic plasticity and spike timing-dependent plasticity. These specific forms of neuroplasticity allow the short-term and long-term selection and differential amplification of cortical neural signals modulating the processes of motor and behavioral selection within the basal ganglia neural circuit.


PLOS ONE | 2010

A53T-Alpha-Synuclein Overexpression Impairs Dopamine Signaling and Striatal Synaptic Plasticity in Old Mice

Alexander Kurz; Kay L. Double; Isabel Lastres-Becker; Alessandro Tozzi; Michela Tantucci; Vanessa Bockhart; Michael Bonin; Moisés García-Arencibia; Silke Nuber; Falk Schlaudraff; Birgit Liss; Javier Fernández-Ruiz; Manfred Gerlach; Ullrich Wüllner; Hartmut Lüddens; Paolo Calabresi; Georg Auburger; Suzana Gispert

Background Parkinsons disease (PD), the second most frequent neurodegenerative disorder at old age, can be caused by elevated expression or the A53T missense mutation of the presynaptic protein alpha-synuclein (SNCA). PD is characterized pathologically by the preferential vulnerability of the dopaminergic nigrostriatal projection neurons. Methodology/Principal Findings Here, we used two mouse lines overexpressing human A53T-SNCA and studied striatal dysfunction in the absence of neurodegeneration to understand early disease mechanisms. To characterize the progression, we employed young adult as well as old mice. Analysis of striatal neurotransmitter content demonstrated that dopamine (DA) levels correlated directly with the level of expression of SNCA, an observation also made in SNCA-deficient (knockout, KO) mice. However, the elevated DA levels in the striatum of old A53T-SNCA overexpressing mice may not be transmitted appropriately, in view of three observations. First, a transcriptional downregulation of the extraneural DA degradation enzyme catechol-ortho-methytransferase (COMT) was found. Second, an upregulation of DA receptors was detected by immunoblots and autoradiography. Third, extensive transcriptome studies via microarrays and quantitative real-time RT-PCR (qPCR) of altered transcript levels of the DA-inducible genes Atf2, Cb1, Freq, Homer1 and Pde7b indicated a progressive and genotype-dependent reduction in the postsynaptic DA response. As a functional consequence, long term depression (LTD) was absent in corticostriatal slices from old transgenic mice. Conclusions/Significance Taken together, the dysfunctional neurotransmission and impaired synaptic plasticity seen in the A53T-SNCA overexpressing mice reflect early changes within the basal ganglia prior to frank neurodegeneration. As a model of preclinical stages of PD, such insights may help to develop neuroprotective therapeutic approaches.


Brain | 2012

Mechanisms underlying the impairment of hippocampal long-term potentiation and memory in experimental Parkinson's disease.

Cinzia Costa; Carmelo Sgobio; Sabrina Siliquini; Alessandro Tozzi; Michela Tantucci; Veronica Ghiglieri; Massimiliano Di Filippo; Valentina Pendolino; Matteo Marti; Michele Morari; Maria Grazia Spillantini; Emanuele Claudio Latagliata; Tiziana Pascucci; Stefano Puglisi-Allegra; Fabrizio Gardoni; Monica Di Luca; Barbara Picconi; Paolo Calabresi

Although patients with Parkinsons disease show impairments in cognitive performance even at the early stage of the disease, the synaptic mechanisms underlying cognitive impairment in this pathology are unknown. Hippocampal long-term potentiation represents the major experimental model for the synaptic changes underlying learning and memory and is controlled by endogenous dopamine. We found that hippocampal long-term potentiation is altered in both a neurotoxic and transgenic model of Parkinsons disease and this plastic alteration is associated with an impaired dopaminergic transmission and a decrease of NR2A/NR2B subunit ratio in synaptic N-methyl-d-aspartic acid receptors. Deficits in hippocampal-dependent learning were also found in hemiparkinsonian and mutant animals. Interestingly, the dopamine precursor l-DOPA was able to restore hippocampal synaptic potentiation via D1/D5 receptors and to ameliorate the cognitive deficit in parkinsonian animals suggesting that dopamine-dependent impairment of hippocampal long-term potentiation may contribute to cognitive deficits in patients with Parkinsons disease.


Neurobiology of Disease | 2013

Effects of central and peripheral inflammation on hippocampal synaptic plasticity.

Massimiliano Di Filippo; Davide Chiasserini; Fabrizio Gardoni; Barbara Viviani; Alessandro Tozzi; Carmela Giampà; Cinzia Costa; Michela Tantucci; Elisa Zianni; Mariaserena Boraso; Sabrina Siliquini; Veronica Ghiglieri; Elisa Colcelli; David Baker; Paola Sarchielli; Francesca Fusco; Monica Di Luca; Paolo Calabresi

The central nervous system (CNS) and the immune system are known to be engaged in an intense bidirectional crosstalk. In particular, the immune system has the potential to influence the induction of brain plastic phenomena and neuronal networks functioning. During direct CNS inflammation, as well as during systemic, peripheral, inflammation, the modulation exerted by neuroinflammatory mediators on synaptic plasticity might negatively influence brain neuronal networks functioning. The aim of the present study was to investigate, by using electrophysiological techniques, the ability of hippocampal excitatory synapses to undergo synaptic plasticity during the initial clinical phase of an experimental model of CNS (experimental autoimmune encephalomyelitis, EAE) as well as following a systemic inflammatory trigger. Moreover, we compared the morphologic, synaptic and molecular consequences of central neuroinflammation with those accompanying peripheral inflammation. Hippocampal long-term potentiation (LTP) has been studied by extracellular field potential recordings in the CA1 region. Immunohistochemistry was performed to investigate microglia activation. Western blot and ELISA assays have been performed to assess changes in the subunit composition of the synaptic glutamate NMDA receptor and the concentration of pro-inflammatory cytokines in the hippocampus. Significant microglial activation together with an impairment of CA1 LTP was present in the hippocampus of mice with central as well as peripheral inflammation. Interestingly, exclusively during EAE but not during systemic inflammation, the impairment of hippocampal LTP was paralleled by a selective reduction of the NMDA receptor NR2B subunit levels and a selective increase of interleukin-1β (IL1β) levels. Both central and peripheral inflammation-triggered mechanisms can activate CNS microglia and influence the function of CNS synapses. During direct CNS inflammation these events are accompanied by detectable changes in synaptic glutamate receptors subunit composition and in the levels of the pro-inflammatory cytokine IL1β.


Neuropharmacology | 2007

Interaction of A2A adenosine and D2 dopamine receptors modulates corticostriatal glutamatergic transmission

Alessandro Tozzi; Anne Tscherter; Vincenzo Belcastro; Michela Tantucci; Cinzia Costa; Barbara Picconi; Diego Centonze; Paolo Calabresi; Franco Borsini

Adenosine and dopamine (DA) strongly modulate the neuronal activity in the striatum by pre- and postsynaptic mechanisms. As several behavioral and molecular studies indicate a functional antagonism between A2A adenosine and D2 DA receptors, compounds that are able to block A2A receptors are of particular interest as antiparkinsonian agents. To study the interaction of A2A and D2 receptors in the striatum, we performed intracellular recordings with sharp microelectrodes and whole-cell patch clamp recordings from spiny neurons in rat corticostriatal slices. The amplitude of the evoked excitatory postsynaptic potentials (EPSPs), as well as the frequency and the amplitude of spontaneous excitatory postsynaptic currents (sEPSCs), were affected neither by the A2A receptor antagonists ST1535 and ZM241385, nor by the D2 receptor agonist quinpirole when applied in isolation. However, co-application of quinpirole and ST1535 or ZM241385 significantly reduced the EPSPs amplitude. This inhibitory effect was associated with an increased paired-pulse facilitation suggesting a presynaptic mechanism of action. Accordingly, whole-cell recordings showed that the concomitant activation of D2 receptors and the antagonism of A2A receptors decreased the frequency of sEPSCs without affecting their amplitude. These results suggest that A2A and D2 receptors converge in the control of corticostriatal glutamatergic transmission by exerting an opposite function.


The Journal of Neuroscience | 2008

Electrophysiology and Pharmacology of Striatal Neuronal Dysfunction Induced by Mitochondrial Complex I Inhibition

Cinzia Costa; Vincenzo Belcastro; Alessandro Tozzi; Massimiliano Di Filippo; Michela Tantucci; Sabrina Siliquini; Alessia Autuori; Barbara Picconi; Maria Grazia Spillantini; Ernesto Fedele; Anna Pittaluga; Maurizio Raiteri; Paolo Calabresi

Reduced activity of the mitochondrial respiratory chain and in particular of complex I is implicated not only in the etiology of Parkinsons disease but also in other forms of parkinsonism in which striatal neurodegeneration occurs, such as progressive supranuclear palsy. The pesticide rotenone inhibits mitochondrial complex I and reproduces features of these basal ganglia neurological disorders in animal models. We have characterized the electrophysiological effects of rotenone in the striatum as well as potential neuroprotective strategies to counteract the detrimental effects of this neurotoxin. We found that rotenone causes a dose-dependent and irreversible loss of the corticostriatal field potential amplitude, which was related to the development of a membrane depolarization/inward current in striatal spiny neurons, coupled to an increased release of both excitatory amino acids and dopamine (DA). In particular, we have investigated whether glutamate, DA, and GABA systems might represent possible targets for neuroprotection against rotenone-induced striatal neuronal dysfunction. Interestingly, whereas modulation of glutamatergic transmission was not neuroprotective, blockade of D2-like but not D1-like DA receptors significantly reduced the rotenone-induced effects via a GABA-mediated mechanism. In addition, because antiepileptic drugs (AEDs) modulate multiple transmitter systems, we have analyzed the possible neuroprotective effects of some AEDs against rotenone. We found that carbamazepine, unlike other tested AEDs, exerts a potent neuroprotective action against rotenone-induced striatal neuronal dysfunction. This neuroprotection was observed at therapeutically relevant concentrations requiring endogenous GABA. Differential targeting of GABAergic transmission may represent a possible therapeutic strategy against basal ganglia neurodegenerative disorders involving mitochondrial complex I dysfunction.


Blood | 2013

Reperfusion of cerebral artery thrombosis by the GPIb/vWF blockade with the nanobody ALX-0081 reduces brain infarct size in guinea pigs

Stefania Momi; Michela Tantucci; Maarten Van Roy; Hans Ulrichts; Giovanni Ricci; Paolo Gresele

Thrombolytic therapy is the cornerstone of treatment of acute atherothrombotic ischemic stroke but is associated with brain hemorrhage; antiplatelet therapy has limited efficacy and is still associated with intracranial bleeding. Therefore, new antithrombotic approaches with a better efficacy/safety ratio are required. We have assessed the effect of ALX-0081, a Nanobody against the A1 domain of von Willebrand factor (VWF) that blocks VWF binding to GPIb, of the thrombolytic agent recombinant tissue plasminogen activator (rtPA), and of the GPIIb/IIIa antagonist tirofiban, in a middle cerebral artery (MCA) thrombosis model in guinea pigs. Drugs were administered before, immediately after, or 15 or 60 minutes after the total occlusion of the MCA. ALX-0081 prevented MCA thrombosis and induced reperfusion when given immediately after and 15 minutes after complete occlusion and reduced brain damage without inducing hemorrhage, whereas tirofiban prevented thrombosis but did not induce reperfusion and induced striking brain hemorrhage. rtPA also induced reperfusion when given 60 minutes after occlusion but provoked brain hemorrhage. Skin bleeding time was not modified or was moderately prolonged by ALX-0081, whereas tirofiban and rtPA prolonged it. The inhibition of the GPIb-VWF axis in guinea pigs prevents cerebral artery thrombosis and induces early reperfusion without provoking intracerebral bleeding thus reducing brain infarct area.


Experimental Neurology | 2007

Memantine reduces neuronal dysfunctions triggered by in vitro ischemia and 3-nitropropionic acid

Alessandro Tozzi; Cinzia Costa; M. Di Filippo; Michela Tantucci; Sabrina Siliquini; Vincenzo Belcastro; Lucilla Parnetti; Barbara Picconi; Paolo Calabresi

Memantine, a low-affinity uncompetitive NMDA receptor antagonist, has been widely utilized for the treatment of Alzheimers disease. A possible neuroprotective role of this drug in pathophysiological conditions involving an altered energetic metabolism of the basal ganglia has never been addressed. Thus, we have characterized the electrophysiological effect of memantine on striatal spiny neurons recorded under control conditions and after in vitro ischemia (oxygen and glucose deprivation). Memantine reduced in a dose-dependent manner (EC(50)=5 microM) the irreversible loss of field potential amplitude induced by in vitro ischemia. The neuroprotective effect of memantine against in vitro ischemia was even more potent (EC(50)=3.2 microM) in the absence of external magnesium, a condition enhancing NMDA-mediated glutamatergic transmission. Memantine was also able to block long-term potentiation recorded from spiny neurons following a brief ischemic episode. Moreover, memantine showed protection against irreversible field potential loss induced by 3-nitropropionic acid (3-NP), an inhibitor of the mitochondrial complex II, without influencing toxicity induced by rotenone, a complex I inhibitor. Memantine could represent a potential neuroprotective agent in pathophysiological conditions involving an altered energy metabolism of basal ganglia.

Collaboration


Dive into the Michela Tantucci'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