Network


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

Hotspot


Dive into the research topics where Stefania Preda is active.

Publication


Featured researches published by Stefania Preda.


PLOS ONE | 2012

Dual effect of beta-amyloid on α7 and α4β2 nicotinic receptors controlling the release of glutamate, aspartate and GABA in rat hippocampus.

Elisa Mura; Stefania Zappettini; Stefania Preda; Fabrizio Biundo; Cristina Lanni; Massimo Grilli; Anna Cavallero; Guendalina Olivero; Alessia Salamone; Stefano Govoni; Mario Marchi

Background We previously showed that beta-amyloid (Aβ), a peptide considered as relevant to Alzheimers Disease, is able to act as a neuromodulator affecting neurotransmitter release in absence of evident sign of neurotoxicity in two different rat brain areas. In this paper we focused on the hippocampus, a brain area which is sensitive to Alzheimers Disease pathology, evaluating the effect of Aβ (at different concentrations) on the neurotransmitter release stimulated by the activation of pre-synaptic cholinergic nicotinic receptors (nAChRs, α4β2 and α7 subtypes). Particularly, we focused on some neurotransmitters that are usually involved in learning and memory: glutamate, aspartate and GABA. Methodology/Findings We used a dual approach: in vivo experiments (microdialysis technique on freely moving rats) in parallel to in vitro experiments (isolated nerve endings derived from rat hippocampus). Both in vivo and in vitro the administration of nicotine stimulated an overflow of aspartate, glutamate and GABA. This effect was greatly inhibited by the highest concentrations of Aβ considered (10 µM in vivo and 100 nM in vitro). In vivo administration of 100 nM Aβ (the lowest concentration considered) potentiated the GABA overflow evoked by nicotine. All these effects were specific for Aβ and for nicotinic secretory stimuli. The in vitro administration of either choline or 5-Iodo-A-85380 dihydrochloride (α7 and α4β2 nAChRs selective agonists, respectively) elicited the hippocampal release of aspartate, glutamate, and GABA. High Aβ concentrations (100 nM) inhibited the overflow of all three neurotransmitters evoked by both choline and 5-Iodo-A-85380 dihydrochloride. On the contrary, low Aβ concentrations (1 nM and 100 pM) selectively acted on α7 subtypes potentiating the choline-induced release of both aspartate and glutamate, but not the one of GABA. Conclusions/Significance The results reinforce the concept that Aβ has relevant neuromodulatory effects, which may span from facilitation to inhibition of stimulated release depending upon the concentration used.


Neuropsychopharmacology | 2008

Acute β -Amyloid Administration Disrupts the Cholinergic Control of Dopamine Release in the Nucleus Accumbens

Stefania Preda; Stefano Govoni; Cristina Lanni; Marco Racchi; Elisa Mura; Massimo Grilli; Mario Marchi

The clinical presentation of Alzheimers disease is characterized by memory deficits but it also involves the impairment of several cognitive functions. Some of these cognitive and executive functions are mediated by limbic areas and are regulated by dopaminergic neurotransmission. Furthermore, literature data suggest that β-amyloid (Aβ) can influence synaptic activity in absence of neurotoxicity and in particular can impair cholinergic modulation of other neurotransmitter actions. In the present study, we evaluated whether small concentrations of Aβ could disrupt cholinergic control of dopamine (DA) release in nucleus accumbens using in vivo (brain dialysis) and in vitro (isolated synaptosomes) models. The cholinergic agonist carbachol (CCh) greatly enhanced DA release from dopaminergic nerve endings in nucleus accumbens both in vivo and in vitro. This effect was mainly exerted on muscarinic receptors because it was inhibited by the muscarinic antagonist atropine and it was unaffected by the nicotinic antagonist mecamylamine. Also the nicotinic agonists epibatidine and nicotine evoked a dopaminergic outflow in nucleus accumbens, which, however, was lower. Aβ 1–40 in absence of neurotoxicity fully inhibited the DA release evoked by CCh and only marginally affected the DA release evoked by epibatidine. The PKC inhibitor GF109203X mimicked the effect of Aβ on DA release and, in turn, Aβ impaired PKC activation by CCh. We can suggest that, in nucleus accumbens, Aβ disrupted in vivo and in vitro cholinergic control of DA release by acting on muscarinic transduction machinery.


Current Pharmaceutical Design | 2010

β -Amyloid: A Disease Target or a Synaptic Regulator Affecting Age-Related Neurotransmitter Changes?

Elisa Mura; Cristina Lanni; Stefania Preda; Francesca Pistoia; M. Sara; Marco Racchi; G. Schettini; Mario Marchi; Stefano Govoni

The amyloid cascade hypothesis sustains that beta-amyloid (Abeta) is the main pathogenetic factor of Alzheimers Disease (AD). Although the direct and indirect neurotoxic role of Abeta are unchallenged, recent findings suggest that the peptide may have so far unforeseen physiological roles. In this regard, the observations showing the ability of Abeta to exert synaptic activities in absence of neurotoxicity are very intriguing. In particular, the peptide is able to affect synaptic transmission of different neurotransmitter systems in key brain areas that regulate executive and cognitive functions, an observation that points Abeta as a new neuromodulator. However, in a pathological context, Abeta may drive functional alterations of several neurotransmitter systems in the early phases of the disease, in turn producing subtle cognitive and behavioural disturbances in addition and before the well known neurodegenerative events. On the other hand, advancing age is the most significant risk factor for the development of AD. In fact, during aging increased Abeta levels have been reported. Moreover, several neurotransmitter systems undergo age-related changes in parallel to a decline of cognitive functions. However, the putative neuromodulatory role of Abeta in the context of aging is nowadays unknown. For these reasons, future studies about the spectrum of action of Abeta (brain areas and neurotransmitter systems affected) are particularly interesting since may suggest new therapeutic targets in order to sustain those functions which may be altered during aging.


Neuropsychobiology | 1996

Catecholaminergic, Neuroendocrine and Anxiety Responses to Acute Psychological Stress in Healthy Subjects: Influence of Alprazolam Administration

Giuseppe Santagostino; Guido Amoretti; Pietro Frattini; F. Zerbi; Maria Laura Cucchi; Stefania Preda; Gian Luigi Corona

We studied the effect of alprazolam (APZ) in 12 healthy volunteers on the psychological stress-induced activation of emotion and on the pituitary-adrenal, adrenomedullary and sympathoneuronal systems. After 3 days of placebo or APZ (1 mg/day orally) administration, we examined plasma levels of adrenocorticotropic hormone, cortisol, L-3,4 dihydroxyphenylalanine, dopamine, norepinephrine (NE), epinephrine, metanephrine, normetanephrine, homovanillic acid, vanillylmandelic acid, 3-methoxy-4-hydroxy phenyglycol, urinary levels of cortisol and catecholamines, circulatory responses and state anxiety levels in subjects undergoing psychological stress based on viewing horror, violence, danger and war film clips. Film viewing produced modest rises of state anxiety levels, of plasma NE concentration and of diastolic blood pressure in both the placebo and drug groups. APZ significantly reduced anxiety levels at the beginning of the experimental session and caused a decrease of noradrenergic and dopaminergic neurotransmitter and cortisol concentrations. Our data suggest that APZ reduced anxiety related to the expectation of the event, while the circuitry between structures responsible for anxiety and peripheral sympathoneural function was still found to be partly sensitive to film viewing.


Journal of Alzheimer's Disease | 2010

Specific Neuromodulatory Actions of Amyloid-β on Dopamine Release in Rat Nucleus Accumbens and Caudate Putamen

Elisa Mura; Stefania Preda; Stefano Govoni; Cristina Lanni; Luigia Trabace; Massimo Grilli; Federica Lagomarsino; Anna Pittaluga; Mario Marchi

We previously demonstrated that amyloid-beta (Abeta) has a neuromodulatory action in the nucleus accumbens (NAc). In this area of the brain, the peptide disrupts the cholinergic control of dopamine (DA) release both in vivo and in vitro. The aim of the present work was to extend the research on the neuromodulatory effect of Abeta (1-40) on DA transmission to different release stimuli and to another dopaminergic brain area, the caudate putamen (CPu), in order to clarify whether the effect of the peptide is stimulus- or brain area-selective. We performed both in vivo (microdialysis associated to HPLC) and in vitro studies (synaptosomes in superfusion). Both in NAc and in CPu and both in vivo and in vitro, Abeta did not affect either basal or potassium-stimulated DA release. In CPu, the Abeta ability to impair the DA release evoked by the cholinergic agonist carbachol, observed in NAc, was confirmed only in vitro. Moreover, in vitro Abeta affected a specific component of the DA overflow evoked by the non-selective metabotropic glutamate receptors agonist t-ACPD. Altogether, these results show that Abeta may have different neuromodulatory actions depending upon the secretory stimulus and, in vivo, the brain area investigated.


Frontiers in Pharmacology | 2012

Beta Amyloid Differently Modulate Nicotinic and Muscarinic Receptor Subtypes which Stimulate in vitro and in vivo the Release of Glycine in the Rat Hippocampus

Stefania Zappettini; Massimo Grilli; Guendalina Olivero; Elisa Mura; Stefania Preda; Stefano Govoni; Alessia Salamone; Mario Marchi

Using both in vitro (hippocampal synaptosomes in superfusion) and in vivo (microdialysis) approaches we investigated whether and to what extent β amyloid peptide 1–40 (Aβ 1–40) interferes with the cholinergic modulation of the release of glycine (GLY) in the rat hippocampus. The nicotine-evoked overflow of endogenous GLY in hippocampal synaptosomes in superfusion was significantly inhibited by Aβ 1–40 (10 nM) while increasing the concentration to 100 nM the inhibitory effect did not further increase. Both the Choline (Ch; α7 agonist; 1 mM) and the 5-Iodo-A-85380 dihydrochloride (5IA85380, α4β2 agonist; 10 nM)-evoked GLY overflow were inhibited by Aβ 1–40 at 100 nM but not at 10 nM concentrations. The KCl evoked [3H]GLY and [3H]Acetylcholine (ACh) overflow were strongly inhibited in presence of oxotremorine; however this inhibitory muscarinic effect was not affected by Aβ 1–40. The effects of Aβ 1–40 on the administration of nicotine, veratridine, 5IA85380, and PHA543613 hydrochloride (PHA543613; a selective agonist of α7 subtypes) on hippocampal endogenous GLY release in vivo were also studied. Aβ 1–40 significantly reduced (at 10 μM but not at 1 μM) the nicotine-evoked in vivo release of GLY. Aβ 1–40 (at 10 μM but not at 1 μM) significantly inhibited the PHA543613 (1 mM)-elicited GLY overflow while was ineffective on the GLY overflow evoked by 5IA85380 (1 mM). Aβ 40–1 (10 μM) did not produce any inhibitory effect on nicotine-evoked GLY overflow both in the in vitro and in vivo experiments. Our results indicate that (a) the cholinergic modulation of the release of GLY occurs by the activation of both α7 and α4β2 nicotinic ACh receptors (nAChRs) as well as by the activation of inhibitory muscarinic ACh receptors (mAChRs) and (b) Aβ 1–40 can modulate cholinergic evoked GLY release exclusively through the interaction with α7 and the α4β2 nAChR nicotinic receptors but not through mAChR subtypes.


Current Pharmaceutical Design | 2014

Dangerous Liaisons between Beta-Amyloid and Cholinergic Neurotransmission

Stefano Govoni; Elisa Mura; Stefania Preda; Marco Racchi; Cristina Lanni; Massimo Grilli; Stefania Zappettini; Alessia Salamone; Guendalina Olivero; Anna Pittaluga; Mario Marchi

The review examines the multifaceted interactions between cholinergic transmission and beta-amyloid suggesting a continuum in the action of the peptide that at low concentrations (picomolar-low nanomolar) may directly stimulate nicotinic cholinergic receptor while desensitizing them at increasing concentrations (high nanomolar-low micromolar). In addition high beta-amyloid concentrations may reduce the synaptic release of several neurotransmitters, including glutamate, aspartate, GABA, glycine and dopamine, when the release is elicited through cholinergic stimulation but not following depolarization. The effect of beta-amyloid has been observed both in vitro and in vivo in at least three different brain areas (nucleus accumbens, striatum, hippocampus) suggesting that the peptide may exert some general effects even if not all the brain areas have been evaluated. In turn the activation of cholinergic receptors may affect the amyloid precursor protein processing diverting the metabolism toward non-amyloidogenic products. These actions, dissociated from those described in the case of high beta-amyloid concentrations leading to neurotoxic oligomers, may participate to cause dysfunctions in the neurotransmitter activity, in turn leading, at least from a theoretical point of view, to early neuropsychiatric disturbances in the disease. Complexively these observations underscore novel relationships between two main players in Alzheimers disease pathogenesis that are beta-amyloid and cholinergic transmission. Also emerges the inherent difficulty of targeting beta-amyloid in a context in which the peptide exerts several actions beyond neurotoxicity.


Neuroscience | 2010

Specific inhibitory effect of amyloid-β on presynaptic muscarinic receptor subtypes modulating neurotransmitter release in the rat nucleus accumbens

Massimo Grilli; Federica Lagomarsino; Stefania Zappettini; Stefania Preda; Elisa Mura; Stefano Govoni; Mario Marchi

In this study we investigate on the effect of amyloid-beta1-40 (A beta 1-40) on the oxotremorine (OXO)-induced release of [(3)H] dopamine (DA), [(3)H]GABA and [(3)H]acetylcholine (ACh) from synaptosomes in the rat nucleus accumbens (NAc). OXO in presence of himbacine (HIMBA) was able to increase the basal release of [(3)H]GABA. The OXO-elicited [(3)H]GABA overflow was significantly antagonized by atropine (A; 94%), by the M3 antagonists DAU5884 (96%) and 4-DAMP (70%), and by A beta 1-40 (65%). Exposure of NAc synaptosomes to OXO produced a dose-dependent increase of [(3)H]DA overflow which was antagonized by A, partially inhibited by A beta 1-40 (100 nM) but unaffected by DAU5884 and 4-DAMP. The K(+)-evoked [(3)H]ACh overflow was inhibited by OXO. This effect was counteracted by the M2 antagonist AFDX-116 but not by the selective M4 antagonist mamba toxin 3 (MT3). The K(+)-evoked [(3)H]GABA overflow was also inhibited by OXO but conversely, this effect was counteracted by MT3 and not by AFDX-116. A beta 1-40 (100 nM) did not modify the inhibitory effect of OXO both on the K(+)-evoked [(3)H]ACh and [(3)H]GABA overflow. The results show that in the rat NAc, A beta 1-40 selectively inhibits the function of the muscarinic subtypes which stimulate neurotransmitter release and not those which modulate negatively the stimulated release.


Journal of Bioactive and Compatible Polymers | 2016

Local biological effects of adipose stromal vascular fraction delivery systems after subcutaneous implantation in a murine model

Barbara Vigani; Luca Mastracci; Federica Grillo; Sara Perteghella; Stefania Preda; Barbara Crivelli; Barbara Antonioli; Marta Galuzzi; Marta Cecilia Tosca; Mario Marazzi; Maria Luisa Torre; Theodora Chlapanidas

The aim of this study was to test alginate beads and silk fibroin non-woven mats as stromal vascular fraction delivery systems to support cell implantation for tissue repair and regeneration, through trophic and immunomodulant paracrine signaling. Furthermore, in vivo scaffold biocompatibility was histologically analyzed in a murine model at different time endpoints, with particular focus on construct-induced vascularization and neoangiogenesis. The fibroin mat induced a typical foreign body reaction, recruiting macrophages and giant cells and concurrently promoted neovascularization of the implanted construct. Conversely, alginate beads triggered a more circumscribed, chronic inflammatory reaction, which decreased over time. The combined in vivo implantation of alginate beads and fibroin mat with stromal vascular fraction promoted vascularization and integration of scaffolds into the surrounding subcutaneous environment. The new blood vessel ingrowth should, hopefully, support engineered cell viability and functionality, as well as the transport of soluble bioactive molecules. Due to their neovascularization properties, stromal vascular fraction administration, using alginate or fibroin scaffolds, is a new, promising, cost-effective tissue engineering approach.


Neurochemistry International | 2011

Different presynaptic nicotinic receptor subtypes modulate in vivo and in vitro the release of glycine in the rat hippocampus

Stefania Zappettini; Elisa Mura; Massimo Grilli; Stefania Preda; Alessia Salamone; Guendalina Olivero; Stefano Govoni; Mario Marchi

In the present study, using an in vivo approach (a microdialysis technique associated to HPLC with fluorimetric detection) and in vitro purified hippocampal synaptosomes in superfusion, we investigated the glycinergic transmission in the hippocampus, focusing on the nicotinic control of glycine (GLY) release. The acute administration of nicotine in vivo was able to evoke endogenous GLY release in the rat hippocampus. The specific nicotinic agonists PHA-543613 hydrochloride (PHA543613) selective for the α7 nicotinic receptor subtype administered in vivo also elicited GLY release in a similar extent, while the α4β2 agonist 5-IA85380 dihydrochloride (5IA85380) was less effective. Nicotine elicited GLY overflow also from hippocampal synaptosomes in vitro. This overflow was Ca(2+)-dependent and inhibited by methyllycaconitine (MLA), but was not modified by dihydro-beta-erythroidine (DHβE, 1 μM). Choline(Ch)-evoked GLY overflow was Ca(2+) dependent, unaltered in presence of DHβE and blocked by methyllycaconitine (MLA). Additionally, 5IA85380 elicited a GLY overflow, which in turn was Ca(2+) dependent, was significantly inhibited by DHβE but was unaffected by MLA. The GLY overflow produced by these nicotinic agonists quantitatively resembles that evoked by 9 mM KCl. The effects of a high concentration of 5IA85380 (1mM), in the presence of 2 μM DHβE, on the release of GLY was also studied comparatively to that on glutamate and aspartate release. The nicotinic agonist 5IA85380 tested at high concentration (1mM) was able to produce a stimulatory effect of endogenous release of the three amino acids, even in the presence of 2 μM DHβE, indicating the existence of a DHβE resistant, α4β2 nAChR subtype with a functional role in the modulation of GLY, ASP, and GLU release. Our results show that in the rat hippocampus the release of GLY is, at least in part, of neuronal origin and is modulated by the activation of both α7 and α4β2 (low and high affinity) nAChR subtypes.

Collaboration


Dive into the Stefania Preda'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