Network


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

Hotspot


Dive into the research topics where Gianluca Amato is active.

Publication


Featured researches published by Gianluca Amato.


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

Dissecting the involvement of tropomyosin-related kinase A and p75 neurotrophin receptor signaling in NGF deficit-induced neurodegeneration

Simona Capsoni; Cecilia Tiveron; Domenico Vignone; Gianluca Amato; Antonino Cattaneo

NGF, the principal neurotrophic factor for basal forebrain cholinergic neurons (BFCNs), has been correlated to Alzheimers disease (AD) because of the selective vulnerability of BFCNs in AD. These correlative links do not substantiate a comprehensive cause–effect mechanism connecting NGF deficit to overall AD neurodegeneration. A demonstration that neutralizing NGF activity could have consequences beyond a direct interference with the cholinergic system came from studies in the AD11 mouse model, in which the expression of a highly specific anti-NGF antibody determines a neurodegeneration that encompasses several features of human AD. Because the transgenic antibody binds to mature NGF much more strongly than to proNGF and prevents binding of mature NGF to the tropomyosin-related kinase A (TrkA) receptor and to p75 neurotrophin receptor (p75NTR), we postulated that neurodegeneration in AD11 mice is provoked by an imbalance of proNGF/NGF signaling and, consequently, of TrkA/p75NTR signaling. To test this hypothesis, in this study we characterize the phenotype of two lines of transgenic mice, one in which TrkA signaling is inhibited by neutralizing anti-TrkA antibodies and a second one in which anti-NGF mice were crossed to p75NTRexonIII(−/−) mice to abrogate p75NTR signaling. TrkA neutralization determines a strong cholinergic deficit and the appearance of β-amyloid peptide (Aβ) but no tau-related pathology. In contrast, abrogating p75NTR signaling determines a full rescue of the cholinergic and Aβ phenotype of anti-NGF mice, but tau hyperphosphorylation is exacerbated. Thus, we demonstrate that inhibiting TrkA signaling activates Aβ accumulation and that different streams of AD neurodegeneration are related in complex ways to TrkA versus p75NTR signaling.


The Journal of Neuroscience | 2010

Transgenic Mice with Chronic NGF Deprivation and Alzheimer's Disease-Like Pathology Display Hippocampal Region-Specific Impairments in Short- and Long-Term Plasticities

Gry Houeland; Armando Romani; Cristina Marchetti; Gianluca Amato; Simona Capsoni; Antonino Cattaneo; Hélène Marie

The etiology of Alzheimers disease (AD) remains elusive. The “amyloid” hypothesis states that toxic action of accumulated β-amyloid peptide (Aβ) on synaptic function causes AD cognitive decline. This hypothesis is supported by analysis of familial AD (FAD)-based transgenic mouse models, where altered amyloid precursor protein (APP) processing leads to Aβ accumulation correlating with hippocampal-dependent memory deficits. Some studies report prominent dentate gyrus (DG) glutamatergic plasticity alterations in these mice, while CA1 plasticity remains relatively unaffected. The “neurotrophic unbalance” hypothesis, on the other hand, states that AD-related loss of cholinergic signaling and altered APP processing are due to alterations in nerve growth factor (NGF) trophic support. This hypothesis is supported by analysis of the AD11 mouse, which exhibits chronic NGF deprivation during adulthood and displays AD-like pathology, including Aβ accumulation and hippocampal-dependent memory deficits. In this study, we analyzed CA1 and DG glutamatergic plasticity in AD11 mice to evaluate whether these mice also share with FAD models a common phenotype in hippocampal synaptic dysfunction. We report that AD11 mice display age-dependent short- and long-term DG plasticity deficits, while CA1 plasticity remains relatively spared. We also report that both structures exhibit enhanced glutamatergic transmission under lower, yet physiological, neurotransmitter release conditions, a defect that should be considered when further evaluating hippocampal synaptic deficits underlying AD pathology. We conclude that severe deficits in DG plasticity represent another common denominator between these two etiologically different types of AD mouse models, independent of the initial insult (overexpression of FAD mutation or NGF deprivation).


Journal of Alzheimer's Disease | 2009

Delivery of NGF to the brain: intranasal versus ocular administration in anti-NGF transgenic mice.

Simona Capsoni; Sonia Covaceuszach; Gabriele Ugolini; Francesca Spirito; Domenico Vignone; Barbara Stefanini; Gianluca Amato; Antonino Cattaneo

Nerve growth factor (NGF) has a great potential for the treatment of Alzheimers disease. However, the therapeutic administration of NGF represents a significant challenge, due to the difficulty to deliver relevant doses to the brain, in a safe and non-invasive way. We previously demonstrated the efficacy of a non-invasive delivery of NGF to the brain in animal models, by an intranasal route. Recently, topical eye application of NGF was proposed, as an option for the delivery of NGF to the brain. Here, we compare the efficacy of the two delivery routes of hNGF-61, a recombinant traceable form of human NGF, in the mouse neurodegeneration model AD11. The intranasal administration appeared to be significantly more effective than the ocular one, in rescuing the neurodegenerative phenotypic hallmarks in AD11 mice. The ocular administration of hNGF-61 showed a more limited efficacy, even at higher doses. Thus, NGF nasal drops represent a viable and effective option to successfully deliver therapeutic NGF to the brain in a non-invasive manner.


PLOS ONE | 2012

Intranasal “painless” Human Nerve Growth Factors Slows Amyloid Neurodegeneration and Prevents Memory Deficits in App X PS1 Mice

Simona Capsoni; Sara Marinelli; Marcello Ceci; Domenico Vignone; Gianluca Amato; Francesca Malerba; Francesca Paoletti; Giovanni Meli; Alessandro Viegi; Flaminia Pavone; Antonino Cattaneo

Nerve Growth Factor (NGF) is being considered as a therapeutic candidate for Alzheimers disease (AD) treatment but the clinical application is hindered by its potent pro-nociceptive activity. Thus, to reduce systemic exposure that would induce pain, in recent clinical studies NGF was administered through an invasive intracerebral gene-therapy approach. Our group demonstrated the feasibility of a non-invasive intranasal delivery of NGF in a mouse model of neurodegeneration. NGF therapeutic window could be further increased if its nociceptive effects could be avoided altogether. In this study we exploit forms of NGF, mutated at residue R100, inspired by the human genetic disease HSAN V (Hereditary Sensory Autonomic Neuropathy Type V), which would allow increasing the dose of NGF without triggering pain. We show that “painless” hNGF displays full neurotrophic and anti-amyloidogenic activities in neuronal cultures, and a reduced nociceptive activity in vivo. When administered intranasally to APPxPS1 mice ( n = 8), hNGFP61S/R100E prevents the progress of neurodegeneration and of behavioral deficits. These results demonstrate the in vivo neuroprotective and anti-amyloidogenic properties of hNGFR100 mutants and provide a rational basis for the development of “painless” hNGF variants as a new generation of therapeutics for neurodegenerative diseases.


Cell Death & Differentiation | 2013

ProNGF\NGF imbalance triggers learning and memory deficits, neurodegeneration and spontaneous epileptic-like discharges in transgenic mice

C Tiveron; Luisa Fasulo; S Capsoni; Francesca Malerba; Silvia Marinelli; Francesca Paoletti; Sonia Piccinin; Raffaella Scardigli; Gianluca Amato; Rossella Brandi; P Capelli; S D'Aguanno; Fulvio Florenzano; F La Regina; A Lecci; A Manca; Giovanni Meli; L Pistillo; Nicola Berretta; Robert Nisticò; Flaminia Pavone; Antonino Cattaneo

ProNGF, the precursor of mature nerve growth factor (NGF), is the most abundant form of NGF in the brain. ProNGF and mature NGF differ significantly in their receptor interaction properties and in their bioactivity. ProNGF increases markedly in the cortex of Alzheimer’s disease (AD) brains and proNGF\NGF imbalance has been postulated to play a role in neurodegeneration. However, a direct proof for a causal link between increased proNGF and AD neurodegeneration is lacking. In order to evaluate the consequences of increased levels of proNGF in the postnatal brain, transgenic mice expressing a furin cleavage-resistant form of proNGF, under the control of the neuron-specific mouse Thy1.2 promoter, were derived and characterized. Different transgenic lines displayed a phenotypic gradient of neurodegenerative severity features. We focused the analysis on the two lines TgproNGF#3 and TgproNGF#72, which shared learning and memory impairments in behavioral tests, cholinergic deficit and increased Aβ-peptide immunoreactivity. In addition, TgproNGF#3 mice developed Aβ oligomer immunoreactivity, as well as late diffuse astrocytosis. Both TgproNGF lines also display electrophysiological alterations related to spontaneous epileptic-like events. The results provide direct evidence that alterations in the proNGF/NGF balance in the adult brain can be an upstream driver of neurodegeneration, contributing to a circular loop linking alterations of proNGF/NGF equilibrium to excitatory/inhibitory synaptic imbalance and amyloid precursor protein (APP) dysmetabolism.


Journal of Alzheimer's Disease | 2010

Peripheral neutralization of nerve growth factor induces immunosympathectomy and central neurodegeneration in transgenic mice.

Simona Capsoni; Cecilia Tiveron; Gianluca Amato; Domenico Vignone; Antonino Cattaneo

We previously showed that anti-nerve growth factor (NGF) antibodies expressed in transgenic mice (AD11) elicit a progressive neurodegeneration, comprising the triad of Alzheimers disease (AD) hallmarks: cholinergic loss, tau hyperphosphorylation, and amyloid-beta peptide formation. However, since anti-NGF antibodies are produced both in the brain and in peripheral tissues of AD11 mice, the contribution of peripheral neutralization of NGF to the onset of brain neurodegeneration was still unexplored. To address this question, we characterized a line of transgenic mice (AD10) in which anti-NGF antibodies are obligatorily produced only in lymphocytes, being initially found in blood. In AD10 mice, peripheral NGF neutralization elicits shrinkage of superior cervical ganglia (immunosympathectomy) and, as a consequence of this, peripheral anti-NGF antibodies cross the blood brain barrier (BBB) and reach the brain, generating an NGF-dependent neurodegeneration, largely superimposable to that observed in AD11 mice. This demonstrates that peripherally originated anti-NGF antibodies can generate a neurodegeneration in the central nervous system of an animal model. Consistently, peripherally-delivered NGF is effective in preventing the onset of the central cholinergic deficit. These findings could have a direct relevance for some human sporadic AD cases, highlighting the role of the BBB disruption and suggesting a causally relevant role of circulating antibodies in AD pathology.


Biochemical and Biophysical Research Communications | 2013

Dissecting the role of sortilin receptor signaling in neurodegeneration induced by NGF deprivation

Simona Capsoni; Gianluca Amato; Domenico Vignone; Chiara Criscuolo; Anders Nykjaer; Antonino Cattaneo

Highlights ► We hypothesize that neurodegeneration in anti-NGF is due to NGF/proNGF imbalance. ► We propose that the proNGF co- receptor sortilin contributes to the neurodegeneration. ► We analyzed anti-NGF mice crossed to sortilin knockout mice. ► Sortilin loss partially protected AD10 anti-NGF mice from neurodegeneration.


Journal of Alzheimer's Disease | 2012

SorLA deficiency dissects amyloid pathology from tau and cholinergic neurodegeneration in a mouse model of Alzheimer's disease.

Simona Capsoni; Anne-Sophie Carlo; Domenico Vignone; Gianluca Amato; Chiara Criscuolo; Thomas E. Willnow; Antonino Cattaneo

Sortilin-related receptor with A-type repeats (SorLA, also known as LR11) has been implicated in Alzheimers disease (AD). Thus, genetic studies associated SorLA gene variants with the risk of sporadic AD. Also, in vitro and in vivo studies showed that SorLA impairs processing of the amyloid-β protein precursor (AβPP) to amyloid-β. In particular, it has been found that loss of SorLA accelerates senile plaque deposition in mouse models overexpressing mutant forms of human AβPP and presenilin 1. Here we tested the possibility that SorLA deficiency also interferes with behavioral and neuropathological endpoints in an alternative murine AD model, the AD10 anti-nerve growth factor (NGF) mouse, in which amyloid-β accumulation derives from the altered processing of endogenous AβPP. In addition to alterations in AβPP processing, AD10 mice also show cholinergic deficit and tau hyperphosphorylation resulting in behavioral deficits in learning and memory paradigms. We found that the loss of SorLA not only exacerbates early amyloid pathology but, at the same time, protects from cholinergic deficit and from early phospho-tau mislocalization. The results show that in the AD10 anti-NGF mouse model the AβPP processing-related aspects of neurodegeneration can be dissociated from those related to tau posttranslational processing and to cholinergic phenotypic maintenance by modulation of SorLA expression. We suggest that SorLA regulates different aspects of neurodegeneration in a complex way, supporting the hypothesis that SorLA expression might be critical not only for amyloid-related pathology but also for other cellular processes altered in AD.


Alzheimers & Dementia | 2008

P2-424: Delivery of NGF to the brain: Intranasal versus ocular administration in transgenic mice

Simona Capsoni; Sonia Covaceuszach; Gabriele Ugolini; Francesca Spirito; Domenico Vignone; Barbara Stefanini; Gianluca Amato; Antonino Cattaneo

Background: The concept of therapeutic administration of human recombinant Nerve Growth Factor (NGF) in AD patients has been well validated by preclinical and clinical studies. However, a significant challenge to the clinical use of NGF is the difficulty associated with its delivery to the brain at therapeutically relevant doses. Indeed, NGF is not orally available and unable to access the blood-brain barrier, following parenteral systemic administration. Clinical studies performed so far were based on intracerebroventricular administration or on a gene therapy based on grafting in the brain parenchyma of cells engineered to secrete NGF. Although encouraging in terms of efficacy, these results can be questioned in terms of widespread clinical applicability, due to the invasiveness of the surgical procedure. A non invasive approach for NGF delivery to the brain would be highly desirable. Our laboratory already obtained a proof of concept for the feasibility of a non invasive strategy to delivery NGF to the brain, by treating intranasally a model of Alzheimer’s disease (the AD11 anti-NGF mouse) and obtaining in such a way the rescue of behavioral deficits and neurodegeneration (Capsoni et al 2002; De Rosa et al, 2005). Recently, it was shown that topical eye application of NGF causes an enhanced expression ChAT immunoreactivity in rat basal forebrain cholinergic neurons, suggesting that ocular NGF application might be a viable option for the effective delivery of NGF to the brain (Lambiase et al., 2006). Here, we compare the efficacy of intranasal versus ocular administration of NGF in transgenic AD11 mice. Methods: By using a new analog of human NGF (hNGF-61), characterized by a single amino acid mutation that allows it to be detected against hNGF background, we compared the efficacy of the two methods of administration in rescuing behavioral deficits and neurodegeneration in AD11 mice. Results: Although the ocular administration of hNGF-61 showed a certain extent of efficacy, the intranasal administration appeared to be significantly more effective in rescuing the whole set of phenotypic hallmarks in AD11 mice, even at lower doses. Conclusions: Thus, NGF nasal drops can represent a non-invasive strategy to successfully deliver NGF to the brain.


Biochemical and Biophysical Research Communications | 2010

In vitro receptor binding properties of a "painless" NGF mutein, linked to hereditary sensory autonomic neuropathy type V.

Sonia Covaceuszach; Simona Capsoni; Sara Marinelli; Flaminia Pavone; Marcello Ceci; Gabriele Ugolini; Domenico Vignone; Gianluca Amato; Francesca Paoletti; Doriano Lamba; Antonino Cattaneo

Collaboration


Dive into the Gianluca Amato's collaboration.

Top Co-Authors

Avatar

Antonino Cattaneo

International School for Advanced Studies

View shared research outputs
Top Co-Authors

Avatar

Simona Capsoni

International School for Advanced Studies

View shared research outputs
Top Co-Authors

Avatar

Francesca Paoletti

International School for Advanced Studies

View shared research outputs
Top Co-Authors

Avatar

Flaminia Pavone

National Research Council

View shared research outputs
Top Co-Authors

Avatar

Francesca Malerba

Scuola Normale Superiore di Pisa

View shared research outputs
Top Co-Authors

Avatar

Giovanni Meli

International School for Advanced Studies

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sonia Covaceuszach

International School for Advanced Studies

View shared research outputs
Top Co-Authors

Avatar

Sara Marinelli

National Research Council

View shared research outputs
Researchain Logo
Decentralizing Knowledge