Network


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

Hotspot


Dive into the research topics where Luciano D’Adamio is active.

Publication


Featured researches published by Luciano D’Adamio.


Neuron | 2005

Presenilin-Dependent Transcriptional Control of the Aβ-Degrading Enzyme Neprilysin by Intracellular Domains of βAPP and APLP

Raphaelle Pardossi-Piquard; Agnès Petit; T. Kawarai; Claire Sunyach; Cristine Alves da Costa; Bruno Vincent; Sabine Ring; Luciano D’Adamio; Jie Shen; Ulrike Müller; Peter St. George Hyslop; Frédéric Checler

Amyloid beta-peptide (Abeta), which plays a central role in Alzheimers disease, is generated by presenilin-dependent gamma-secretase cleavage of beta-amyloid precursor protein (betaAPP). We report that the presenilins (PS1 and PS2) also regulate Abeta degradation. Presenilin-deficient cells fail to degrade Abeta and have drastic reductions in the transcription, expression, and activity of neprilysin, a key Abeta-degrading enzyme. Neprilysin activity and expression are also lowered by gamma-secretase inhibitors and by PS1/PS2 deficiency in mouse brain. Neprilysin activity is restored by transient expression of PS1 or PS2 and by expression of the amyloid intracellular domain (AICD), which is cogenerated with Abeta, during gamma-secretase cleavage of betaAPP. Neprilysin gene promoters are transactivated by AICDs from APP-like proteins (APP, APLP1, and APLP2), but not by Abeta or by the gamma-secretase cleavage products of Notch, N- or E- cadherins. The presenilin-dependent regulation of neprilysin, mediated by AICDs, provides a physiological means to modulate Abeta levels with varying levels of gamma-secretase activity.


Scientific Reports | 2016

Extracellular Tau Oligomers Produce An Immediate Impairment of LTP and Memory

Mauro Fa; Daniela Puzzo; Roberto Piacentini; Agnieszka Staniszewski; Hong Zhang; María Antonia Baltrons; D. D. Li Puma; Ishita Chatterjee; Jing-Cheng Li; Faisal Saeed; Henry L. Berman; Cristian Ripoli; Walter Gulisano; Juana Gonzalez; H. Tian; J. A. Costa; P. Lopez; Eliot J. Davidowitz; Wai Haung Yu; V. Haroutunian; L. M. Brown; Agostino Palmeri; Einar M. Sigurdsson; Karen Duff; Andrew F. Teich; Lawrence S. Honig; M. Sierks; James G. Moe; Luciano D’Adamio; Claudio Grassi

Non-fibrillar soluble oligomeric forms of amyloid-β peptide (oAβ) and tau proteins are likely to play a major role in Alzheimer’s disease (AD). The prevailing hypothesis on the disease etiopathogenesis is that oAβ initiates tau pathology that slowly spreads throughout the medial temporal cortex and neocortices independently of Aβ, eventually leading to memory loss. Here we show that a brief exposure to extracellular recombinant human tau oligomers (oTau), but not monomers, produces an impairment of long-term potentiation (LTP) and memory, independent of the presence of high oAβ levels. The impairment is immediate as it raises as soon as 20 min after exposure to the oligomers. These effects are reproduced either by oTau extracted from AD human specimens, or naturally produced in mice overexpressing human tau. Finally, we found that oTau could also act in combination with oAβ to produce these effects, as sub-toxic doses of the two peptides combined lead to LTP and memory impairment. These findings provide a novel view of the effects of tau and Aβ on memory loss, offering new therapeutic opportunities in the therapy of AD and other neurodegenerative diseases associated with Aβ and tau pathology.


Molecular Neurodegeneration | 2012

Inhibition of γ-secretase worsens memory deficits in a genetically congruous mouse model of Danish dementia

Robert Tamayev; Luciano D’Adamio

BackgroundA mutation in the BRI2/ITM2b gene causes familial Danish dementia (FDD). BRI2 is an inhibitor of amyloid-β precursor protein (APP) processing, which is genetically linked to Alzheimer’s disease (AD) pathogenesis. The FDD mutation leads to a loss of BRI2 protein and to increased APP processing. APP haplodeficiency and inhibition of APP cleavage by β-secretase rescue synaptic/memory deficits of a genetically congruous mouse model of FDD (FDDKI). β-cleavage of APP yields the β-carboxyl-terminal (β-CTF) and the amino-terminal-soluble APPβ (sAPPβ) fragments. γ-secretase processing of β-CTF generates Aβ, which is considered the main cause of AD. However, inhibiting Aβ production did not rescue the deficits of FDDKI mice, suggesting that sAPPβ/β-CTF, and not Aβ, are the toxic species causing memory loss.ResultsHere, we have further analyzed the effect of γ-secretase inhibition. We show that treatment with a γ-secretase inhibitor (GSI) results in a worsening of the memory deficits of FDDKI mice. This deleterious effect on memory correlates with increased levels of the β/α-CTFs APP fragments in synaptic fractions isolated from hippocampi of FDDKI mice, which is consistent with inhibition of γ-secretase activity.ConclusionThis harmful effect of the GSI is in sharp contrast with a pathogenic role for Aβ, and suggests that the worsening of memory deficits may be due to accumulation of synaptic-toxic β/α-CTFs caused by GSI treatment. However, γ-secretase cleaves more than 40 proteins; thus, the noxious effect of GSI on memory may be dependent on inhibition of cleavage of one or more of these other γ-secretase substrates. These two possibilities do not need to be mutually exclusive. Our results are consistent with the outcome of a clinical trial with the GSI Semagacestat, which caused a worsening of cognition, and advise against targeting γ-secretase in the therapy of AD. Overall, the data also indicate that FDDKI is a valuable mouse model to study AD pathogenesis and predict the clinical outcome of therapeutic agents for AD.


Brain Structure & Function | 2010

Modeling familial British and Danish dementia

Holly J. Garringer; Jill R. Murrell; Luciano D’Adamio; Bernardino Ghetti; Ruben Vidal

Familial British dementia (FBD) and familial Danish dementia (FDD) are two autosomal dominant neurodegenerative diseases caused by mutations in the BRI2 gene. FBD and FDD are characterized by widespread cerebral amyloid angiopathy (CAA), parenchymal amyloid deposition, and neurofibrillary tangles. Transgenic mice expressing wild-type and mutant forms of the BRI2 protein, Bri2 knock-in mutant mice, and Bri2 gene knock-out mice have been developed. Transgenic mice expressing a human FDD-mutated form of the BRI2 gene have partially reproduced the neuropathological lesions observed in FDD. These mice develop extensive CAA, parenchymal amyloid deposition, and neuroinflammation in the central nervous system. These animal models allow the study of the molecular mechanism(s) underlying the neuronal dysfunction in these diseases and allow the development of potential therapeutic approaches for these and related neurodegenerative conditions. In this review, a comprehensive account of the advances in the development of animal models for FBD and FDD and of their relevance to the study of Alzheimer disease is presented.


eLife | 2015

APP and APLP2 interact with the synaptic release machinery and facilitate transmitter release at hippocampal synapses

Tomas Fanutza; Dolores Del Prete; Michael J Ford; Pablo E. Castillo; Luciano D’Adamio

The amyloid precursor protein (APP), whose mutations cause familial Alzheimer’s disease, interacts with the synaptic release machinery, suggesting a role in neurotransmission. Here we mapped this interaction to the NH2-terminal region of the APP intracellular domain. A peptide encompassing this binding domain -named JCasp- is naturally produced by a γ-secretase/caspase double-cut of APP. JCasp interferes with the APP-presynaptic proteins interaction and, if linked to a cell-penetrating peptide, reduces glutamate release in acute hippocampal slices from wild-type but not APP deficient mice, indicating that JCasp inhibits APP function.The APP-like protein-2 (APLP2) also binds the synaptic release machinery. Deletion of APP and APLP2 produces synaptic deficits similar to those caused by JCasp. Our data support the notion that APP and APLP2 facilitate transmitter release, likely through the interaction with the neurotransmitter release machinery. Given the link of APP to Alzheimer’s disease, alterations of this synaptic role of APP could contribute to dementia. DOI: http://dx.doi.org/10.7554/eLife.09743.001


PLOS ONE | 2013

An intracellular threonine of amyloid-β precursor protein mediates synaptic plasticity deficits and memory loss.

Franco Lombino; Fabrizio Biundo; Robert Tamayev; Ottavio Arancio; Luciano D’Adamio

Mutations in Amyloid-ß Precursor Protein (APP) and BRI2/ITM2b genes cause Familial Alzheimer and Danish Dementias (FAD/FDD), respectively. APP processing by BACE1, which is inhibited by BRI2, yields sAPPß and ß-CTF. ß-CTF is cleaved by gamma-secretase to produce Aß. A knock-in mouse model of FDD, called FDDKI, shows deficits in memory and synaptic plasticity, which can be attributed to sAPPß/ß-CTF but not Aß. We have investigated further the pathogenic function of ß-CTF focusing on Thr668 of ß-CTF because phosphorylation of Thr668 is increased in AD cases. We created a knock-in mouse bearing a Thr668Ala mutation (APPTA mice) that prevents phosphorylation at this site. This mutation prevents the development of memory and synaptic plasticity deficits in FDDKI mice. These data are consistent with a role for the carboxyl-terminal APP domain in the pathogenesis of dementia and suggest that averting the noxious role of Thr668 is a viable therapeutic strategy for human dementias.


Molecular Neurodegeneration | 2012

Caspase-9 mediates synaptic plasticity and memory deficits of Danish dementia knock-in mice: caspase-9 inhibition provides therapeutic protection

Robert Tamayev; Nsikan Akpan; Ottavio Arancio; Carol M. Troy; Luciano D’Adamio

BackgroundMutations in either Aβ Precursor protein (APP) or genes that regulate APP processing, such as BRI2/ITM2B and PSEN1/PSEN2, cause familial dementias. Although dementias due to APP/PSEN1/PSEN2 mutations are classified as familial Alzheimer disease (FAD) and those due to mutations in BRI2/ITM2B as British and Danish dementias (FBD, FDD), data suggest that these diseases have a common pathogenesis involving toxic APP metabolites. It was previously shown that FAD mutations in APP and PSENs promote activation of caspases leading to the hypothesis that aberrant caspase activation could participate in AD pathogenesis.ResultsHere, we tested whether a similar mechanism applies to the Danish BRI2/ITM2B mutation. We have generated a genetically congruous mouse model of FDD, called FDDKI, which presents memory and synaptic plasticity deficits. We found that caspase-9 is activated in hippocampal synaptic fractions of FDDKI mice and inhibition of caspase-9 activity rescues both synaptic plasticity and memory deficits.ConclusionThese data directly implicate caspase-9 in the pathogenesis of Danish dementia and suggest that reducing caspase-9 activity is a valid therapeutic approach to treating human dementias.


Methods of Molecular Biology | 2007

Functional Cloning of Genes Regulating Apoptosis in Neuronal Cells

Roberta Visconti; Luciano D’Adamio

Here, we describe the use of a functional cloning approach, based on the screen of a genome-wide short hairpin RNA (shRNA) library, to identify novel genes regulating apoptosis in neuronal cells. Apoptosis is induced by doxorubicin and is detected with a fluorometric caspase 3 assay. Moreover, we describe also the screen of the library to identify genes regulating the processing f the beta-amyloid (Abeta) precursor protein (APP), the protein associated with the pathogenesis of lzheimers disease. The levels of the peptide Abeta, produced by the APP processing, are detected with ELISA based on the innovative Delfia method developed by PerkinElmer.


Neurobiology of Learning and Memory | 2015

Interaction of ApoE3 and ApoE4 isoforms with an ITM2b/BRI2 mutation linked to the Alzheimer disease-like Danish dementia: Effects on learning and memory

Fabrizio Biundo; Keita Ishiwari; Dolores Del Prete; Luciano D’Adamio

Mutations in Amyloid β Precursor Protein (APP) and in genes that regulate APP processing--such as PSEN1/2 and ITM2b/BRI2--cause familial dementia, such Familial Alzheimer disease (FAD), Familial Danish (FDD) and British (FBD) dementias. The ApoE gene is the major genetic risk factor for sporadic AD. Three major variants of ApoE exist in humans (ApoE2, ApoE3, and ApoE4), with the ApoE4 allele being strongly associated with AD. ITM2b/BRI2 is also a candidate regulatory node genes predicted to mediate the common patterns of gene expression shared by healthy ApoE4 carriers and late-onset AD patients not carrying ApoE4. This evidence provides a direct link between ITM2b/BRI2 and ApoE4. To test whether ApoE4 and pathogenic ITM2b/BRI2 interact to modulate learning and memory, we crossed a mouse carrying the ITM2b/BRI2 mutations that causes FDD knocked-in the endogenous mouse Itm2b/Bri2 gene (FDDKI mice) with human ApoE3 and ApoE4 targeted replacement mice. The resultant ApoE3, FDDKI/ApoE3, ApoE4, FDDKI/ApoE4 male mice were assessed longitudinally for learning and memory at 4, 6, 12, and 16-17 months of age. The results showed that ApoE4-carrying mice displayed spatial working/short-term memory deficits relative to ApoE3-carrying mice starting in early middle age, while long-term spatial memory of ApoE4 mice was not adversely affected even at 16-17 months, and that the FDD mutation impaired working/short-term spatial memory in ApoE3-carrying mice and produced impaired long-term spatial memory in ApoE4-carrying mice in middle age. The present results suggest that the FDD mutation may differentially affect learning and memory in ApoE4 carriers and non-carriers.


Molecular Neurodegeneration | 2012

β - but not γ-secretase proteolysis of APP causes synaptic and memory deficits in a mouse model of dementia

Robert Tamayev; Shuji Matsuda; Luciano D’Adamio

Background Amyloid deposition of Ab peptide characterizes AD. Ab derives from sequential cleavage of APP by band g-secretases. Interestingly, mutations in either APP or the g-secretase genes PSEN1/2 cause familial AD (FAD). Mutation of BRI2/ITM2b causes an AD-like familial dementia (FDD) with amyloid deposits. The FDD plaques contain Ab and ADan, which derives from processing of mutant BRI2 protein by pro-protein convertases. Since amyloidogenic peptides are believed to cause dementias, transgenic mice carrying mutant APP, PSEN1/2 or BRI2/ITM2b are used to model these dementias, as over-expression is necessary to reproduce amyloidosis. However, over-expression of mutant genes might produce harmful effects unrelated to dementias and lead to erroneous information concerning pathogenesis and therapy of human diseases. The clinical failures of compounds efficacious in transgenic models support this hypothesis.

Collaboration


Dive into the Luciano D’Adamio's collaboration.

Top Co-Authors

Avatar

Robert Tamayev

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Fabrizio Biundo

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dolores Del Prete

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shuji Matsuda

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Agnès Petit

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Claire Sunyach

Centre national de la recherche scientifique

View shared research outputs
Researchain Logo
Decentralizing Knowledge