12 research outputs found

    Conséquences de la surexpression des formes solubles de l’APP dans les mécanismes de mémoire : application à la maladie d'Alzheimer

    No full text
    One of the main characteristic of Alzheimer’s Disease (AD) is the intracerebral accumulation of the neurotoxic Amyloid β peptide (Aβ) either as oligomeric or aggregated forms known as the amyloid plaques. This peptide is produced via the Amyloid Precursor Protein (APP) processing following the amyloidogenic pathway, pathological pathway overactivated in AD. Most of the research performed during the last 25 years focused on pathogenic consequences of this dysregulation, deprioritizing the understanding of the APP physiological functions. Nonetheless, numerous studies show that these physiological functions might be mediated via APP soluble forms (APPs). In the physiological APP processing pathway, the non-amyloidogenic pathway, APP is cleaved by the α secretase, releasing the APPsα which display neuroprotective and synaptotrophic properties, essential for brain normal functions. In the context of AD, the amyloidogenic pathway overactivation leads to APPsβ overproduction at the expense of APPsα. Therefore, AD harmful consequences could be due to the decrease of APPsα concentration associated with an increase of APPsβ. My thesis project aimed to characterize mnemonic and functional properties following the overexpression of these two soluble forms of APP and their therapeutic potential in AD. We firstly overexpressed APPsα in hippocampal neurons of APP/PS1ΔE9 mice, animal model of AD, which display cognitive and synaptic deficits. The continual expression of APPsα, mediated via AAV viruses, enabled restoration of spatial memory, long-term potentiation and dendritic spines density in the hippocampus. This phenotypic rescue was accompanied with the decrease of both Aβ levels and amyloid plaques. This might be due to the activation of microglia, cell type able to internalize and degrade Aβ. In a second hand, I studied the involvement of APPsβ in AD, which remains poorly known. Its overexpression in APP/PS1ΔE9 did not induce neither LTP nor spatial memory restoration. However, APPsβ injection lead to the decrease of Aβ levels without reducing amyloid plaques. This default might be due to the lack of microglial activation. In conclusion, my thesis work show that, unlike APPsβ, APPsα overexpression might overcome the AD inevitable evolution by reducing synaptic and memory alterations, typical of AD. These results reinforce a new way of treatment to cope with AD progression. The use of APPsα as therapeutic agent might be an important tool for future AD therapies.Une des principales caractéristiques de la maladie d'Alzheimer (MA) est l'accumulation intracérébrale du peptide neurotoxique Amyloïde β (Aβ) sous forme oligomérique et sous forme agrégée en plaques amyloïdes. Ce peptide est le produit du clivage de l'Amyloid Precursor Protein (APP) selon la voie amyloïdogène, voie pathologique suractivée dans la MA. La majorité des recherches, au cours des 25 dernières années, se sont concentrées sur les conséquences pathologiques de cette dérégulation, mettant au second plan la compréhension des fonctions physiologiques de l’APP. Cependant, de nombreuses études montrent que ses fonctions physiologiques pourraient être médiées par ses formes solubles (APPs). Dans la voie de clivage physiologique, la voie non-amyloïdogène, l’APP est clivé par l’α secrétase pour libérer l’APPsα, peptide disposant de propriétés neuroprotectrices et synaptotrophiques, essentielles au bon fonctionnement cérébral. Dans le contexte de la MA, la suractivation de la voie amyloïdogène va aboutir à la production de l’APPsβ au détriment de celle d’APPsα. Les conséquences fonctionnelles associées à la maladie d’Alzheimer pourraient ainsi être dues à la diminution de la production d’APPsα associée à une augmentation de la production d’APPsβ. Mon projet de thèse porta sur les conséquences mnésiques et fonctionnelles de la surexpression de ces deux formes et à leur potentiel thérapeutique dans la maladie d’Alzheimer. Nous avons tout d’abord surexprimé l’APPsα dans les neurones de l’hippocampe de souris transgéniques APP/PS1ΔE9, modèle de la MA, qui présentent des déficits cognitifs et synaptiques. L’expression continue d’APPsα, à l’aide de vecteurs AAV, permet la restauration des performances mnésiques des souris, de la potentialisation à long terme (LTP) ainsi que du nombre d’épines dendritiques dans l’hippocampe. Ce sauvetage phénotypique s’accompagne de la diminution conjointe des niveaux d’Aβ et des plaques amyloïdes. Ceci serait en partie la conséquence de l’activation de la microglie, type cellulaire ayant la capacité d’internaliser et de dégrader l’Aβ. Mon second axe de recherche a consisté à étudier l’APPsβ dont l’implication dans la MA reste méconnue. Sa surexpression dans le modèle murin APP/PS1ΔE9 n’induit pas de restauration de la LTP ni de la mémoire spatiale. Néanmoins, l’injection d’APPsβ aboutit à la diminution de la concentration en Aβ solubles sans cependant réduire le nombre de plaques amyloïdes. Ce défaut pourrait-être la conséquence de l’absence d’activation microgliale. En résumé, mon travail de thèse montre que, contrairement à l’APPsβ, la surexpression d’APPsα pourrait contrecarrer l’évolution inéluctable de la maladie et en particulier en réduisant l’atteinte synaptique et mnésique caractéristique de la MA. Ces résultats renforcent une nouvelle voie d’action pour lutter contre la progression de la MA. L’utilisation de l’APPsα en tant qu’agent thérapeutique pourrait ainsi s’avérer être un élément important dans l’arsenal clinique de ces prochaines années

    Overexpression of APP soluble forms : physiological roles and application to Alzheimer’s disease

    No full text
    Une des principales caractéristiques de la maladie d'Alzheimer (MA) est l'accumulation intracérébrale du peptide neurotoxique Amyloïde β (Aβ) sous forme oligomérique et sous forme agrégée en plaques amyloïdes. Ce peptide est le produit du clivage de l'Amyloid Precursor Protein (APP) selon la voie amyloïdogène, voie pathologique suractivée dans la MA. La majorité des recherches, au cours des 25 dernières années, se sont concentrées sur les conséquences pathologiques de cette dérégulation, mettant au second plan la compréhension des fonctions physiologiques de l’APP. Cependant, de nombreuses études montrent que ses fonctions physiologiques pourraient être médiées par ses formes solubles (APPs). Dans la voie de clivage physiologique, la voie non-amyloïdogène, l’APP est clivé par l’α secrétase pour libérer l’APPsα, peptide disposant de propriétés neuroprotectrices et synaptotrophiques, essentielles au bon fonctionnement cérébral. Dans le contexte de la MA, la suractivation de la voie amyloïdogène va aboutir à la production de l’APPsβ au détriment de celle d’APPsα. Les conséquences fonctionnelles associées à la maladie d’Alzheimer pourraient ainsi être dues à la diminution de la production d’APPsα associée à une augmentation de la production d’APPsβ. Mon projet de thèse porta sur les conséquences mnésiques et fonctionnelles de la surexpression de ces deux formes et à leur potentiel thérapeutique dans la maladie d’Alzheimer. Nous avons tout d’abord surexprimé l’APPsα dans les neurones de l’hippocampe de souris transgéniques APP/PS1ΔE9, modèle de la MA, qui présentent des déficits cognitifs et synaptiques. L’expression continue d’APPsα, à l’aide de vecteurs AAV, permet la restauration des performances mnésiques des souris, de la potentialisation à long terme (LTP) ainsi que du nombre d’épines dendritiques dans l’hippocampe. Ce sauvetage phénotypique s’accompagne de la diminution conjointe des niveaux d’Aβ et des plaques amyloïdes. Ceci serait en partie la conséquence de l’activation de la microglie, type cellulaire ayant la capacité d’internaliser et de dégrader l’Aβ. Mon second axe de recherche a consisté à étudier l’APPsβ dont l’implication dans la MA reste méconnue. Sa surexpression dans le modèle murin APP/PS1ΔE9 n’induit pas de restauration de la LTP ni de la mémoire spatiale. Néanmoins, l’injection d’APPsβ aboutit à la diminution de la concentration en Aβ solubles sans cependant réduire le nombre de plaques amyloïdes. Ce défaut pourrait-être la conséquence de l’absence d’activation microgliale. En résumé, mon travail de thèse montre que, contrairement à l’APPsβ, la surexpression d’APPsα pourrait contrecarrer l’évolution inéluctable de la maladie et en particulier en réduisant l’atteinte synaptique et mnésique caractéristique de la MA. Ces résultats renforcent une nouvelle voie d’action pour lutter contre la progression de la MA. L’utilisation de l’APPsα en tant qu’agent thérapeutique pourrait ainsi s’avérer être un élément important dans l’arsenal clinique de ces prochaines années.One of the main characteristic of Alzheimer’s Disease (AD) is the intracerebral accumulation of the neurotoxic Amyloid β peptide (Aβ) either as oligomeric or aggregated forms known as the amyloid plaques. This peptide is produced via the Amyloid Precursor Protein (APP) processing following the amyloidogenic pathway, pathological pathway overactivated in AD. Most of the research performed during the last 25 years focused on pathogenic consequences of this dysregulation, deprioritizing the understanding of the APP physiological functions. Nonetheless, numerous studies show that these physiological functions might be mediated via APP soluble forms (APPs). In the physiological APP processing pathway, the non-amyloidogenic pathway, APP is cleaved by the α secretase, releasing the APPsα which display neuroprotective and synaptotrophic properties, essential for brain normal functions. In the context of AD, the amyloidogenic pathway overactivation leads to APPsβ overproduction at the expense of APPsα. Therefore, AD harmful consequences could be due to the decrease of APPsα concentration associated with an increase of APPsβ. My thesis project aimed to characterize mnemonic and functional properties following the overexpression of these two soluble forms of APP and their therapeutic potential in AD. We firstly overexpressed APPsα in hippocampal neurons of APP/PS1ΔE9 mice, animal model of AD, which display cognitive and synaptic deficits. The continual expression of APPsα, mediated via AAV viruses, enabled restoration of spatial memory, long-term potentiation and dendritic spines density in the hippocampus. This phenotypic rescue was accompanied with the decrease of both Aβ levels and amyloid plaques. This might be due to the activation of microglia, cell type able to internalize and degrade Aβ. In a second hand, I studied the involvement of APPsβ in AD, which remains poorly known. Its overexpression in APP/PS1ΔE9 did not induce neither LTP nor spatial memory restoration. However, APPsβ injection lead to the decrease of Aβ levels without reducing amyloid plaques. This default might be due to the lack of microglial activation. In conclusion, my thesis work show that, unlike APPsβ, APPsα overexpression might overcome the AD inevitable evolution by reducing synaptic and memory alterations, typical of AD. These results reinforce a new way of treatment to cope with AD progression. The use of APPsα as therapeutic agent might be an important tool for future AD therapies

    The APP intracellular domain is required for normal synaptic morphology, synaptic plasticity, and hippocampus-dependent behavior

    Get PDF
    The amyloid precursor protein family (APP/APLPs) has essential roles for neuromuscular synapse development and for the formation and plasticity of synapses within the CNS. Despite this, it has remained unclear whether APP mediates its functions primarily as a cell surface adhesion and signaling molecule or via its numerous proteolytic cleavage products. To address these questions, we followed a genetic approach and used APPΔCT15 knockin mice lacking the last 15 amino acids of APP, including the highly conserved YENPTY protein interaction motif. To circumvent functional compensation by the closely related APLP2, these mice were bred to an APLP2-KO background to generate APPΔCT15-DM double mutants. These APPΔCT15-DM mice were partially viable and displayed defects in neuromuscular synapse morphology and function with impairments in the ability to sustain transmitter release that resulted in muscular weakness. In the CNS, we demonstrate pronounced synaptic deficits including impairments in LTP that were associated with deficits in spatial learning and memory. Thus, the APP-CT15 domain provides essential physiological functions, likely via recruitment of specific interactors. Together with the well-established role of APPsα for synaptic plasticity, this shows that multiple domains of APP, including the conserved C-terminus, mediate signals required for normal PNS and CNS physiology. In addition, we demonstrate that lack of the APP-CT15 domain strongly impairs Aβ generation in vivo, establishing the APP C-terminus as a target for Aβ-lowering strategies. SIGNIFICANCE STATEMENT Synaptic dysfunction and cognitive decline are early hallmark features of Alzheimer's disease. Thus, it is essential to elucidate the in vivo function(s) of APP at the synapse. At present, it is unknown whether APP family proteins function as cell surface receptors, or mainly via shedding of their secreted ectodomains, such as neurotrophic APPsα. Here, to dissect APP functional domains, we used APP mutant mice lacking the last 15 amino acids that were crossed onto an APLP2-KO background. These APPΔCT15-DM mice showed defects in neuromuscular morphology and function. Synaptic deficits in the CNS included impairments of synaptic plasticity, spatial learning, and memory. Collectively, this indicates that multiple APP domains, including the C-terminus, are required for normal nervous system function

    Interleukin-2 improves amyloid pathology, synaptic failure and memory in Alzheimer’s disease mice

    Get PDF
    International audienceInterleukin-2 (IL-2)-deficient mice have cytoarchitectural hippocampal modifications and impaired learning and memory ability reminiscent of Alzheimer’s disease. IL-2 stimulates regulatory T cells whose role is to control inflammation. As neuroinflammation contributes to neurodegeneration, we investigated IL-2 in Alzheimer’s disease. Therefore, we investigated IL-2 levels in hippocampal biopsies of patients with Alzheimer’s disease relative to age-matched control individuals. We then treated APP/PS1ΔE9 mice having established Alzheimer’s disease with IL-2 for 5 months using single administration of an AAV-IL-2 vector. We first found decreased IL-2 levels in hippocampal biopsies of patients with Alzheimer’s disease. In mice, IL-2-induced systemic and brain regulatory T cells expansion and activation. In the hippocampus, IL-2 induced astrocytic activation and recruitment of astrocytes around amyloid plaques, decreased amyloid-β42/40 ratio and amyloid plaque load, improved synaptic plasticity and significantly rescued spine density. Of note, this tissue remodelling was associated with recovery of memory deficits, as assessed in the Morris water maze task. Altogether, our data strongly suggest that IL-2 can alleviate Alzheimer’s disease hallmarks in APP/PS1ΔE9 mice with established pathology. Therefore, this should prompt the investigation of low-dose IL-2 in Alzheimer’s disease and other neuroinflammatory/neurodegenerative disorders

    Alzheimer's disease-like APP processing in wild-type mice identifies synaptic defects as initial steps of disease progression.

    Get PDF
    International audienceAlzheimer's disease (AD) is the most frequent form of dementia in the elderly and no effective treatment is currently available. The mechanisms triggering AD onset and progression are still imperfectly dissected. We aimed at deciphering the modifications occurring in vivo during the very early stages of AD, before the development of amyloid deposits, neurofibrillary tangles, neuronal death and inflammation. Most current AD models based on Amyloid Precursor Protein (APP) overproduction beginning from in utero, to rapidly reproduce the histological and behavioral features of the disease within a few months, are not appropriate to study the early steps of AD development. As a means to mimic in vivo amyloid APP processing closer to the human situation in AD, we used an adeno-associated virus (AAV)-based transfer of human mutant APP and Presenilin 1 (PS1) genes to the hippocampi of two-month-old C57Bl/6 J mice to express human APP, without significant overexpression and to specifically induce its amyloid processing. The human APP, βCTF and Aβ42/40 ratio were similar to those in hippocampal tissues from AD patients. Three months after injection the murine Tau protein was hyperphosphorylated and rapid synaptic failure occurred characterized by decreased levels of both PSD-95 and metabolites related to neuromodulation, on proton magnetic resonance spectroscopy ((1)H-MRS). Astrocytic GLT-1 transporter levels were lower and the tonic glutamatergic current was stronger on electrophysiological recordings of CA1 hippocampal region, revealing the overstimulation of extrasynaptic N-methyl D-aspartate receptor (NMDAR) which precedes the loss of long-term potentiation (LTP). These modifications were associated with early behavioral impairments in the Open-field, Y-maze and Morris Mater Maze tasks. Altogether, this demonstrates that an AD-like APP processing, yielding to levels of APP, βCTF and Aβ42/Aβ40 ratio similar to those observed in AD patients, are sufficient to rapidly trigger early steps of the amyloidogenic and Tau pathways in vivo. With this strategy, we identified a sequence of early events likely to account for disease onset and described a model that may facilitate efforts to decipher the factors triggering AD and to evaluate early neuroprotective strategies

    Inhibition of DYRK1A proteolysis modifies its kinase specificity and rescues Alzheimer phenotype in APP/PS1 mice

    Get PDF
    Abstract Recent evidences suggest the involvement of DYRK1A (dual specificity tyrosine phosphorylation-regulated kinase 1 A) in Alzheimer’s disease (AD). Here we showed that DYRK1A undergoes a proteolytic processing in AD patients hippocampus without consequences on its kinase activity. Resulting truncated forms accumulate in astrocytes and exhibit increased affinity towards STAT3ɑ, a regulator of inflammatory process. These findings were confirmed in APP/PS1 mice, an amyloid model of AD, suggesting that this DYRK1A cleavage is a consequence of the amyloid pathology. We identified in vitro the Leucettine L41 as a compound able to prevent DYRK1A proteolysis in both human and mouse protein extracts. We then showed that intraperitoneal injections of L41 in aged APP/PS1 mice inhibit STAT3ɑ phosphorylation and reduce pro-inflammatory cytokines levels (IL1- β, TNF-ɑ and IL-12) associated to an increased microglial recruitment around amyloid plaques and decreased amyloid-β plaque burden. Importantly, L41 treatment improved synaptic plasticity and rescued memory functions in APP/PS1 mice. Collectively, our results suggest that DYRK1A may contribute to AD pathology through its proteolytic process, reducing its kinase specificity. Further evaluation of inhibitors of DYRK1A truncation promises a new therapeutic approach for AD

    Additional file 2: Figure S2. of Alzheimer’s disease-like APP processing in wild-type mice identifies synaptic defects as initial steps of disease progression

    No full text
    AAV-PS1 and AAV-APP mice do not exhibit neuronal defects in terms of PSD-95, GLT-1 and tonic glutamatergic current. C57Bl/6 J mice (all males) were injected at 8 weeks of age either with AAV-CAG-PS1M146L (AAV-PS1 mice) or AAV-CAG-APPSL (AAV-APP mice). Mice were killed three months later for analyses. (A) Western blot of PSD-95 and GLT-1 (n = 3-4 per group). (B) Densitometric analyses of the antibody immunoreactivities shown in panel A. (C) Tonic glutamatergic current recorded at a holding potential of +40 mV by the whole-cell patch-clamping of CA1 pyramidal cells. No significant difference in tonic glutamatergic current intensity was observed between the AAV-PS1 and AAV-APP groups (whole cell patch-clamp of CA1 pyramidal cells, n = 11/group from n = 10 mice per group). (PNG 447 kb

    Additional file 1: Figure S1. of Alzheimer’s disease-like APP processing in wild-type mice identifies synaptic defects as initial steps of disease progression

    No full text
    APP is processed in sporadic AD cases. Human samples were obtained from late-onset AD cases (Braak 6, Thal 5) and age-matched controls (n = 5 per group). The hippocampus was the studied structure. (A) Western blot analysis of NeuN. (B) Densitometric analyses of western blots showing the expression of human APP in the hippocampus of human controls and AD cases (n = 5 per group). Bars represent means ± SEM, and data were normalized with respect to GAPDH. Statistical analyses were performed with Student’s t-test: **p < 0.01. (C) Densitometric analyses of A, showing the expression of NeuN in the hippocampus of human controls and AD cases (n = 5 per group). Bars represent means ± SEM, and data were normalized with respect to GAPDH. Statistical analyses were performed with Student’s t-test: *p < 0.05. (D) Representation of the APP/NeuN ratio following densitometric analyses of the corresponding western blots (n = 5 per group). Note that APP seems to be processed in sporadic AD cases. Bars represent means ± SEM and data were normalized with respect to GAPDH. Statistical analyses were performed with Student’s t-test: *p < 0.05. (PNG 325 kb
    corecore