38 results on '"Pardossi-Piquard R"'
Search Results
2. Neprilysin activity and expression are controlled by nicastrin
- Author
-
Pardossi-Piquard, R., Dunys, J., Yu, G., George-Hyslop, P. St., da Costa, C. Alves, and Checler, F.
- Published
- 2006
3. Does Intraneuronal Accumulation of Carboxyl-terminal Fragments of the Amyloid Precursor Protein Trigger Early Neurotoxicity in Alzheimer’s Disease?
- Author
-
Lauritzen, I., primary, Pardossi-Piquard, R., additional, Bourgeois, A., additional, Bécot, A., additional, and Checler, F., additional
- Published
- 2019
- Full Text
- View/download PDF
4. Influence of Genetic Background on Apathy-Like Behavior in Triple Transgenic AD Mice
- Author
-
Pardossi-Piquard, R., primary, Lauritzen, I., additional, Bauer, C., additional, Sacco, G., additional, Robert, P., additional, and Checler, F., additional
- Published
- 2016
- Full Text
- View/download PDF
5. A novel presenilin 2 mutation (V393M) in early-onset dementia with profound language impairment
- Author
-
Lindquist, Susanne Granhøj, Hasholt, L., Bahl, J.M.C., Heegaard, N.H.H., Andersen, B.B., Nørremølle, Anne, Stokholm, J., Schwartz, M., Batbayli, M., Laursen, Henning, Pardossi-Piquard, R., Chen, F., George-Hyslop, P. St, Waldemar, Gunhild, Nielsen, J.E., Lindquist, Susanne Granhøj, Hasholt, L., Bahl, J.M.C., Heegaard, N.H.H., Andersen, B.B., Nørremølle, Anne, Stokholm, J., Schwartz, M., Batbayli, M., Laursen, Henning, Pardossi-Piquard, R., Chen, F., George-Hyslop, P. St, Waldemar, Gunhild, and Nielsen, J.E.
- Abstract
Udgivelsesdato: 2008/10
- Published
- 2008
6. P1-9 Etude de la production du fragment C-terminal du précurseur de la protéine beta amyloïde (AICD) à partir des substrats C99 et C83
- Author
-
Flammang, B., primary, Pardossi-Piquard, R., additional, and Checler, F., additional
- Published
- 2009
- Full Text
- View/download PDF
7. Presenilin-Dependent -Secretase-Mediated Control of p53-Associated Cell Death in Alzheimer's Disease
- Author
-
Alves da Costa, C., primary, Sunyach, C., additional, Pardossi-Piquard, R., additional, Sevalle, J., additional, Vincent, B., additional, Boyer, N., additional, Kawarai, T., additional, Girardot, N., additional, St. George-Hyslop, P., additional, and Checler, F., additional
- Published
- 2006
- Full Text
- View/download PDF
8. C2-2 Les présénilines contrôlent la production et la dégradation du peptide amyloïde
- Author
-
Pardossi-Piquard, R., primary, Alves da Costa, C., additional, Sunyach, C., additional, Vincent, B., additional, and Checler, F., additional
- Published
- 2005
- Full Text
- View/download PDF
9. [gamma]-Secretase-Mediated Regulation of Neprilysin: Influence of Cell Density and Aging and Modulation by Imatinib.
- Author
-
Bauer C, Pardossi-Piquard R, Dunys J, Roy M, and Checler F
- Published
- 2011
10. Suppression of MT5-MMP Reveals Early Modulation of Alzheimer's Pathogenic Events in Primary Neuronal Cultures of 5xFAD Mice.
- Author
-
Pilat D, Paumier JM, Louis L, Manrique C, García-González L, Stephan D, Bernard A, Pardossi-Piquard R, Checler F, Khrestchatisky M, Di Pasquale E, Baranger K, and Rivera S
- Subjects
- Animals, Mice, Matrix Metalloproteinases, Membrane-Associated metabolism, Matrix Metalloproteinases, Membrane-Associated genetics, Mice, Transgenic, Cells, Cultured, Amyloid beta-Protein Precursor metabolism, Amyloid beta-Protein Precursor genetics, Amyloid beta-Peptides metabolism, Disease Models, Animal, Mice, Knockout, Alzheimer Disease metabolism, Alzheimer Disease genetics, Alzheimer Disease pathology, Neurons metabolism, Neurons pathology, Interleukin-1beta metabolism
- Abstract
We previously reported that membrane-type 5-matrix metalloproteinase (MT5-MMP) deficiency not only reduces pathological hallmarks of Alzheimer's disease (AD) in 5xFAD (Tg) mice in vivo but also impairs interleukin-1 beta (IL-1β)-mediated neuroinflammation and Aβ production in primary Tg immature neural cell cultures after 11 days in vitro. We now investigate the effect of MT5-MMP on incipient pathogenic pathways that are activated in cortical primary cultures at 21-24 days in vitro (DIV), during which time neurons are organized into a functional mature network. Using wild-type (WT), MT5-MMP
-/- (MT5-/- ), 5xFAD (Tg), and 5xFADxMT5-MMP-/- (TgMT5-/- ) mice, we generated primary neuronal cultures that were exposed to IL-1β and/or different proteolytic system inhibitors. We assessed neuroinflammation, APP metabolism, synaptic integrity, and electrophysiological properties using biochemical, imaging and whole-cell patch-clamp approaches. The absence of MT5-MMP impaired the IL-1β-mediated induction of inflammatory genes in TgMT5-/- cells compared to Tg cells. Furthermore, the reduced density of dendritic spines in Tg neurons was also prevented in TgMT5-/- neurons. IL-1β caused a strong decrease in the dendritic spine density of WT neurons, which was prevented in MT5-/- neurons. However, the latter exhibited fewer spines than the WT under untreated conditions. The spontaneous rhythmic firing frequency of the network was increased in MT5-/- neurons, but not in TgMT5-/- neurons, and IL-1β increased this parameter only in Tg neurons. In terms of induced somatic excitability, Tg and TgMT5-/- neurons exhibited lower excitability than WT and MT5-/- , while IL-1β impaired excitability only in non-AD backgrounds. The synaptic strength of miniature global synaptic currents was equivalent in all genotypes but increased dramatically in WT and MT5-/- neurons after IL-1β. MT5-MMP deficiency decreased endogenous and overexpressed C83 and C99 levels but did not affect Aβ levels. C99 appears to be cleared by several pathways, including γ-secretase, the autophagolysosomal system, and also α-secretase, via its conversion to C83. In summary, this study confirms that MT5-MMP is a pivotal factor affecting not only neuroinflammation and APP metabolism but also synaptogenesis and synaptic activity at early stages of the pathology, and reinforces the relevance of targeting MT5-MMP to fight AD.- Published
- 2024
- Full Text
- View/download PDF
11. Ectodomain shedding of PLA2R1 is mediated by the metalloproteases ADAM10 and ADAM17.
- Author
-
Dolla G, Nicolas S, Dos Santos LR, Bourgeois A, Pardossi-Piquard R, Bihl F, Zaghrini C, Justino J, Payré C, Mansuelle P, Garbers C, Ronco P, Checler F, Lambeau G, and Petit-Paitel A
- Subjects
- Humans, Animals, Mice, HEK293 Cells, Podocytes metabolism, Proteolysis, Protein Domains, Ionomycin pharmacology, ADAM10 Protein metabolism, ADAM10 Protein genetics, ADAM17 Protein metabolism, ADAM17 Protein genetics, Amyloid Precursor Protein Secretases metabolism, Amyloid Precursor Protein Secretases genetics, Membrane Proteins metabolism, Membrane Proteins genetics, Receptors, Phospholipase A2 metabolism, Receptors, Phospholipase A2 genetics
- Abstract
Phospholipase A2 receptor 1 (PLA2R1) is a 180-kDa transmembrane protein that plays a role in inflammation and cancer and is the major autoantigen in membranous nephropathy, a rare but severe autoimmune kidney disease. A soluble form of PLA2R1 has been detected in mouse and human serum. It is likely produced by proteolytic shedding of membrane-bound PLA2R1 but the mechanism is unknown. Here, we show that human PLA2R1 is cleaved by A Disintegrin And Metalloprotease 10 (ADAM10) and ADAM17 in HEK293 cells, mouse embryonic fibroblasts, and human podocytes. By combining site-directed mutagenesis and sequencing, we determined the exact cleavage site within the extracellular juxtamembrane stalk of human PLA2R1. Orthologs and paralogs of PLA2R1 are also shed. By using pharmacological inhibitors and genetic approaches with RNA interference and knock-out cellular models, we identified a major role of ADAM10 in the constitutive shedding of PLA2R1 and a dual role of ADAM10 and ADAM17 in the stimulated shedding. We did not observe evidence for cleavage by β- or γ-secretase, suggesting that PLA2R1 may not be a substrate for regulated intramembrane proteolysis. PLA2R1 shedding occurs constitutively and can be triggered by the calcium ionophore ionomycin, the protein kinase C activator PMA, cytokines, and lipopolysaccharides, in vitro and in vivo. Altogether, our results show that PLA2R1 is a novel substrate for ADAM10 and ADAM17, producing a soluble form that is increased in inflammatory conditions and likely exerts various functions in physiological and pathophysiological conditions including inflammation, cancer, and membranous nephropathy., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
12. The amyloid precursor protein and its derived fragments concomitantly contribute to the alterations of mitochondrial transport machinery in Alzheimer's disease.
- Author
-
Vaillant-Beuchot L, Eysert F, Duval B, Kinoshita PF, Pardossi-Piquard R, Bauer C, Eddarkaoui S, Buée L, Checler F, and Chami M
- Subjects
- Animals, Humans, Mice, Mice, Transgenic, Neurons metabolism, Amyloid beta-Peptides metabolism, Mitochondrial Proteins metabolism, Mitochondrial Proteins genetics, Amyloid Precursor Protein Secretases metabolism, Kinesins metabolism, Biological Transport, Mitophagy, Nerve Tissue Proteins, rho GTP-Binding Proteins, Intracellular Signaling Peptides and Proteins, Alzheimer Disease metabolism, Alzheimer Disease pathology, Alzheimer Disease genetics, Amyloid beta-Protein Precursor metabolism, Amyloid beta-Protein Precursor genetics, Mitochondria metabolism
- Abstract
Mitochondria dysfunctions and mitophagy failure have been associated with several Alzheimer's disease (AD) related molecular actors including amyloid beta (Aβ) and recently the amyloid precursor protein-C terminal fragments (APP-CTFs). The efficacy of the mitophagy process in neurons relies on regulated mitochondrial transport along axons involving a complex molecular machinery. The contribution of the amyloid precursor protein (APP) and its derived fragments to the mitochondrial transport machinery alterations in AD have not been investigated before. We report herein a change of the expression of mitochondrial transport proteins (SNPH and Miro1), motor adapters (TRANK1 and TRAK2), and components of the dynein and kinesin motors (i.e., IC1,2 and Kif5 (A, B, C) isoforms) by endogenous APP and by overexpression of APP carrying the familial Swedish mutation (APPswe). We show that APP-CTFs and Aβ concomitantly regulate the expression of a set of transport proteins as demonstrated in APPswe cells treated with β- and γ-secretase inhibitors and in cells Knock-down for presenilin 1 and 2. We further report the impact of APP-CTFs on the expression of transport proteins in AAV-injected C99 mice brains. Our data also indicate that both Aβ oligomers (Aβo) and APP-CTFs impair the colocalization of mitochondria and transport proteins. This has been demonstrated in differentiated SH-SY5Y naive cells treated with Aβo and in differentiated SH-SY5Y and murine primary neurons expressing APPswe and treated with the γ-secretase inhibitor. Importantly, we uncover that the expression of a set of transport proteins is modulated in a disease-dependent manner in 3xTgAD mice and in human sporadic AD brains. This study highlights molecular mechanisms underlying mitochondrial transport defects in AD that likely contribute to mitophagy failure and disease progression., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
13. Correction to: Intraneuronal aggregation of the β‑CTF fragment of APP (C99) induces Aβ‑independent lysosomal-autophagic pathology.
- Author
-
Lauritzen I, Pardossi-Piquard R, Bourgeois A, Pagnotta S, Biferi MG, Barkats M, Lacor P, Klein W, Bauer C, and Checler F
- Published
- 2023
- Full Text
- View/download PDF
14. The η-secretase-derived APP fragment ηCTF is localized in Golgi, endosomes and extracellular vesicles and contributes to Aβ production.
- Author
-
Afram E, Lauritzen I, Bourgeois A, El Manaa W, Duplan E, Chami M, Valverde A, Charlotte B, Pardossi-Piquard R, and Checler F
- Subjects
- Mice, Animals, Amyloid beta-Protein Precursor genetics, Amyloid beta-Protein Precursor metabolism, Amyloid Precursor Protein Secretases genetics, Amyloid Precursor Protein Secretases metabolism, Plaque, Amyloid, Mice, Transgenic, Endosomes metabolism, Amyloid beta-Peptides metabolism, Alzheimer Disease metabolism, Extracellular Vesicles metabolism
- Abstract
The processing of the amyloid precursor protein (APP) is one of the key events contributing to Alzheimer's disease (AD) etiology. Canonical cleavages by β- and γ-secretases lead to Aβ production which accumulate in amyloid plaques. Recently, the matrix metalloprotease MT5-MMP, referred to as η-secretase, has been identified as a novel APP cleaving enzyme producing a transmembrane fragment, ηCTF that undergoes subsequent cleavages by α- and β-secretases yielding the Aηα and Aηβ peptides, respectively. The functions and contributions of ηCTF and its related fragments to AD pathology are poorly understood. In this study, we designed a novel immunological probe referred to as ηCTF-NTer antibody that specifically interacts with the N-terminal part of ηCTF targeting ηCTF, Aηα, Aηβ but not C99, C83 and Aβ. We examined the fate and localization of ηCTF fragment in various cell models and in mice. We found that overexpressed ηCTF undergoes degradation in the proteasomal and autophagic pathways and accumulates mainly in the Golgi and in endosomes. Moreover, we observed the presence of ηCTF in small extracellular vesicles purified from neuroblastoma cells or from mouse brains expressing ηCTF. Importantly, the expression of ηCTF in fibroblasts devoid on APP leads to Aβ production demonstrating its contribution to the amyloidogenic pathway. Finally, we observed an ηCTF-like immunoreactivity around amyloid plaques and an age-dependent accumulation of ηCTF in the triple-transgenic mouse AD model. Thus, our study suggests that the ηCTF fragment likely contributes to AD pathology by its exosomal spreading and involvement in Aβ production., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
15. Is γ-secretase a beneficial inactivating enzyme of the toxic APP C-terminal fragment C99?
- Author
-
Checler F, Afram E, Pardossi-Piquard R, and Lauritzen I
- Subjects
- Alzheimer Disease metabolism, Amyloid beta-Peptides chemistry, Animals, Humans, Amyloid Precursor Protein Secretases metabolism, Amyloid beta-Peptides metabolism
- Abstract
Genetic, biochemical, and anatomical grounds led to the proposal of the amyloid cascade hypothesis centered on the accumulation of amyloid beta peptides (Aβ) to explain Alzheimer's disease (AD) etiology. In this context, a bulk of efforts have aimed at developing therapeutic strategies seeking to reduce Aβ levels, either by blocking its production (γ- and β-secretase inhibitors) or by neutralizing it once formed (Aβ-directed immunotherapies). However, so far the vast majority of, if not all, clinical trials based on these strategies have failed, since they have not been able to restore cognitive function in AD patients, and even in many cases, they have worsened the clinical picture. We here propose that AD could be more complex than a simple Aβ-linked pathology and discuss the possibility that a way to reconcile undoubted genetic evidences linking processing of APP to AD and a consistent failure of Aβ-based clinical trials could be to envision the pathological contribution of the direct precursor of Aβ, the β-secretase-derived C-terminal fragment of APP, βCTF, also referred to as C99. In this review, we summarize scientific evidences pointing to C99 as an early contributor to AD and postulate that γ-secretase should be considered as not only an Aβ-generating protease, but also a beneficial C99-inactivating enzyme. In that sense, we discuss the limitations of molecules targeting γ-secretase and propose alternative strategies seeking to reduce C99 levels by other means and notably by enhancing its lysosomal degradation., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
16. Accumulation of amyloid precursor protein C-terminal fragments triggers mitochondrial structure, function, and mitophagy defects in Alzheimer's disease models and human brains.
- Author
-
Vaillant-Beuchot L, Mary A, Pardossi-Piquard R, Bourgeois A, Lauritzen I, Eysert F, Kinoshita PF, Cazareth J, Badot C, Fragaki K, Bussiere R, Martin C, Mary R, Bauer C, Pagnotta S, Paquis-Flucklinger V, Buée-Scherrer V, Buée L, Lacas-Gervais S, Checler F, and Chami M
- Subjects
- Aged, Aged, 80 and over, Alzheimer Disease metabolism, Amyloid Precursor Protein Secretases antagonists & inhibitors, Amyloid Precursor Protein Secretases metabolism, Animals, Aspartic Acid Endopeptidases antagonists & inhibitors, Aspartic Acid Endopeptidases metabolism, Autopsy, Cell Line, Female, Humans, Membrane Potential, Mitochondrial, Mice, Mitochondria metabolism, Peptide Fragments genetics, Peptide Fragments metabolism, Reactive Oxygen Species metabolism, Alzheimer Disease pathology, Amyloid beta-Protein Precursor genetics, Amyloid beta-Protein Precursor metabolism, Brain pathology, Mitochondria pathology, Mitochondria ultrastructure, Mitophagy genetics
- Abstract
Several lines of recent evidence indicate that the amyloid precursor protein-derived C-terminal fragments (APP-CTFs) could correspond to an etiological trigger of Alzheimer's disease (AD) pathology. Altered mitochondrial homeostasis is considered an early event in AD development. However, the specific contribution of APP-CTFs to mitochondrial structure, function, and mitophagy defects remains to be established. Here, we demonstrate in neuroblastoma SH-SY5Y cells expressing either APP Swedish mutations, or the β-secretase-derived APP-CTF fragment (C99) combined with β- and γ-secretase inhibition, that APP-CTFs accumulation independently of Aβ triggers excessive mitochondrial morphology alteration (i.e., size alteration and cristae disorganization) associated with enhanced mitochondrial reactive oxygen species production. APP-CTFs accumulation also elicit basal mitophagy failure illustrated by enhanced conversion of LC3, accumulation of LC3-I and/or LC3-II, non-degradation of SQSTM1/p62, inconsistent Parkin and PINK1 recruitment to mitochondria, enhanced levels of membrane and matrix mitochondrial proteins, and deficient fusion of mitochondria with lysosomes. We confirm the contribution of APP-CTFs accumulation to morphological mitochondria alteration and impaired basal mitophagy in vivo in young 3xTgAD transgenic mice treated with γ-secretase inhibitor as well as in adeno-associated-virus-C99 injected mice. Comparison of aged 2xTgAD and 3xTgAD mice indicates that, besides APP-CTFs, an additional contribution of Aβ to late-stage mitophagy activation occurs. Importantly, we report on mitochondrial accumulation of APP-CTFs in human post-mortem sporadic AD brains correlating with mitophagy failure molecular signature. Since defective mitochondria homeostasis plays a pivotal role in AD pathogenesis, targeting mitochondrial dysfunctions and/or mitophagy by counteracting early APP-CTFs accumulation may represent relevant therapeutic interventions in AD.
- Published
- 2021
- Full Text
- View/download PDF
17. The Transcription Factor EB Reduces the Intraneuronal Accumulation of the Beta-Secretase-Derived APP Fragment C99 in Cellular and Mouse Alzheimer’s Disease Models.
- Author
-
Bécot A, Pardossi-Piquard R, Bourgeois A, Duplan E, Xiao Q, Diwan A, Lee JM, Lauritzen I, and Checler F
- Subjects
- Animals, Autophagy genetics, Cell Line, Disease Models, Animal, Humans, Lysosomes metabolism, Mice, Transgenic, Stereotaxic Techniques, Alzheimer Disease metabolism, Amyloid Precursor Protein Secretases metabolism, Amyloid beta-Peptides metabolism, Aspartic Acid Endopeptidases metabolism, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors metabolism, Neurons metabolism
- Abstract
: Brains that are affected by Alzheimer's disease (AD) are characterized by the overload of extracellular amyloid β (Aβ) peptides, but recent data from cellular and animal models propose that Aβ deposition is preceded by intraneuronal accumulation of the direct precursor of Aβ, C99. These studies indicate that C99 accumulation firstly occurs within endosomal and lysosomal compartments and that it contributes to early-stage AD-related endosomal-lysosomal-autophagic defects. Our previous work also suggests that C99 accumulation itself could be a consequence of defective lysosomal-autophagic degradation. Thus, in the present study, we analyzed the influence of the overexpression of the transcription factor EB (TFEB), a master regulator of autophagy and lysosome biogenesis, on C99 accumulation occurring in both AD cellular models and in the triple-transgenic mouse model (3xTgAD). In the in vivo experiments, TFEB overexpression was induced via adeno-associated viruses (AAVs), which were injected either into the cerebral ventricles of newborn mice or administrated at later stages (3 months of age) by stereotaxic injection into the subiculum. In both cells and the 3xTgAD mouse model, exogenous TFEB strongly reduced C99 load and concomitantly increased the levels of many lysosomal and autophagic proteins, including cathepsins, key proteases involved in C99 degradation. Our data indicate that TFEB activation is a relevant strategy to prevent the accumulation of this early neurotoxic catabolite., Competing Interests: The authors have no conflicts or financial disclosures to disclose.
- Published
- 2020
- Full Text
- View/download PDF
18. Palmitate Is Increased in the Cerebrospinal Fluid of Humans with Obesity and Induces Memory Impairment in Mice via Pro-inflammatory TNF-α.
- Author
-
Melo HM, Seixas da Silva GDS, Sant'Ana MR, Teixeira CVL, Clarke JR, Miya Coreixas VS, de Melo BC, Fortuna JTS, Forny-Germano L, Ledo JH, Oliveira MS, Figueiredo CP, Pardossi-Piquard R, Checler F, Delgado-García JM, Gruart A, Velloso LA, Balthazar MLF, Cintra DE, Ferreira ST, and De Felice FG
- Subjects
- Animals, Humans, Memory Disorders pathology, Mice, Obesity pathology, Memory Disorders etiology, Obesity cerebrospinal fluid, Palmitates cerebrospinal fluid, Tumor Necrosis Factor-alpha metabolism
- Abstract
Obesity has been associated with cognitive decline, atrophy of brain regions related to learning and memory, and higher risk of developing dementia. However, the molecular mechanisms underlying these neurological alterations are still largely unknown. Here, we investigate the effects of palmitate, a saturated fatty acid present at high amounts in fat-rich diets, in the brain. Palmitate is increased in the cerebrospinal fluid (CSF) of overweight and obese patients with amnestic mild cognitive impairment. In mice, intracerebroventricular infusion of palmitate impairs synaptic plasticity and memory. Palmitate induces astroglial and microglial activation in the mouse hippocampus, and its deleterious impact is mediated by microglia-derived tumor necrosis factor alpha (TNF-α) signaling. Our results establish that obesity is associated with increases in CSF palmitate. By defining a pro-inflammatory mechanism by which abnormal levels of palmitate in the brain impair memory, the results further suggest that anti-inflammatory strategies may attenuate memory impairment in obesity., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
19. Targeting γ-secretase triggers the selective enrichment of oligomeric APP-CTFs in brain extracellular vesicles from Alzheimer cell and mouse models.
- Author
-
Lauritzen I, Bécot A, Bourgeois A, Pardossi-Piquard R, Biferi MG, Barkats M, and Checler F
- Abstract
Background: We recently demonstrated an endolysosomal accumulation of the β-secretase-derived APP C-terminal fragment (CTF) C99 in brains of Alzheimer disease (AD) mouse models. Moreover, we showed that the treatment with the γ-secretase inhibitor (D6) led to further increased endolysosomal APP-CTF levels, but also revealed extracellular APP-CTF-associated immunostaining. We here hypothesized that this latter staining could reflect extracellular vesicle (EV)-associated APP-CTFs and aimed to characterize these γ-secretase inhibitor-induced APP-CTFs., Methods: EVs were purified from cell media or mouse brains from vehicle- or D6-treated C99 or APP
swedish expressing cells/mice and analyzed for APP-CTFs by immunoblot. Combined pharmacological, immunological and genetic approaches (presenilin invalidation and C99 dimerization mutants (GXXXG)) were used to characterize vesicle-containing APP-CTFs. Subcellular APP-CTF localization was determined by immunocytochemistry., Results: Purified EVs from both AD cell or mouse models were enriched in APP-CTFs as compared to EVs from control cells/brains. Surprisingly, EVs from D6-treated cells not only displayed increased C99 and C99-derived C83 levels but also higher molecular weight (HMW) APP-CTF-immunoreactivities that were hardly detectable in whole cell extracts. Accordingly, the intracellular levels of HMW APP-CTFs were amplified by the exosomal inhibitor GW4869. By combined pharmacological, immunological and genetic approaches, we established that these HMW APP-CTFs correspond to oligomeric APP-CTFs composed of C99 and/or C83. Immunocytochemical analysis showed that monomers were localized mainly to the trans -Golgi network, whereas oligomers were confined to endosomes and lysosomes, thus providing an anatomical support for the selective recovery of HMW APP-CTFs in EVs. The D6-induced APP-CTF oligomerization and subcellular mislocalization was indeed due to γ-secretase blockade, since it similarly occurred in presenilin-deficient fibroblasts. Further, our data proposed that besides favoring APP-CTF oligomerization by preventing C99 proteolysis, γ-secretase inhibiton also led to a defective SorLA-mediated retrograde transport of HMW APP-CTFs from endosomal compartments to the TGN., Conclusions: This is the first study to demonstrate the presence of oligomeric APP-CTFs in AD mouse models, the levels of which are selectively enriched in endolysosomal compartments including exosomes and amplified by γ-secretase inhibition. Future studies should evaluate the putative contribution of these exosome-associated APP-CTFs in AD onset, progression and spreading., Competing Interests: Competing interestsThe authors declare that they have no competing interests., (© The Author(s). 2019.)- Published
- 2019
- Full Text
- View/download PDF
20. Intraneuronal accumulation of C99 contributes to synaptic alterations, apathy-like behavior, and spatial learning deficits in 3×TgAD and 2×TgAD mice.
- Author
-
Bourgeois A, Lauritzen I, Lorivel T, Bauer C, Checler F, and Pardossi-Piquard R
- Subjects
- Amyloid beta-Protein Precursor genetics, Animals, Disease Models, Animal, Female, Hippocampus metabolism, Locomotion, Male, Mice, Transgenic, Peptide Fragments genetics, Peptide Fragments metabolism, Phenotype, Phosphorylation, tau Proteins genetics, tau Proteins metabolism, Alzheimer Disease metabolism, Amyloid beta-Protein Precursor metabolism, Apathy physiology, Long-Term Potentiation, Neurons metabolism, Spatial Learning physiology, Synapses physiology
- Abstract
The triple transgenic mouse model (3×TgAD: APPswe, Tau
P301L , PS1M146V ) recapitulates both amyloid β (Aβ)- and tau-related lesions as well as synaptic and memory deficits. In these mice, we reported an early apathy-like behavior and alterations in synaptic plasticity appearing concomitantly with intraneuronal accumulation of C99 in the subiculum. To delineate the genuine contribution of C99 on the above phenotypes, we generated double transgenic mice (2×TgAD: APPswe, TauP301L ) that accumulate C99 without Aβ deposition or hyperphosphorylation of tau and compared them to 3×TgAD mice. Here, we show that both TgAD mice display similar decreases in long-term potentiation and in spontaneous locomotor activity measured by actimetry suggesting that the synaptic alterations and the apathy-like behavior were likely linked to C99 rather than Aβ. However, spatial learning alterations, assessed by the Morris water maze task, are more pronounced in 3×TgAD than in 2×TgAD, suggesting that both Aβ and C99 contribute to defects in the acquisition of spatial information. Finally, even if similar results are observed in males, cognitive and non-cognitive deficits are more severe in females., (Copyright © 2018 Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
21. β-Amyloid Precursor Protein Intracellular Domain Controls Mitochondrial Function by Modulating Phosphatase and Tensin Homolog-Induced Kinase 1 Transcription in Cells and in Alzheimer Mice Models.
- Author
-
Goiran T, Duplan E, Chami M, Bourgeois A, El Manaa W, Rouland L, Dunys J, Lauritzen I, You H, Stambolic V, Biféri MG, Barkats M, Pimplikar SW, Sergeant N, Colin M, Morais VA, Pardossi-Piquard R, Checler F, and Alves da Costa C
- Subjects
- Animals, Cell Line, Disease Models, Animal, Embryo, Mammalian, Fibroblasts, HEK293 Cells, Humans, Intracellular Space metabolism, Male, Mice, Mice, 129 Strain, Mice, Inbred C57BL, Mice, Transgenic, Alzheimer Disease metabolism, Amyloid Precursor Protein Secretases metabolism, Amyloid beta-Protein Precursor metabolism, Forkhead Box Protein O3 metabolism, Hippocampus metabolism, Mitochondria metabolism, Presenilins metabolism, Protein Kinases metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
Background: Mitophagy and mitochondrial dynamics alterations are two major hallmarks of neurodegenerative diseases. Dysfunctional mitochondria accumulate in Alzheimer's disease-affected brains by yet unexplained mechanisms., Methods: We combined cell biology, molecular biology, and pharmacological approaches to unravel a novel molecular pathway by which presenilins control phosphatase and tensin homolog-induced kinase 1 (Pink-1) expression and transcription. In vivo approaches were carried out on various transgenic and knockout animals as well as in adeno-associated virus-infected mice. Functional readout and mitochondrial physiology (mitochondrial potential) were assessed by combined procedures including flow cytometry, live imaging analysis, and immunohistochemistry., Results: We show that presenilins 1 and 2 trigger opposite effects on promoter transactivation, messenger RNA, and protein expression of Pink-1. This control is linked to γ-secretase activity and β-amyloid precursor protein but is independent of phosphatase and tensin homolog. We show that amyloid precursor protein intracellular domain (AICD) accounts for presenilin-dependent phenotype and upregulates Pink-1 transactivation in cells as well as in vivo in a Forkhead box O3a-dependent manner. Interestingly, the modulation of γ-secretase activity or AICD expression affects Pink-1-related control of mitophagy and mitochondrial dynamics. Finally, we show that parkin acts upstream of presenilins to control Pink-1 promoter transactivation and protein expression., Conclusions: Overall, we delineate a molecular cascade presenilins-AICD-Forkhead box O3a linking parkin to Pink-1. Our study demonstrates AICD-mediated Pink-1-dependent control of mitochondrial physiology by presenilins. Furthermore, it unravels a parkin-Pink-1 feedback loop controlling mitochondrial physiology that could be disrupted in neurodegenerative conditions., (Copyright © 2017 Society of Biological Psychiatry. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
22. Intraneuronal aggregation of the β-CTF fragment of APP (C99) induces Aβ-independent lysosomal-autophagic pathology.
- Author
-
Lauritzen I, Pardossi-Piquard R, Bourgeois A, Pagnotta S, Biferi MG, Barkats M, Lacor P, Klein W, Bauer C, and Checler F
- Subjects
- Alzheimer Disease metabolism, Amyloid beta-Peptides metabolism, Animals, Autophagy physiology, Brain metabolism, Disease Models, Animal, Endosomes metabolism, Lysosomes metabolism, Mice, Inbred C57BL, Mice, Transgenic, Neurons pathology, Alzheimer Disease pathology, Amyloid Precursor Protein Secretases metabolism, Amyloid beta-Protein Precursor metabolism, Brain pathology, Neurons metabolism
- Abstract
Endosomal-autophagic-lysosomal (EAL) dysfunction is an early and prominent neuropathological feature of Alzheimers's disease, yet the exact molecular mechanisms contributing to this pathology remain undefined. By combined biochemical, immunohistochemical and ultrastructural approaches, we demonstrate a link between EAL pathology and the intraneuronal accumulation of the β-secretase-derived βAPP fragment (C99) in two in vivo models, 3xTgAD mice and adeno-associated viral-mediated C99-infected mice. We present a pathological loop in which the accumulation of C99 is both the effect and causality of impaired lysosomal-autophagic function. The deleterious effect of C99 was found to be linked to its aggregation within EAL-vesicle membranes leading to disrupted lysosomal proteolysis and autophagic impairment. This effect was Aβ independent and was even exacerbated when γ-secretase was pharmacologically inhibited. No effect was observed in inhibitor-treated wild-type animals suggesting that lysosomal dysfunction was indeed directly linked to C99 accumulation. In some brain areas, strong C99 expression also led to inflammatory responses and synaptic dysfunction. Taken together, this work demonstrates a toxic effect of C99 which could underlie some of the early-stage anatomical hallmarks of Alzheimer's disease pathology. Our work also proposes molecular mechanisms likely explaining some of the unfavorable side-effects associated with γ-secretase inhibitor-directed therapies.
- Published
- 2016
- Full Text
- View/download PDF
23. The β-secretase-derived C-terminal fragment of βAPP, C99, but not Aβ, is a key contributor to early intraneuronal lesions in triple-transgenic mouse hippocampus.
- Author
-
Lauritzen I, Pardossi-Piquard R, Bauer C, Brigham E, Abraham JD, Ranaldi S, Fraser P, St-George-Hyslop P, Le Thuc O, Espin V, Chami L, Dunys J, and Checler F
- Subjects
- Aging physiology, Amyloid Precursor Protein Secretases antagonists & inhibitors, Amyloid beta-Protein Precursor chemistry, Animals, Blotting, Western, Electrophoresis, Polyacrylamide Gel, Enzyme-Linked Immunosorbent Assay, Hippocampus enzymology, Hippocampus growth & development, Hippocampus metabolism, Immunohistochemistry, Immunoprecipitation, Mice, Mice, Transgenic, Peptide Fragments chemistry, Presenilin-1 genetics, Presenilin-1 metabolism, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, tau Proteins genetics, tau Proteins metabolism, Amyloid Precursor Protein Secretases metabolism, Amyloid beta-Peptides physiology, Amyloid beta-Protein Precursor physiology, Hippocampus pathology, Interneurons pathology, Peptide Fragments physiology
- Abstract
Triple-transgenic mice (3xTgAD) overexpressing Swedish-mutated β-amyloid precursor protein (βAPP(swe)), P310L-Tau (Tau(P301L)), and physiological levels of M146V-presenilin-1 (PS1(M146V)) display extracellular amyloid-β peptides (Aβ) deposits and Tau tangles. More disputed is the observation that these mice accumulate intraneuronal Aβ that has been linked to synaptic dysfunction and cognitive deficits. Here, we provide immunohistological, genetic, and pharmacological evidences for early, age-dependent, and hippocampus-specific accumulation of the β-secretase-derived βAPP fragment C99 that is observed from 3 months of age and enhanced by pharmacological blockade of γ-secretase. Notably, intracellular Aβ is only detectable several months later and appears, as is the case of C99, in enlarged cathepsin B-positive structures, while extracellular Aβ deposits are detected ~12 months of age and beyond. Early C99 production occurs mainly in the CA1/subicular interchange area of the hippocampus corresponding to the first region exhibiting plaques and tangles in old mice. Furthermore, the comparison of 3xTgAD mice with double-transgenic mice bearing the βAPP(swe) and Tau(P301L) mutations but expressing endogenous PS1 (2xTgAD) demonstrate that C99 accumulation is not accounted for by a loss of function triggered by PS1 mutation that would have prevented C99 secondary cleavage by γ-secretase. Together, our work identifies C99 as the earliest βAPP catabolite and main contributor to the intracellular βAPP-related immunoreactivity in 3xTgAD mice, suggesting its implication as an initiator of the neurodegenerative process and cognitive alterations taking place in this mouse model.
- Published
- 2012
- Full Text
- View/download PDF
24. The physiology of the β-amyloid precursor protein intracellular domain AICD.
- Author
-
Pardossi-Piquard R and Checler F
- Subjects
- Alzheimer Disease enzymology, Alzheimer Disease genetics, Animals, Cytoplasm physiology, Gene Targeting methods, Humans, Intracellular Fluid physiology, Protein Structure, Tertiary physiology, Transcription Factors genetics, Transcription Factors metabolism, Alzheimer Disease metabolism, Amyloid Precursor Protein Secretases physiology, Amyloid beta-Protein Precursor chemistry, Amyloid beta-Protein Precursor physiology, Cytoplasm chemistry, Intracellular Fluid chemistry
- Abstract
The amyloid-β precursor protein (βAPP) undergoes several cleavages by enzymatic activities called secretases. Numerous studies aimed at studying the biogenesis and catabolic fate of Aβ peptides, the proteinaceous component of the senile plaques that accumulate in Alzheimer's disease-affected brains. Relatively recently, another secretase-mediated β-APP-derived catabolite called APP IntraCellular Domain (AICD) entered the game. Whether AICD corresponded to a biologically inert by-pass product of βAPP processing or whether it could harbor its own function remained questionable. In this study, we review the mechanisms by which AICD is generated and how its production is regulated. Furthermore, we discuss the degradation mechanism underlying its rapid catabolic fate. Finally, we review putative AICD-related functions and more particularly, the numerous studies indicating that AICD could translocate to the nucleus and control at a transcriptional level, the expression of a series of proteins involved in various functions including the control of cell death and Aβ degradation., (© 2011 The Authors. Journal of Neurochemistry © 2011 International Society for Neurochemistry.)
- Published
- 2012
- Full Text
- View/download PDF
25. Evidence that the amyloid-β protein precursor intracellular domain, AICD, derives from β-secretase-generated C-terminal fragment.
- Author
-
Flammang B, Pardossi-Piquard R, Sevalle J, Debayle D, Dabert-Gay AS, Thévenet A, Lauritzen I, and Checler F
- Subjects
- Amyloid beta-Protein Precursor chemistry, Amyloid beta-Protein Precursor genetics, Cell Line, Transformed, Cell Line, Tumor, Chromatography, High Pressure Liquid, Humans, Immunoprecipitation, Intracellular Fluid drug effects, Neuroblastoma pathology, Peptide Fragments pharmacology, Protein Structure, Tertiary, Proteolysis drug effects, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Substrate Specificity drug effects, Time Factors, Transfection, Amyloid Precursor Protein Secretases metabolism, Amyloid beta-Protein Precursor metabolism, Intracellular Fluid metabolism
- Abstract
One of the major pathological hallmarks of brains affected with Alzheimer's disease (AD) is the senile plaque, an extracellular deposit mainly composed of a set of highly insoluble peptides of various lengths (39-43 amino acids) referred to as amyloid-β (Aβ) peptides. Aβ peptides are derived from combined proteolytic cleavages undergone on the amyloid-β protein precursor (AβPP) by a set of enzymes called secretases. Several lines of anatomical and biological evidence suggest that Aβ peptides would not account for all pathological stigmata and molecular dysfunctions taking place in AD. In amyloidogenic and non-amyloidogenic pathways, AβPP first undergoes β- or α-secretases-mediated cleavages yielding C99 and C83, respectively. These two membrane-embedded C-terminal fragments are both potential targets of subsequent γ-secretase-mediated proteolysis. The latter cleavage not only generates either p3 or Aβ peptides but similarly gives rise to an AβPP IntraCellular Domain (AICD fragment) that could modulate the transcription of several genes linked to AD pathology. It is therefore striking that AICD theoretically derives from both amyloidogenic and non-amyloidogenic AβPP processing pathways. Here we show that AICD predominantly derives from C99 by means of recombinant substrates and transiently transfected cells expressing C99. Our data suggest a preferred pathogenic pathway for AICD production and suggests that this fragment, in addition to C99 and Aβ peptides, could contribute to AD pathology.
- Published
- 2012
- Full Text
- View/download PDF
26. γ-Secretase-mediated regulation of neprilysin: influence of cell density and aging and modulation by imatinib.
- Author
-
Bauer C, Pardossi-Piquard R, Dunys J, Roy M, and Checler F
- Subjects
- Animals, Benzamides, Cell Count methods, Cellular Senescence drug effects, HEK293 Cells, Humans, Imatinib Mesylate, Mice, Mice, Knockout, Neprilysin antagonists & inhibitors, Amyloid Precursor Protein Secretases physiology, Cellular Senescence physiology, Neprilysin physiology, Piperazines pharmacology, Protein Kinase Inhibitors pharmacology, Pyrimidines pharmacology
- Abstract
Proteolytic degradation has emerged as a key pathway involved in controlling levels of the Alzheimer's disease (AD)-associated amyloid-β peptides (Aβ) in the brain. The ectopeptidase, neprilysin (NEP), has been reported as the major Aβ-degrading enzyme in mice and human brains. We have previously shown that NEP expression and activity are regulated by AICD, the intracellular domain of the amyloid-β protein precursor (AβPP) generated by γ-secretase. Thus, NEP transcription, expression, and enzymatic activity are dramatically reduced in fibroblasts devoid of AβPP (the precursor of AICD) or lacking both presenilin (PS) 1 and 2 (two parent proteins contributing to AICD formation). We demonstrate here that NEP expression and activity are influenced by a number of cell passages and density, and we confirm a drastic reduction of NEP expression and activity in AβPP and PS null fibroblasts examined at similar passages and cell densities. Furthermore, Imatinib (Gleevec), a known tyrosine kinase inhibitor was recently shown to elevate AICD in H4 human neuroglioma cells, and this was accompanied by concomitant increases of NEP protein, mRNA levels, and activity. However, the demonstration of a causal link between Imatinib and AICD levels was still lacking. We show here an Imatinib-dependent effect on NEP expression and activity in murine fibroblasts and establish that Imatinib-induced modulation of NEP was abolished by the depletion of AβPP or its homologues APLP1 and APLP2, thereby confirming that Imatinib-mediated control of NEP could indeed be accounted for its effect on AICD.
- Published
- 2011
- Full Text
- View/download PDF
27. p53 is regulated by and regulates members of the gamma-secretase complex.
- Author
-
Checler F, Dunys J, Pardossi-Piquard R, and Alves da Costa C
- Subjects
- Alzheimer Disease genetics, Amyloid Precursor Protein Secretases genetics, Animals, Cell Death genetics, Cell Line, Transformed, Gene Expression Regulation genetics, Humans, Mice, Models, Biological, Presenilin-1 genetics, Presenilin-1 metabolism, Presenilin-2 genetics, Presenilin-2 metabolism, Transfection methods, Tumor Suppressor Protein p53 genetics, Alzheimer Disease metabolism, Amyloid Precursor Protein Secretases metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
Amyloid beta-peptides is the generic term for a set of hydrophobic peptides that accumulate in Alzheimer's disease (AD)-affected brains. These amyloid-beta peptide fragments are mainly generated by an enzymatic machinery referred to as gamma-secretase complex that is built up by the association of four distinct proteins, namely presenilin 1 (PS1) or PS2, nicastrin, Aph-1 and Pen-2. AD is also characterized by exacerbated cell death that appears linked to the tumor suppressor p53. Interestingly, all members of the gamma-secretase complex control p53-dependent cell death. On the other hand, p53 appears to be able to regulate directly or indirectly the expression and transcription of PS1, PS2 and Pen-2. This review will focus on the functional cross-talk between the members of the gamma-secretase complex and p53 and will discuss the putative implication of this oncogene in AD pathology., (Copyright 2010 S. Karger AG, Basel.)
- Published
- 2010
- Full Text
- View/download PDF
28. p53-dependent control of transactivation of the Pen2 promoter by presenilins.
- Author
-
Dunys J, Sevalle J, Giaime E, Pardossi-Piquard R, Vitek MP, Renbaum P, Levy-Lahad E, Zhang YW, Xu H, Checler F, and da Costa CA
- Subjects
- Alzheimer Disease genetics, Alzheimer Disease metabolism, Amyloid Precursor Protein Secretases metabolism, Amyloid beta-Protein Precursor genetics, Amyloid beta-Protein Precursor metabolism, Animals, Cell Line, Cells, Cultured, Fibroblasts metabolism, Humans, Membrane Proteins metabolism, Mice, Mice, Knockout, Presenilin-1 genetics, Presenilin-2 genetics, Tumor Suppressor Protein p53 genetics, Amyloid Precursor Protein Secretases genetics, Membrane Proteins genetics, Presenilin-1 metabolism, Presenilin-2 metabolism, Promoter Regions, Genetic, Transcriptional Activation, Tumor Suppressor Protein p53 metabolism
- Abstract
The senile plaques found in the brains of patients with Alzheimer's disease are mainly due to the accumulation of amyloid beta-peptides (A beta) that are liberated by gamma-secretase, a high molecular weight complex including presenilins, PEN-2, APH-1 and nicastrin. The depletion of each of these proteins disrupts the complex assembly into a functional protease. Here, we describe another level of regulation of this multimeric protease. The depletion of both presenilins drastically reduces Pen2 mRNA levels and its promoter transactivation. Furthermore, overexpression of presenilin-1 lowers Pen2 promoter transactivation, a phenotype abolished by a double mutation known to prevent presenilin-dependent gamma-secretase activity. PEN-2 expression is decreased by depletion of beta-amyloid precursor protein (APP) and increased by the APP intracellular domain (AICD). We show that AICD and APP complement for Pen2 mRNA levels in APP/APLP1-2 knockout fibroblasts. Interestingly, overexpression of presenilin-2 greatly increases Pen2 promoter transactivation. The opposite effect triggered by both presenilins was reminiscent of our previous study, which showed that these two proteins elicit antagonistic effects on p53. Therefore, we examined the contribution of p53 on Pen2 transcription. Pen2 promoter transactivation, and Pen2 mRNA and protein levels were drastically reduced in p53(-/-) fibroblasts. Furthermore, PEN-2 expression could be rescued by p53 complementation in p53- and APP-deficient cells. Interestingly, PEN-2 expression was also reduced in p53-deficient mouse brain. Overall, our study describes a p53-dependent regulation of PEN-2 expression by other members of the gamma-secretase complex, namely presenilins.
- Published
- 2009
- Full Text
- View/download PDF
29. TMP21 transmembrane domain regulates gamma-secretase cleavage.
- Author
-
Pardossi-Piquard R, Böhm C, Chen F, Kanemoto S, Checler F, Schmitt-Ulms G, St George-Hyslop P, and Fraser PE
- Subjects
- Amyloid beta-Peptides chemistry, Cell Line, Cell Membrane metabolism, Cell-Free System, Electrophoresis, Polyacrylamide Gel, Enzyme-Linked Immunosorbent Assay, Genes, Reporter, Humans, Lysine chemistry, Mutagenesis, Nucleocytoplasmic Transport Proteins, Protein Structure, Tertiary, Recombinant Fusion Proteins chemistry, Amyloid Precursor Protein Secretases metabolism, Membrane Proteins metabolism, Peptides chemistry
- Abstract
TMP21 has been shown to be associated with the gamma-secretase complex and can specifically regulate gamma-cleavage without affecting epsilon-mediated proteolysis. To explore the basis of this activity, TMP21 modulation of gamma-secretase activity was investigated independent of epsilon-cleavage using an amyloid-beta precursor proteinepsilon (APPepsilon) construct which lacks the amyloid intracellular domain domain. The APPepsilon construct behaves similarly to the full-length precursor protein with respect to alpha- and beta-cleavages and is able to undergo normal gamma-processing. Co-expression of APPepsilon and TMP21 resulted in the accumulation of membrane-embedded higher molecular weight Abeta-positive fragments, consistent with an inhibition of gamma-secretase cleavage. The APPepsilon system was used to examine the functional domains of TMP21 through the investigation of a series of TMP21-p24a chimera proteins. It was found that chimeras containing the transmembrane domain bound to the gamma-secretase complex and could decrease gamma-secretase proteolytic processing. This was confirmed though investigation of a synthetic peptide corresponding to the TMP21 transmembrane helix. The isolated TMP21 TM peptide but not the homologous p24a domain was able to reduce Abeta production in a dose-dependent fashion. These observations suggest that the TMP21 transmembrane domain promotes its association with the presenilin complex that results in decreased gamma-cleavage activity.
- Published
- 2009
- Full Text
- View/download PDF
30. APH1 polar transmembrane residues regulate the assembly and activity of presenilin complexes.
- Author
-
Pardossi-Piquard R, Yang SP, Kanemoto S, Gu Y, Chen F, Böhm C, Sevalle J, Li T, Wong PC, Checler F, Schmitt-Ulms G, St George-Hyslop P, and Fraser PE
- Subjects
- Amino Acid Sequence, Amino Acid Substitution, Amyloid Precursor Protein Secretases metabolism, Animals, Aspartic Acid genetics, Catalysis, Cells, Cultured, Conserved Sequence, Endopeptidases, Fibroblasts cytology, Fibroblasts physiology, Histidine genetics, Humans, Membrane Proteins chemistry, Mice, Molecular Sequence Data, Mutagenesis, Site-Directed, Peptide Hydrolases chemistry, Protein Structure, Tertiary, Transfection, Membrane Proteins genetics, Membrane Proteins metabolism, Multiprotein Complexes metabolism, Peptide Hydrolases genetics, Peptide Hydrolases metabolism, Presenilins metabolism
- Abstract
Complexes involved in the gamma/epsilon-secretase-regulated intramembranous proteolysis of substrates such as the amyloid-beta precursor protein are composed primarily of presenilin (PS1 or PS2), nicastrin, anterior pharynx defective-1 (APH1), and PEN2. The presenilin aspartyl residues form the catalytic site, and similar potentially functional polar transmembrane residues in APH1 have been identified. Substitution of charged (E84A, R87A) or polar (Q83A) residues in TM3 had no effect on complex assembly or activity. In contrast, changes to either of two highly conserved histidines (H171A, H197A) located in TM5 and TM6 negatively affected PS1 cleavage and altered binding to other secretase components, resulting in decreased amyloid generating activity. Charge replacement with His-to-Lys substitutions rescued nicastrin maturation and PS1 endoproteolysis leading to assembly of the formation of structurally normal but proteolytically inactive gamma-secretase complexes. Substitution with a negatively charged side chain (His-to-Asp) or altering the structural location of the histidines also disrupted gamma-secretase binding and abolished functionality of APH1. These results suggest that the conserved transmembrane histidine residues contribute to APH1 function and can affect presenilin catalytic activity.
- Published
- 2009
- Full Text
- View/download PDF
31. p53-dependent control of cell death by nicastrin: lack of requirement for presenilin-dependent gamma-secretase complex.
- Author
-
Pardossi-Piquard R, Dunys J, Giaime E, Guillot-Sestier MV, St George-Hyslop P, Checler F, and Alves da Costa C
- Subjects
- Amyloid Precursor Protein Secretases biosynthesis, Amyloid Precursor Protein Secretases genetics, Amyloid Precursor Protein Secretases physiology, Cell Death physiology, Cell Line, Cell Survival genetics, Humans, Membrane Glycoproteins genetics, Phosphatidylinositol 3-Kinases physiology, Presenilins genetics, Proto-Oncogene Proteins c-akt physiology, Signal Transduction genetics, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 physiology, Amyloid Precursor Protein Secretases metabolism, Membrane Glycoproteins biosynthesis, Presenilins metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
Nicastrin (NCT) is a component of the presenilin (PS)-dependent gamma-secretase complexes that liberate amyloid beta-peptides from the beta-Amyloid Precursor Protein. Several lines of evidence indicate that the members of these complexes could also contribute to the control of cell death. Here we show that over-expression of NCT increases the viability of human embryonic kidney (HEK293) cells and decreases staurosporine (STS)- and thapsigargin (TPS)-induced caspase-3 activation in various cell lines from human and neuronal origins by Akt-dependent pathway. NCT lowers p53 expression, transcriptional activity and promoter transactivation and reduces p53 phosphorylation. NCT-associated protection against STS-stimulated cell death was completely abolished by p53 deficiency. Conversely, the depletion of NCT drastically enhances STS-induced caspase-3 activation and p53 pathway and favored p53 nuclear translocation. We examined whether NCT protective function depends on PS-dependent gamma-secretase activity. First, a 29-amino acid deletion known to reduce NCT-dependent amyloid beta-peptide production did not affect NCT-associated protective phenotype. Second, NCT still reduces STS-induced caspase-3 activation in fibroblasts lacking PS1 and PS2. Third, the gamma-secretase inhibitor DFK167 did not affect NCT-mediated reduction of p53 activity. Altogether, our study indicates that NCT controls cell death via phosphoinositide 3-kinase/Akt and p53-dependent pathways and that this function remains independent of the activity and molecular integrity of the gamma-secretase complexes.
- Published
- 2009
- Full Text
- View/download PDF
32. Overexpression of human CRB1 or related isoforms, CRB2 and CRB3, does not regulate the human presenilin complex in culture cells.
- Author
-
Pardossi-Piquard R, Chen F, Silva-Gagliardi NF, Szego M, McInnes R, McGlade CJ, St George-Hyslop P, and Fraser PE
- Subjects
- Animals, Blotting, Western, Cell Line, Electrophoresis, Polyacrylamide Gel, Enzyme-Linked Immunosorbent Assay, Fluorescent Antibody Technique, Humans, Immunoprecipitation, Mice, Carrier Proteins metabolism, Eye Proteins metabolism, Membrane Glycoproteins metabolism, Membrane Proteins metabolism, Nerve Tissue Proteins metabolism, Presenilins metabolism
- Abstract
The presenilin proteins (PS1 and PS2) with their partners (NCT, Aph1, and Pen2) are the major components of the high molecular weight gamma-secretase complex which facilitates the intramembraneous cleavage of various type 1 transmembrane proteins, including the amyloid-beta precursor protein (APP) and the Notch receptor. Additional gamma-secretase complex components may be involved in regulation of its activity and specificity. A recent investigation indicated that the Crumbs protein is a negative regulator of Notch signaling and may act by repressing gamma/epsilon-secretase activity in Drosophila [Herranz, H., Stamataki, E., Feiguin, F., and Milan, M. (2006) EMBO Rep. 7, 297-302]. To address this question, we investigated potential functional interactions between the human Crumbs homologues (CRB1, CRB2, and CRB3) and presenilin complexes which mediate gamma/epsilon-secretase cleavage of APP and Notch. We found no evidence for direct interaction between CRB1, CRB2, or CRB3 and presenilin complex components. Furthermore, overexpression of human CRB1 and related isoforms, CRB2 and CRB3, had no effect on the levels of presenilin complex components, on NCT maturation or on PS endoproteolysis, and did not alter Abeta AICD or NICD production. These results suggest that, in mammalian cells at least, Crumbs is unlikely to be a significant direct modulator of presenilin-dependent gamma/epsilon-secretase activity.
- Published
- 2007
- Full Text
- View/download PDF
33. The gamma/epsilon-secretase-derived APP intracellular domain fragments regulate p53.
- Author
-
Checler F, Sunyach C, Pardossi-Piquard R, Sévalle J, Vincent B, Kawarai T, Girardot N, St George-Hyslop P, and da Costa CA
- Subjects
- Amyloid beta-Peptides chemistry, Animals, Humans, Models, Biological, Protein Structure, Tertiary, Amyloid Precursor Protein Secretases metabolism, Amyloid beta-Peptides metabolism, Gene Expression Regulation drug effects, Peptides pharmacology, Tumor Suppressor Protein p53 metabolism
- Abstract
Amyloid beta-peptide (Abeta), which plays a central role in Alzheimer Disease, is generated by presenilin-dependent and presenilin-independent gamma-secretase cleavages of beta-amyloid precursor protein (betaAPP). We report that the presenilins (PS1 and PS2) also regulate p53-associated cell death. Thus, we established that PS deficiency, catalytically inactive PS mutants, gamma-secretase inhibitors and betaAPP or APLP2 depletion reduced the expression and activity of p53, and lowered the transactivation of its promoter and mRNA levels. p53 expression was also reduced in the brains or betaAPP-deficient mice or in brains where both PS had been invalidated by double conditional knock out. AICDC59 and AICDC50, the gamma- and epsilon-secretase-derived C-terminal fragments of betaAPP, respectively, trigger the activation of caspase-3, p53-dependent cell death, and increase p53 activity and mRNA. Finally, HEK293 cells expressing PS1 harboring familial AD (FAD) mutations or FAD-affected brains, all display enhanced p53 activity and p53 expression. Our studies demonstrate that AICDs control p53 at a transcriptional level, in vitro and in vivo and unravel a still unknown function for presenilins.
- Published
- 2007
- Full Text
- View/download PDF
34. Response to correspondence: Pardossi-Piquard et al., "Presenilin-dependent transcriptional control of the Abeta-degrading enzyme neprilysin by intracellular domains of betAAPP and APLP." Neuron 46, 541-554.
- Author
-
Pardossi-Piquard R, Dunys J, Kawarai T, Sunyach C, Alves da Costa C, Vincent B, Sévalle J, Pimplikar S, St George-Hyslop P, and Checler F
- Subjects
- Amyloid Precursor Protein Secretases deficiency, Animals, Cells, Cultured, Cytidine Deaminase metabolism, Down-Regulation physiology, Fibroblasts metabolism, Humans, Membrane Glycoproteins deficiency, Mice, Mice, Knockout, Neprilysin, Presenilins deficiency, Presenilins metabolism
- Published
- 2007
- Full Text
- View/download PDF
35. The neuronal sortilin-related receptor SORL1 is genetically associated with Alzheimer disease.
- Author
-
Rogaeva E, Meng Y, Lee JH, Gu Y, Kawarai T, Zou F, Katayama T, Baldwin CT, Cheng R, Hasegawa H, Chen F, Shibata N, Lunetta KL, Pardossi-Piquard R, Bohm C, Wakutani Y, Cupples LA, Cuenco KT, Green RC, Pinessi L, Rainero I, Sorbi S, Bruni A, Duara R, Friedland RP, Inzelberg R, Hampe W, Bujo H, Song YQ, Andersen OM, Willnow TE, Graff-Radford N, Petersen RC, Dickson D, Der SD, Fraser PE, Schmitt-Ulms G, Younkin S, Mayeux R, Farrer LA, and St George-Hyslop P
- Subjects
- Age of Onset, Amyloid beta-Peptides metabolism, Amyloid beta-Protein Precursor metabolism, Cell Line, Endosomes metabolism, Genetic Variation, Haplotypes, Humans, Introns, Models, Genetic, Organ Specificity, Polymorphism, Single Nucleotide, Protease Nexins, Receptors, Cell Surface metabolism, Vesicular Transport Proteins metabolism, Alzheimer Disease genetics, LDL-Receptor Related Proteins genetics, Membrane Transport Proteins genetics
- Abstract
The recycling of the amyloid precursor protein (APP) from the cell surface via the endocytic pathways plays a key role in the generation of amyloid beta peptide (Abeta) in Alzheimer disease. We report here that inherited variants in the SORL1 neuronal sorting receptor are associated with late-onset Alzheimer disease. These variants, which occur in at least two different clusters of intronic sequences within the SORL1 gene (also known as LR11 or SORLA) may regulate tissue-specific expression of SORL1. We also show that SORL1 directs trafficking of APP into recycling pathways and that when SORL1 is underexpressed, APP is sorted into Abeta-generating compartments. These data suggest that inherited or acquired changes in SORL1 expression or function are mechanistically involved in causing Alzheimer disease.
- Published
- 2007
- Full Text
- View/download PDF
36. Presenilin-dependent gamma-secretase-mediated control of p53-associated cell death in Alzheimer's disease.
- Author
-
Alves da Costa C, Sunyach C, Pardossi-Piquard R, Sévalle J, Vincent B, Boyer N, Kawarai T, Girardot N, St George-Hyslop P, and Checler F
- Subjects
- Adult, Aged, Alzheimer Disease genetics, Alzheimer Disease pathology, Amyloid Precursor Protein Secretases, Amyloid beta-Protein Precursor deficiency, Amyloid beta-Protein Precursor genetics, Amyloid beta-Protein Precursor metabolism, Aspartic Acid Endopeptidases, Brain metabolism, Case-Control Studies, Caspase 3, Caspases metabolism, Cell Death, Cell Line, Enzyme Activation, Female, Humans, Male, Membrane Proteins genetics, Middle Aged, Mutation, Nerve Tissue Proteins deficiency, Peptide Fragments metabolism, Presenilin-1, Presenilin-2, Promoter Regions, Genetic, Protein Structure, Tertiary, Transcription, Genetic, Transcriptional Activation, Tumor Suppressor Protein p53 genetics, Alzheimer Disease physiopathology, Endopeptidases metabolism, Membrane Proteins metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
Presenilins (PSs) are part of the gamma-secretase complex that produces the amyloid beta-peptide (Abeta) from its precursor [beta-amyloid precursor protein (betaAPP)]. Mutations in PS that cause familial Alzheimer's disease (FAD) increase Abeta production and trigger p53-dependent cell death. We demonstrate that PS deficiency, catalytically inactive PS mutants, gamma-secretase inhibitors, and betaAPP or amyloid precursor protein-like protein 2 (APLP2) depletion all reduce the expression and activity of p53 and lower the transactivation of its promoter and mRNA expression. p53 expression also is diminished in the brains of PS- or betaAPP-deficient mice. The gamma- and epsilon-secretase-derived amyloid intracellular C-terminal domain (AICD) fragments (AICDC59 and AICDC50, respectively) of betaAPP trigger p53-dependent cell death and increase p53 activity and mRNA. Finally, PS1 mutations enhance p53 activity in human embryonic kidney 293 cells and p53 expression in FAD-affected brains. Thus our study shows that AICDs control p53 at a transcriptional level, in vitro and in vivo, and that FAD mutations increase p53 expression and activity in cells and human brains.
- Published
- 2006
- Full Text
- View/download PDF
37. TMP21 is a presenilin complex component that modulates gamma-secretase but not epsilon-secretase activity.
- Author
-
Chen F, Hasegawa H, Schmitt-Ulms G, Kawarai T, Bohm C, Katayama T, Gu Y, Sanjo N, Glista M, Rogaeva E, Wakutani Y, Pardossi-Piquard R, Ruan X, Tandon A, Checler F, Marambaud P, Hansen K, Westaway D, St George-Hyslop P, and Fraser P
- Subjects
- Amyloid Precursor Protein Secretases, Amyloid beta-Peptides biosynthesis, Amyloid beta-Peptides genetics, Animals, Aspartic Acid Endopeptidases, Cell Line, Endopeptidases chemistry, Humans, Membrane Proteins chemistry, Membrane Proteins deficiency, Membrane Proteins genetics, Mice, Models, Biological, Nucleocytoplasmic Transport Proteins, Presenilin-1, Presenilin-2, Protein Binding, Substrate Specificity, Endopeptidases metabolism, Membrane Proteins metabolism, Multiprotein Complexes chemistry, Multiprotein Complexes metabolism
- Abstract
The presenilin proteins (PS1 and PS2) and their interacting partners nicastrin, aph-1 (refs 4, 5) and pen-2 (ref. 5) form a series of high-molecular-mass, membrane-bound protein complexes that are necessary for gamma-secretase and epsilon-secretase cleavage of selected type 1 transmembrane proteins, including the amyloid precursor protein, Notch and cadherins. Modest cleavage activity can be generated by reconstituting these four proteins in yeast and Spodoptera frugiperda (sf9) cells. However, a critical but unanswered question about the biology of the presenilin complexes is how their activity is modulated in terms of substrate specificity and/or relative activities at the gamma and epsilon sites. A corollary to this question is whether additional proteins in the presenilin complexes might subsume these putative regulatory functions. The hypothesis that additional proteins might exist in the presenilin complexes is supported by the fact that enzymatically active complexes have a mass that is much greater than predicted for a 1:1:1:1 stoichiometric complex (at least 650 kDa observed, compared with about 220 kDa predicted). To address these questions we undertook a search for presenilin-interacting proteins that differentially affected gamma- and epsilon-site cleavage events. Here we report that TMP21, a member of the p24 cargo protein family, is a component of presenilin complexes and differentially regulates gamma-secretase cleavage without affecting epsilon-secretase activity.
- Published
- 2006
- Full Text
- View/download PDF
38. Presenilin-dependent transcriptional control of the Abeta-degrading enzyme neprilysin by intracellular domains of betaAPP and APLP.
- Author
-
Pardossi-Piquard R, Petit A, Kawarai T, Sunyach C, Alves da Costa C, Vincent B, Ring S, D'Adamio L, Shen J, Müller U, St George Hyslop P, and Checler F
- Subjects
- Aged, Amyloid beta-Peptides chemistry, Amyloid beta-Peptides pharmacology, Amyloid beta-Protein Precursor chemistry, Amyloid beta-Protein Precursor deficiency, Animals, Blotting, Western methods, Cadherins metabolism, Cells, Cultured, Cloning, Molecular methods, Drug Interactions, Electrophoretic Mobility Shift Assay methods, Enzyme Activation drug effects, Enzyme Activation physiology, Enzyme Inhibitors pharmacology, Female, Fibroblasts drug effects, Fibroblasts metabolism, Fluorescent Antibody Technique methods, Humans, Membrane Proteins deficiency, Membrane Proteins genetics, Mice, Middle Aged, Models, Biological, Mutagenesis physiology, Neprilysin genetics, Peptide Fragments pharmacology, Promoter Regions, Genetic physiology, Protein Processing, Post-Translational drug effects, Protein Structure, Tertiary physiology, Receptors, Notch, Recombinant Proteins, Time Factors, Transfection, Amyloid beta-Peptides physiology, Amyloid beta-Protein Precursor physiology, Extracellular Space metabolism, Membrane Proteins metabolism, Neprilysin metabolism, Protein Processing, Post-Translational physiology
- Abstract
Amyloid beta-peptide (Abeta), which plays a central role in Alzheimer's 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.
- Published
- 2005
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.