Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 15(5): 367, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806484

ABSTRACT

Mitochondria dysfunctions and mitophagy failure have been associated with several Alzheimer's disease (AD) related molecular actors including amyloid beta (Aß) 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 Aß concomitantly regulate the expression of a set of transport proteins as demonstrated in APPswe cells treated with ß- 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 Aß oligomers (Aßo) and APP-CTFs impair the colocalization of mitochondria and transport proteins. This has been demonstrated in differentiated SH-SY5Y naive cells treated with Aßo 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.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Mitochondria , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Mitochondria/metabolism , 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
2.
Cells ; 12(24)2023 12 08.
Article in English | MEDLINE | ID: mdl-38132122

ABSTRACT

The SORL1 gene encodes LR11/SorLA, a protein that binds ß-amyloid precursor protein (APP) and drives its intracellular trafficking. SORL1 mutations, occurring frequently in a subset of familial cases of Alzheimer's disease (AD), have been documented, but their pathogenic potential is not yet clear and questions remain concerning their putative influence on the physiopathological processing of APP. We have assessed the influence of two SORL1 mutations that were described as likely disease-causing and that were associated with either benign (SorLA924) or severe (SorLA511) AD phenotypes. We examined the influence of wild-type and mutants SorLA in transiently transfected HEK293 cells expressing either wild-type or Swedish mutated APP on APP expression, secreted Aß and sAPPα levels, intracellular Aß 40 and Aß42 peptides, APP-CTFs (C99 and C83) expressions, α-, ß- and γ-secretases expressions and activities as well as Aß and CTFs-degrading enzymes. These paradigms were studied in control conditions or after pharmacological proteasomal modulation. We also established stably transfected CHO cells expressing wild-type SorLA and established the colocalization of APP and either wild-type or mutant SorLA. SorLA mutations partially disrupt co-localization of wild-type sorLA with APP. Overall, although we mostly confirmed previous data concerning the influence of wild-type SorLA on APP processing, we were unable to evidence significant alterations triggered by our set of SorLA mutants, whatever the cells or pharmacological conditions examined. Our study , however, does not rule out the possibility that other AD-linked SORL1 mutations could indeed affect APP processing, and that pathogenic mutations examined in the present study could interfere with other cellular pathways/triggers in AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Animals , Cricetinae , Humans , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Cricetulus , HEK293 Cells , LDL-Receptor Related Proteins/metabolism , Membrane Transport Proteins/genetics , Mutation/genetics
5.
Mol Psychiatry ; 26(10): 5592-5607, 2021 10.
Article in English | MEDLINE | ID: mdl-33144711

ABSTRACT

Although APP metabolism is being intensively investigated, a large fraction of its modulators is yet to be characterized. In this context, we combined two genome-wide high-content screenings to assess the functional impact of miRNAs and genes on APP metabolism and the signaling pathways involved. This approach highlighted the involvement of FERMT2 (or Kindlin-2), a genetic risk factor of Alzheimer's disease (AD), as a potential key modulator of axon guidance, a neuronal process that depends on the regulation of APP metabolism. We found that FERMT2 directly interacts with APP to modulate its metabolism, and that FERMT2 underexpression impacts axonal growth, synaptic connectivity, and long-term potentiation in an APP-dependent manner. Last, the rs7143400-T allele, which is associated with an increased AD risk and localized within the 3'UTR of FERMT2, induced a downregulation of FERMT2 expression through binding of miR-4504 among others. This miRNA is mainly expressed in neurons and significantly overexpressed in AD brains compared to controls. Altogether, our data provide strong evidence for a detrimental effect of FERMT2 underexpression in neurons and insight into how this may influence AD pathogenesis.


Subject(s)
Alzheimer Disease , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Humans , Membrane Proteins , Neoplasm Proteins , Neuronal Plasticity/genetics , Neurons , Risk Factors
6.
Acta Neuropathol ; 141(1): 39-65, 2021 01.
Article in English | MEDLINE | ID: mdl-33079262

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 ß-secretase-derived APP-CTF fragment (C99) combined with ß- and γ-secretase inhibition, that APP-CTFs accumulation independently of Aß 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 Aß 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.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Brain/pathology , Mitochondria/pathology , Mitochondria/ultrastructure , Mitophagy/genetics , 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
7.
Neurobiol Aging ; 71: 21-31, 2018 11.
Article in English | MEDLINE | ID: mdl-30071370

ABSTRACT

The triple transgenic mouse model (3×TgAD: APPswe, TauP301L, PS1M146V) recapitulates both amyloid ß (Aß)- 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 Aß 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 Aß. However, spatial learning alterations, assessed by the Morris water maze task, are more pronounced in 3×TgAD than in 2×TgAD, suggesting that both Aß 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.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Apathy/physiology , Long-Term Potentiation , Neurons/metabolism , Spatial Learning/physiology , Synapses/physiology , 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
8.
Acta Neuropathol ; 134(5): 749-767, 2017 11.
Article in English | MEDLINE | ID: mdl-28631094

ABSTRACT

The mechanisms underlying ryanodine receptor (RyR) dysfunction associated with Alzheimer disease (AD) are still not well understood. Here, we show that neuronal RyR2 channels undergo post-translational remodeling (PKA phosphorylation, oxidation, and nitrosylation) in brains of AD patients, and in two murine models of AD (3 × Tg-AD, APP +/- /PS1 +/-). RyR2 is depleted of calstabin2 (KFBP12.6) in the channel complex, resulting in endoplasmic reticular (ER) calcium (Ca2+) leak. RyR-mediated ER Ca2+ leak activates Ca2+-dependent signaling pathways, contributing to AD pathogenesis. Pharmacological (using a novel RyR stabilizing drug Rycal) or genetic rescue of the RyR2-mediated intracellular Ca2+ leak improved synaptic plasticity, normalized behavioral and cognitive functions and reduced Aß load. Genetically altered mice with congenitally leaky RyR2 exhibited premature and severe defects in synaptic plasticity, behavior and cognitive function. These data provide a mechanism underlying leaky RyR2 channels, which could be considered as potential AD therapeutic targets.


Subject(s)
Alzheimer Disease/metabolism , Calcium/metabolism , Cognition Disorders/metabolism , Protein Processing, Post-Translational , Ryanodine Receptor Calcium Release Channel/metabolism , Alzheimer Disease/pathology , Animals , Calcium Signaling , Cognition Disorders/pathology , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Humans , Male , Maze Learning/physiology , Mice , Mice, Transgenic , Oxidative Stress/physiology , Phosphorylation , Recognition, Psychology/physiology , Sarcoplasmic Reticulum/metabolism
9.
Acta Neuropathol ; 133(6): 955-966, 2017 06.
Article in English | MEDLINE | ID: mdl-27933404

ABSTRACT

Genome-wide association studies (GWASs) have identified 19 susceptibility loci for Alzheimer's disease (AD). However, understanding how these genes are involved in the pathophysiology of AD is one of the main challenges of the "post-GWAS" era. At least 123 genes are located within the 19 susceptibility loci; hence, a conventional approach (studying the genes one by one) would not be time- and cost-effective. We therefore developed a genome-wide, high-content siRNA screening approach and used it to assess the functional impact of gene under-expression on APP metabolism. We found that 832 genes modulated APP metabolism. Eight of these genes were located within AD susceptibility loci. Only FERMT2 (a ß3-integrin co-activator) was also significantly associated with a variation in cerebrospinal fluid Aß peptide levels in 2886 AD cases. Lastly, we showed that the under-expression of FERMT2 increases Aß peptide production by raising levels of mature APP at the cell surface and facilitating its recycling. Taken as a whole, our data suggest that FERMT2 modulates the AD risk by regulating APP metabolism and Aß peptide production.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , RNA, Small Interfering/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Biomarkers/cerebrospinal fluid , Cell Membrane/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Genetic Loci , Genetic Predisposition to Disease , Genome-Wide Association Study , HEK293 Cells , Hippocampus/metabolism , Hippocampus/pathology , Humans , Neurons/metabolism , Neurons/pathology , RNA Interference , Rats
10.
J Alzheimers Dis ; 55(4): 1549-1570, 2017.
Article in English | MEDLINE | ID: mdl-27911326

ABSTRACT

Alteration of mitochondria-associated membranes (MAMs) has been proposed to contribute to the pathogenesis of Alzheimer's disease (AD). We studied herein the subcellular distribution, the processing, and the protein interactome of the amyloid-ß protein precursor (AßPP) and its proteolytic products in MAMs. We reveal that AßPP and its catabolites are present in MAMs in cellular models overexpressing wild type AßPP or AßPP harboring the double Swedish or London familial AD mutations, and in brains of transgenic mice model of AD. Furthermore, we evidenced that both ß- and γ-secretases are present and harbor AßPP processing activities in MAMs. Interestingly, cells overexpressing APPswe show increased ER-mitochondria contact sites. We also document increased neutral lipid accumulation linked to Aß production and reversed by inhibiting ß- or γ-secretases. Using a proteomic approach, we show that AßPP and its catabolites interact with key proteins of MAMs controlling mitochondria and ER functions. These data highlight the role of AßPP processing and proteomic interactome in MAMs deregulation taking place in AD.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cell Membrane/metabolism , Mitochondria/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , CHO Cells , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/ultrastructure , Cricetulus , Electron Transport Complex IV/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Immunoprecipitation , Mice , Mice, Transgenic , Microscopy, Electron , Mitochondria/drug effects , Mitochondria/ultrastructure , Mutation/genetics , Neuroblastoma/pathology , Presenilin-1/genetics , Presenilin-1/metabolism , Pyrazoles/pharmacology , Quinolines/pharmacology , Ryanodine Receptor Calcium Release Channel/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transfection , Voltage-Dependent Anion Channel 1/metabolism
11.
Sci Rep ; 6: 37436, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27853315

ABSTRACT

The aspartyl protease ß-site APP cleaving enzyme, BACE1, is the rate-limiting enzyme involved in the production of amyloid-ß peptide, which accumulates in both sporadic and familial cases of Alzheimer's disease and is at the center of gravity of the amyloid cascade hypothesis. In this context, unravelling the molecular mechanisms controlling BACE1 expression and activity in both physiological and pathological conditions remains of major importance. We previously demonstrated that Aß controlled BACE1 transcription in an NFκB-dependent manner. Here, we delineate an additional cellular pathway by which natural and synthetic Aß42 oligomers enhance active X-box binding protein XBP-1s. XBP-1s lowers BACE1 expression and activity indirectly, via the up-regulation of the ubiquitin-ligase HRD1 that acts as an endogenous down-regulator of BACE1. Thus, we delineate a novel pathway by which cells could compensate for Aß42 oligomers production and thus, associated toxicity, by triggering a compensatory mechanism aimed at lowering BACE-1-mediated Aß production by a molecular cascade involving XBP-1s and HRD1. It thus identifies HRD1 as a potential target for a novel Aß-centered therapeutic strategy.


Subject(s)
Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/pharmacology , Aspartic Acid Endopeptidases/genetics , Neurons/drug effects , Peptide Fragments/pharmacology , Ubiquitin-Protein Ligases/genetics , X-Box Binding Protein 1/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Cell Line , Cell Line, Tumor , Cycloheximide/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Luciferases/genetics , Luciferases/metabolism , Mice , Neurons/cytology , Neurons/metabolism , Plasmids/chemistry , Plasmids/metabolism , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Transfection , Ubiquitin-Protein Ligases/metabolism , X-Box Binding Protein 1/metabolism
12.
EBioMedicine ; 9: 278-292, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27333034

ABSTRACT

Although several ADAMs (A disintegrin-like and metalloproteases) have been shown to contribute to the amyloid precursor protein (APP) metabolism, the full spectrum of metalloproteases involved in this metabolism remains to be established. Transcriptomic analyses centred on metalloprotease genes unraveled a 50% decrease in ADAM30 expression that inversely correlates with amyloid load in Alzheimer's disease brains. Accordingly, in vitro down- or up-regulation of ADAM30 expression triggered an increase/decrease in Aß peptides levels whereas expression of a biologically inactive ADAM30 (ADAM30(mut)) did not affect Aß secretion. Proteomics/cell-based experiments showed that ADAM30-dependent regulation of APP metabolism required both cathepsin D (CTSD) activation and APP sorting to lysosomes. Accordingly, in Alzheimer-like transgenic mice, neuronal ADAM30 over-expression lowered Aß42 secretion in neuron primary cultures, soluble Aß42 and amyloid plaque load levels in the brain and concomitantly enhanced CTSD activity and finally rescued long term potentiation alterations. Our data thus indicate that lowering ADAM30 expression may favor Aß production, thereby contributing to Alzheimer's disease development.


Subject(s)
ADAM Proteins/metabolism , Amyloid beta-Peptides/metabolism , Cathepsin D/metabolism , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amino Acid Sequence , Animals , Brain/metabolism , Brain/pathology , Cathepsin D/chemistry , Cell Line, Tumor , Down-Regulation/drug effects , HEK293 Cells , Humans , Lysosomes/metabolism , Macrolides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Patch-Clamp Techniques , Pepstatins/pharmacology , RNA Interference , RNA, Small Interfering/metabolism
13.
Acta Neuropathol ; 132(2): 257-276, 2016 08.
Article in English | MEDLINE | ID: mdl-27138984

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 ß-secretase-derived ßAPP 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 Aß 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.


Subject(s)
Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Brain/pathology , Neurons/metabolism , 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
14.
Front Mol Neurosci ; 9: 163, 2016.
Article in English | MEDLINE | ID: mdl-28119565

ABSTRACT

We previously reported that deficiency of membrane-type five matrix metalloproteinase (MT5-MMP) prevents amyloid pathology in the cortex and hippocampus of 5xFAD mice, and ameliorates the functional outcome. We have now investigated whether the integrity of another important area affected in Alzheimer's disease (AD), the frontal cortex, was also preserved upon MT5-MMP deficiency in 4-month old mice at prodromal stages of the pathology. We used the olfactory H-maze (OHM) to show that learning impairment associated with dysfunctions of the frontal cortex in 5xFAD was prevented in bigenic 5xFAD/MT5-MMP-/- mice. The latter exhibited concomitant drastic reductions of amyloid beta peptide (Aß) assemblies (soluble, oligomeric and fibrillary) and its immediate precursor, C99. Simultaneously, astrocyte reactivity and tumor necrosis factor alpha (TNF-α) levels were also lowered. Moreover, MT5-MMP deficiency induced a decrease in N-terminal soluble fragments of amyloid precursor protein (APP), including soluble APPα (sAPPα), sAPPß and the MT5-MMP-linked fragment of 95 kDa, sAPP95. However, the lack of MT5-MMP did not affect the activity of ß- and γ-secretases. In cultured HEKswe cells, transiently expressed MT5-MMP localized to early endosomes and increased the content of APP and Aß40 in these organelles, as well as Aß levels in cell supernatants. This is the first evidence that the pro-amyloidogenic features of MT5-MMP lie, at least in part, on the ability of the proteinase to promote trafficking into one of the amyloidogenic subcellular loci. Together, our data further support the pathogenic role of MT5-MMP in AD and that its inhibition improves the functional and pathological outcomes, in this case in the frontal cortex. These data also support the idea that MT5-MMP could become a novel therapeutic target in AD.

15.
Cell Mol Life Sci ; 73(1): 217-36, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26202697

ABSTRACT

Membrane-type 5-matrix metalloproteinase (MT5-MMP) is a proteinase mainly expressed in the nervous system with emerging roles in brain pathophysiology. The implication of MT5-MMP in Alzheimer's disease (AD), notably its interplay with the amyloidogenic process, remains elusive. Accordingly, we crossed the genetically engineered 5xFAD mouse model of AD with MT5-MMP-deficient mice and examined the impact of MT5-MMP deficiency in bigenic 5xFAD/MT5-MMP(-/-) mice. At early stages (4 months) of the pathology, the levels of amyloid beta peptide (Aß) and its amyloid precursor protein (APP) C-terminal fragment C99 were largely reduced in the cortex and hippocampus of 5xFAD/MT5-MMP(-/-), compared to 5xFAD mice. Reduced amyloidosis in bigenic mice was concomitant with decreased glial reactivity and interleukin-1ß (IL-1ß) levels, and the preservation of long-term potentiation (LTP) and spatial learning, without changes in the activity of α-, ß- and γ-secretases. The positive impact of MT5-MMP deficiency was still noticeable at 16 months of age, as illustrated by reduced amyloid burden and gliosis, and a better preservation of the cortical neuronal network and synaptophysin levels in bigenic mice. MT5-MMP expressed in HEKswe cells colocalized and co-immunoprecipitated with APP and significantly increased the levels of Aß and C99. MT5-MMP also promoted the release of a soluble APP fragment of 95 kDa (sAPP95) in HEKswe cells. sAPP95 levels were significantly reduced in brain homogenates of 5xFAD/MT5-MMP(-/-) mice, supporting altogether the idea that MT5-MMP influences APP processing. MT5-MMP emerges as a new pro-amyloidogenic regulator of APP metabolism, whose deficiency alleviates amyloid pathology, neuroinflammation and cognitive decline.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/physiopathology , Hippocampus/enzymology , Hippocampus/physiopathology , Matrix Metalloproteinases, Membrane-Associated/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/analysis , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/analysis , Amyloid beta-Protein Precursor/metabolism , Animals , Cognition , Female , Gene Deletion , HEK293 Cells , Hippocampus/metabolism , Hippocampus/pathology , Humans , Long-Term Potentiation , Male , Matrix Metalloproteinases, Membrane-Associated/analysis , Matrix Metalloproteinases, Membrane-Associated/genetics , Mice, Inbred C57BL , Mice, Transgenic , Spatial Learning
16.
Neurobiol Aging ; 35(7): 1570-81, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24495834

ABSTRACT

The ß-amyloid precursor protein undergoes cleavages by ß- and γ-secretasses yielding amyloid-ß peptides (Aß) that accumulate in Alzheimer's disease. Subsequently, Aß peptides are targets of additional truncations or endoproteolytic cleavages explaining the diversity of Aß-related fragments recovered in cell media or pathologic human fluids. Here, we focused on Aß1-34 (Aß34) that has been detected both in vitro and in vivo and that derives from the hydrolysis of Aß by ß-secretase. We have obtained and fully characterized by immunologic and biochemical approaches, a polyclonal antibody that specifically recognizes the C-terminus of Aßx-34. We present immunohistochemical evidence for the presence of Aßx-34 in the brain of 3xTg mice and Alzheimer's disease-affected human brains. Finally, we demonstrate a neprilysin-mediated degradation process of Aß34 and the ability of synthetic Aß34 to protect HEK cells overexpressing either wild type or Swedish-mutated ß-amyloid precursor protein from apoptosis.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Brain/metabolism , Peptide Fragments/metabolism , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Protein Precursor/metabolism , Animals , Apoptosis/genetics , Aspartic Acid Endopeptidases/metabolism , Caspase 3/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , HEK293 Cells , Humans , Mice , Mice, Transgenic , Neprilysin/physiology
17.
J Mol Cell Biol ; 5(2): 132-42, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23359614

ABSTRACT

We previously established that besides its canonical function as E3-ubiquitin ligase, parkin also behaves as a transcriptional repressor of p53. Here we show that parkin differently modulates presenilin-1 and presenilin-2 expression and functions at transcriptional level. Thus, parkin enhances/reduces the protein expression, promoter activity and mRNA levels of presenilin-1 and presenilin-2, respectively, in cells and in vivo. This parkin-associated function is independent of its ubiquitin-ligase activity and remains unrelated to its capacity to repress p53. Accordingly, physical interaction of endogenous or overexpressed parkin with presenilins promoters is demonstrated by chromatin immunoprecipitation assays (ChIP). Furthermore, we identify a consensus sequence, the deletion of which abolishes parkin-dependent modulation of presenilins-1/2 and p53 promoter activities. Interestingly, electrophoretic mobility shift assays (EMSA) revealed a physical interaction between this consensus sequence and wild-type but not mutated parkin. Finally, we demonstrate that the RING1-IBR-RING2 domain of parkin harbors parkin's potential to modulate presenilins promoters. This transcriptional control impacts on presenilins-associated phenotypes, since parkin increases presenilin-1-associated γ-secretase activity and reduces presenilin-2-linked caspase-3 activation. Overall, our data delineate a promoter responsive element targeted by parkin that drives differential regulation of presenilin-1 and presenilin-2 transcription with functional consequences for γ-secretase activity and cell death.


Subject(s)
Presenilin-1/metabolism , Presenilin-2/metabolism , Promoter Regions, Genetic/physiology , Transcription, Genetic/physiology , Ubiquitin-Protein Ligases/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Caspase 3/genetics , Caspase 3/metabolism , Cell Death/physiology , Enzyme Activation/physiology , Mice , Mice, Knockout , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Presenilin-1/genetics , Presenilin-2/genetics , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics
18.
J Neurosci ; 32(46): 16243-1655a, 2012 Nov 14.
Article in English | MEDLINE | ID: mdl-23152608

ABSTRACT

Triple-transgenic mice (3xTgAD) overexpressing Swedish-mutated ß-amyloid precursor protein (ßAPP(swe)), P310L-Tau (Tau(P301L)), and physiological levels of M146V-presenilin-1 (PS1(M146V)) display extracellular amyloid-ß peptides (Aß) deposits and Tau tangles. More disputed is the observation that these mice accumulate intraneuronal Aß 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 ß-secretase-derived ßAPP fragment C99 that is observed from 3 months of age and enhanced by pharmacological blockade of γ-secretase. Notably, intracellular Aß is only detectable several months later and appears, as is the case of C99, in enlarged cathepsin B-positive structures, while extracellular Aß 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 ßAPP(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 ßAPP catabolite and main contributor to the intracellular ßAPP-related immunoreactivity in 3xTgAD mice, suggesting its implication as an initiator of the neurodegenerative process and cognitive alterations taking place in this mouse model.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/physiology , Amyloid beta-Protein Precursor/physiology , Hippocampus/pathology , Interneurons/pathology , Peptide Fragments/physiology , 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
19.
J Biol Chem ; 287(7): 5021-32, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-22184125

ABSTRACT

In physiological conditions, both ß-amyloid precursor protein (ßAPP) and cellular prion (PrP(c)) undergo similar disintegrin-mediated α-secretase cleavage yielding N-terminal secreted products referred to as soluble amyloid precursor protein-α (sAPPα) and N1, respectively. We recently demonstrated that N1 displays neuroprotective properties by reducing p53-dependent cell death both in vitro and in vivo. In this study, we examined the potential of N1 as a neuroprotector against amyloid ß (Aß)-mediated toxicity. We first show that both recombinant sAPPα and N1, but not its inactive parent fragment N2, reduce staurosporine-stimulated caspase-3 activation and TUNEL-positive cell death by lowering p53 promoter transactivation and activity in human cells. We demonstrate that N1 also lowers toxicity, cell death, and p53 pathway exacerbation triggered by Swedish mutated ßAPP overexpression in human cells. We designed a CHO cell line overexpressing the London mutated ßAPP (APP(LDN)) that yields Aß oligomers. N1 protected primary cultured neurons against toxicity and cell death triggered by oligomer-enriched APP(LDN)-derived conditioned medium. Finally, we establish that N1 also protects neurons against oligomers extracted from Alzheimer disease-affected brain tissues. Overall, our data indicate that a cellular prion catabolite could interfere with Aß-associated toxicity and that its production could be seen as a cellular protective mechanism aimed at compensating for an sAPPα deficit taking place at the early asymptomatic phase of Alzheimer disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Peptides/metabolism , PrPC Proteins/metabolism , Protein Multimerization , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/genetics , Animals , CHO Cells , Caspase 3/genetics , Caspase 3/metabolism , Cell Death , Cricetinae , Cricetulus , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Mice , Neurons/metabolism , Neurons/pathology , Peptides/genetics , PrPC Proteins/genetics , Staurosporine/pharmacology
20.
J Alzheimers Dis ; 27(3): 511-20, 2011.
Article in English | MEDLINE | ID: mdl-21841248

ABSTRACT

Proteolytic degradation has emerged as a key pathway involved in controlling levels of the Alzheimer's disease (AD)-associated amyloid-ß peptides (Aß) in the brain. The ectopeptidase, neprilysin (NEP), has been reported as the major Aß-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-ß protein precursor (AßPP) generated by γ-secretase. Thus, NEP transcription, expression, and enzymatic activity are dramatically reduced in fibroblasts devoid of AßPP (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 AßPP 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 AßPP or its homologues APLP1 and APLP2, thereby confirming that Imatinib-mediated control of NEP could indeed be accounted for its effect on AICD.


Subject(s)
Amyloid Precursor Protein Secretases/physiology , Cellular Senescence/physiology , Neprilysin/physiology , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Animals , Benzamides , Cell Count/methods , Cellular Senescence/drug effects , HEK293 Cells , Humans , Imatinib Mesylate , Mice , Mice, Knockout , Neprilysin/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...