Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 297(2): 100963, 2021 08.
Article in English | MEDLINE | ID: mdl-34265307

ABSTRACT

The amyloid cascade hypothesis, which proposes a prominent role for full-length amyloid ß peptides in Alzheimer's disease, is currently being questioned. In addition to full-length amyloid ß peptide, several N-terminally truncated fragments of amyloid ß peptide could well contribute to Alzheimer's disease setting and/or progression. Among them, pyroGlu3-amyloid ß peptide appears to be one of the main components of early anatomical lesions in Alzheimer's disease-affected brains. Little is known about the proteolytic activities that could account for the N-terminal truncations of full-length amyloid ß, but they appear as the rate-limiting enzymes yielding the Glu3-amyloid ß peptide sequence that undergoes subsequent cyclization by glutaminyl cyclase, thereby yielding pyroGlu3-amyloid ß. Here, we investigated the contribution of dipeptidyl peptidase 4 in Glu3-amyloid ß peptide formation and the functional influence of its genetic depletion or pharmacological blockade on spine maturation as well as on pyroGlu3-amyloid ß peptide and amyloid ß 42-positive plaques and amyloid ß 42 load in the triple transgenic Alzheimer's disease mouse model. Furthermore, we examined whether reduction of dipeptidyl peptidase 4 could rescue learning and memory deficits displayed by these mice. Our data establish that dipeptidyl peptidase 4 reduction alleviates anatomical, biochemical, and behavioral Alzheimer's disease-related defects. Furthermore, we demonstrate that dipeptidyl peptidase 4 activity is increased early in sporadic Alzheimer's disease brains. Thus, our data demonstrate that dipeptidyl peptidase 4 participates in pyroGlu3-amyloid ß peptide formation and that targeting this peptidase could be considered as an alternative strategy to interfere with Alzheimer's disease progression.


Subject(s)
Alzheimer Disease , Animals , Brain/metabolism , Dipeptidyl Peptidase 4 , Disease Models, Animal , Humans , Mice , Plaque, Amyloid
2.
Acta Neuropathol ; 141(6): 823-839, 2021 06.
Article in English | MEDLINE | ID: mdl-33881611

ABSTRACT

One of the main components of senile plaques in Alzheimer's disease (AD)-affected brain is the Aß peptide species harboring a pyroglutamate at position three pE3-Aß. Several studies indicated that pE3-Aß is toxic, prone to aggregation and serves as a seed of Aß aggregation. The cyclisation of the glutamate residue is produced by glutaminyl cyclase, the pharmacological and genetic reductions of which significantly alleviate AD-related anatomical lesions and cognitive defects in mice models. The cyclisation of the glutamate in position 3 requires prior removal of the Aß N-terminal aspartyl residue to allow subsequent biotransformation. The enzyme responsible for this rate-limiting catalytic step and its relevance as a putative trigger of AD pathology remained yet to be established. Here, we identify aminopeptidase A as the main exopeptidase involved in the N-terminal truncation of Aß and document its key contribution to AD-related anatomical and behavioral defects. First, we show by mass spectrometry that human recombinant aminopeptidase A (APA) truncates synthetic Aß1-40 to yield Aß2-40. We demonstrate that the pharmacological blockade of APA with its selective inhibitor RB150 restores the density of mature spines and significantly reduced filopodia-like processes in hippocampal organotypic slices cultures virally transduced with the Swedish mutated Aß-precursor protein (ßAPP). Pharmacological reduction of APA activity and lowering of its expression by shRNA affect pE3-42Aß- and Aß1-42-positive plaques and expressions in 3xTg-AD mice brains. Further, we show that both APA inhibitors and shRNA partly alleviate learning and memory deficits observed in 3xTg-AD mice. Importantly, we demonstrate that, concomitantly to the occurrence of pE3-42Aß-positive plaques, APA activity is augmented at early Braak stages in sporadic AD brains. Overall, our data indicate that APA is a key enzyme involved in Aß N-terminal truncation and suggest the potential benefit of targeting this proteolytic activity to interfere with AD pathology.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Brain/metabolism , Cognitive Dysfunction/metabolism , Glutamyl Aminopeptidase/metabolism , Animals , Brain/pathology , Cell Line , Disease Models, Animal , Glutamyl Aminopeptidase/antagonists & inhibitors , Glutamyl Aminopeptidase/physiology , Hippocampus/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plaque, Amyloid/pathology
3.
J Biol Chem ; 293(40): 15419-15428, 2018 10 05.
Article in English | MEDLINE | ID: mdl-30143530

ABSTRACT

The histopathology of Alzheimer's disease (AD) is characterized by neuronal loss, neurofibrillary tangles, and senile plaque formation. The latter results from an exacerbated production (familial AD cases) or altered degradation (sporadic cases) of 40/42-amino acid-long ß-amyloid peptides (Aß peptides) that are produced by sequential cleavages of Aß precursor protein (ßAPP) by ß- and γ-secretases. The amyloid cascade hypothesis proposes a key role for the full-length Aß42 and the Aß40/42 ratio in AD etiology, in which soluble Aß oligomers lead to neurotoxicity, tau hyperphosphorylation, aggregation, and, ultimately, cognitive defects. However, following this postulate, during the last decade, several clinical approaches aimed at decreasing full-length Aß42 production or neutralizing it by immunotherapy have failed to reduce or even stabilize AD-related decline. Thus, the Aß peptide (Aß40/42)-centric hypothesis is probably a simplified view of a much more complex situation involving a multiplicity of APP fragments and Aß catabolites. Indeed, biochemical analyses of AD brain deposits and fluids have unraveled an Aß peptidome consisting of additional Aß-related species. Such Aß catabolites could be due to either primary enzymatic cleavages of ßAPP or secondary processing of Aß itself by exopeptidases. Here, we review the diversity of N- and C-terminally truncated Aß peptides and their biosynthesis and outline their potential function/toxicity. We also highlight their potential as new pharmaceutical targets and biomarkers.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , CA1 Region, Hippocampal/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/chemistry , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/antagonists & inhibitors , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/genetics , Animals , Antibodies, Monoclonal/pharmacology , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/pathology , Disease Models, Animal , Gene Expression , Humans , Mice , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Nootropic Agents/pharmacology , Protein Domains , Proteolysis , tau Proteins/chemistry , tau Proteins/genetics , tau Proteins/metabolism
4.
Cell Death Differ ; 25(5): 873-884, 2018 05.
Article in English | MEDLINE | ID: mdl-29352272

ABSTRACT

p53 is a transcription factor that is implicated in the control of both apoptotic and autophagic cell death. This tumor suppressor elicits both pro-autophagic and anti-autophagic phenotypes depending of its intracellular localization. The ability of p53 to repress autophagy has been exclusively associated to its cytoplasmic localization. Here, we show that transcriptional activity of p53 also contributes to autophagy down-regulation. Thus, nuclear p53 controls PINK1, a key protein involved in the control of mitophagy, by repressing its promoter activity, protein and mRNA levels, ex-vivo and in vivo. We establish that deletion of an identified p53 responsive element on PINK1 promoter impacts p53-mediated PINK1 transcriptional repression and we demonstrate a p53-PINK1 physical interaction by chromatin immunoprecipitation. Accordingly, we show that only nuclear p53 accounts for its ability to repress PINK1 gene transcription. Further, we demonstrate ex-vivo and in vivo that p53 invalidation in human cells increases LC3 maturation as well as optineurin and NDP52 autophagy receptors expression and down-regulates TIM23, TOM20 and HSP60 mitophagy markers. Importantly, this phenotype is mimicked by TP53 invalidation in mice brain. Finally, by combining pharmacological and genetic approaches, we show that the p53-mediated negative regulation of autophagy is PINK1-dependent. Thus pifithrin-α-mediated blockade of p53 transcriptional activity enhances LC3 maturation and reduces p62, TIM23, TOM20 and HSP60 protein levels. This pifithrin-α-associated pro-mitophagy phenotype is fully abolished by PINK1 depletion. This data unravels a novel pathway by which nuclear p53 can repress autophagy/mitophagy that could underlie important dysfunctions in both neurodegenerative and cancer diseases.


Subject(s)
Autophagy , Down-Regulation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Neoplasms/metabolism , Protein Kinases/biosynthesis , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cytosol/metabolism , HEK293 Cells , Humans , Mice , Mice, Knockout , Mitophagy , Neoplasms/genetics , Neoplasms/pathology , Protein Kinases/genetics , Tumor Suppressor Protein p53/genetics
5.
Biol Psychiatry ; 83(5): 416-427, 2018 03 01.
Article in English | MEDLINE | ID: mdl-28587718

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 ß-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.


Subject(s)
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 , 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
6.
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
7.
Mol Neurodegener ; 9: 35, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25216759

ABSTRACT

Endoplasmic reticulum (ER) is the cellular compartment where secreted and integral membrane proteins are folded and matured. The accumulation of unfolded or misfolded proteins triggers a stress that is physiologically controlled by an adaptative protective response called Unfolded Protein Response (UPR). UPR is primordial to induce a quality control response and to restore ER homeostasis. When this adaptative response is defective, protein aggregates overwhelm cells and affect, among other mechanisms, synaptic function, signaling transduction and cell survival. Such dysfunction likely contributes to several neurodegenerative diseases that are indeed characterized by exacerbated protein aggregation, protein folding impairment, increased ER stress and UPR activation. This review briefly documents various aspects of the biology of the transcription factor XBP-1 (X-box Binding Protein-1) and summarizes recent findings concerning its putative contribution to the altered UPR response observed in various neurodegenerative disorders including Parkinson's and Alzheimer's diseases.


Subject(s)
DNA-Binding Proteins/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Transcription Factors/metabolism , Unfolded Protein Response/physiology , Animals , Humans , Regulatory Factor X Transcription Factors , X-Box Binding Protein 1
8.
Neurobiol Aging ; 34(2): 499-510, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22475621

ABSTRACT

Activation of EphB receptors by ephrinB (efnB) ligands on neuronal cell surface regulates important functions, including neurite outgrowth, axonal guidance, and synaptic plasticity. Here, we show that efnB rescues primary cortical neuronal cultures from necrotic cell death induced by glutamate excitotoxicity and that this function depends on EphB receptors. Importantly, the neuroprotective function of the efnB/EphB system depends on presenilin 1 (PS1), a protein that plays crucial roles in Alzheimer's disease (AD) neurodegeneration. Furthermore, absence of one PS1 allele results in significantly decreased neuroprotection, indicating that both PS1 alleles are necessary for full expression of the neuroprotective activity of the efnB/EphB system. We also show that the ability of brain-derived neurotrophic factor (BDNF) to protect neuronal cultures from glutamate-induced cell death depends on PS1. Neuroprotective functions of both efnB and BDNF, however, were independent of γ-secretase activity. Absence of PS1 decreases cell surface expression of neuronal TrkB and EphB2 without affecting total cellular levels of the receptors. Furthermore, PS1-knockout neurons show defective ligand-dependent internalization and decreased ligand-induced degradation of TrkB and Eph receptors. Our data show that PS1 mediates the neuroprotective activities of efnB and BDNF against excitotoxicity and regulates surface expression and ligand-induced metabolism of their cognate receptors. Together, our observations indicate that PS1 promotes neuronal survival by regulating neuroprotective functions of ligand-receptor systems.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Cerebral Cortex/metabolism , Ephrin-B2/pharmacology , Neurons/metabolism , Presenilin-1/metabolism , Receptor, EphB2/metabolism , Receptor, trkB/metabolism , Animals , Cell Survival/drug effects , Cell Survival/physiology , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Mice , Mice, Knockout , Neurons/cytology , Neurons/drug effects , Presenilin-1/genetics , Rats , Receptor, EphB2/genetics , Receptor, trkB/genetics , Signal Transduction/drug effects , Signal Transduction/physiology
9.
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
10.
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
11.
Neurodegener Dis ; 7(1-3): 50-5, 2010.
Article in English | MEDLINE | ID: mdl-20160459

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.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Tumor Suppressor Protein p53/metabolism , 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
12.
J Cell Sci ; 122(Pt 21): 4003-8, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19889971

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.


Subject(s)
Amyloid Precursor Protein Secretases/genetics , Membrane Proteins/genetics , Presenilin-1/metabolism , Presenilin-2/metabolism , Promoter Regions, Genetic , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism , 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
13.
J Neurochem ; 109(1): 225-37, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19187441

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.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Membrane Glycoproteins/biosynthesis , Presenilins/metabolism , Tumor Suppressor Protein p53/metabolism , 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
14.
Biochem Biophys Res Commun ; 371(1): 69-74, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18405662

ABSTRACT

The presenilin (PS)-dependent gamma-secretase activity refers to a high molecular mass-complex including, besides PS1 or PS2, three other proteins recently identified, namely nicastrin, Aph-1, and Pen-2. This proteolytic complex has been shown to contribute to both gamma- and epsilon-cleavages of the beta-amyloid precursor protein (betaAPP), thereby generating beta-amyloid peptides (Abeta) and the APP intracellular domain (AICD), respectively. TMP21, a member of the p24 cargo protein family, was recently shown to interact with PS complexes. Interestingly, TMP21 modulates gamma-secretase-mediated Abeta production but does not regulate epsilon-secretase-derived AICD formation [F. Chen, H. Hasegawa, G. Schmitt-ulms, T. Kawarai, C. Bohm, T. Katayama, Y. Gu, N. Sanjo, M. Glista, E. Rogaeva, Y. Wakutami, R. Pardossi-Piquard, X. Ruan, A. Tandon, F. Checler, P. Marambaud, K. Hansen, D. Westaway, P. St. George-Hyslop, P. Fraser, TMP21 is a presenilin complex component that modulates gamma- but not epsilon-secretase activities, Nature 440 (2006) 1208-1212]. Here we investigate the functional incidence of the over-expression or depletion of TMP21 on both intracellular and secreted Abeta recoveries and AICD-associated phenotypes. First we confirm that TMP21 depletion yields increased levels of secreted Abeta40. However, we demonstrate that both staurosporine-stimulated caspase-3 activation, p53 and neprilysin expression and activity were not affected by TMP21 over-expression or depletion. Overall, our functional data further reinforce the view that TMP21 behaves as a regulator of gamma- but not epsilon-cleavages generated by PS-dependent gamma-secretase complex.


Subject(s)
Amyloid beta-Peptides/metabolism , Membrane Proteins/metabolism , Peptide Fragments/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/genetics , Animals , Caspase 3/genetics , Caspase 3/metabolism , Cell Line , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Neprilysin/genetics , Neprilysin/metabolism , Nucleocytoplasmic Transport Proteins , Peptide Fragments/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
15.
Neurodegener Dis ; 4(2-3): 156-63, 2007.
Article in English | MEDLINE | ID: mdl-17596710

ABSTRACT

The presenilin-dependent gamma-secretase complex is mainly composed of four distinct proteins, namely presenilin 1 or presenilin 2, nicastrin, anterior pharynx defective-1 (Aph-1) and presenilin enhancer (Pen-2). The mechanisms by which the complex is assembled, how its stoichiometry is controlled and how its catalytic activity is regulated are poorly understood. Recent studies indicated that Aph-1 and Pen-2 undergo proteolysis by the proteasome. We have examined the susceptibility of endogenous and overexpressed Aph-1a and Pen-2 to proteolysis by endogenous and purified proteasome as well as by recombinant caspases. We show that endogenous Aph-1a and Pen-2 resist proteolysis by caspases and by the proteasome. Furthermore, we show that unexpected interference of proteasome inhibitors with the cmv promoter region driving expression of Aph-1a and Pen-2 led to artifactual enhancement of overexpressed Aph-1a and Pen-2-like immunoreactivities but that these proteins also resist to in vitro degradation by endogenous and purified proteasome.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Caspases/metabolism , Membrane Proteins/metabolism , Peptide Hydrolases/metabolism , Proteasome Endopeptidase Complex/metabolism , Amyloid Precursor Protein Secretases/genetics , Animals , Cattle , Cell Line , Dose-Response Relationship, Drug , Endopeptidases , Gene Expression/drug effects , Leupeptins/pharmacology , Membrane Proteins/genetics , Mutation/physiology , Oligopeptides/pharmacology , Peptide Hydrolases/genetics , Protease Inhibitors/pharmacology , Transfection/methods
17.
J Biol Chem ; 282(14): 10516-25, 2007 Apr 06.
Article in English | MEDLINE | ID: mdl-17276981

ABSTRACT

The presenilin-dependent gamma-secretase activity, which is responsible for the generation of amyloid beta-peptide, is a high molecular weight complex composed of at least four components, namely, presenilin-1 (or presenilin-2), nicastrin, Aph-1, and Pen-2. Previous data indicated that presenilins, which are thought to harbor the catalytic core of the complex, also control p53-dependent cell death. Whether the other components of the gamma-secretase complex could also modulate the cell death process in mammalian neurons remained to be established. Here, we examined the putative contribution of Aph-1 and Pen-2 in the control of apoptosis in TSM1 cells from a neuronal origin. We show by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling and DNA fragmentation analyses that the overexpression of Aph-1a, Aph-1b, or Pen-2 drastically lowered staurosporine-induced cellular toxicity. In support of an apoptosis rather than necrosis process, Aph-1 and Pen-2 also lower staurosporine- and etoposide-induced caspase-3 expression and diminished caspase-3 activity and poly(ADP-ribose) polymerase inactivation. The Aph-1 and Pen-2 anti-apoptotic phenotype was associated with a drastic reduction of p53 expression and activity and lowered p53 mRNA transcription. Furthermore, the Aph-1- and Pen-2-associated reduction of staurosporine-induced caspase-3 activation was fully abolished by p53 deficiency. Conversely, Aph-1a, Aph-1b, and Pen-2 gene inactivation increases both caspase-3 activity and p53 mRNA levels. Finally, we show that Aph-1 and Pen-2 did not trigger an anti-apoptotic response in cells devoid of presenilins or nicastrin, whereas the protective response was still observed in fibroblasts devoid of beta-amyloid precursor protein and amyloid precursor protein like-protein 2. Furthermore, Aph-1- and Pen-2-associated protection against staurosporine-induced caspase-3 activation was not affected by the gamma-secretase inhibitors N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester and difluoromethylketone. Altogether, our study indicates that Aph-1 and Pen-2 trigger an anti-apoptotic response by lowering p53-dependent control of caspase-3. Our work also demonstrates that this phenotype is strictly dependent on the molecular integrity of the gamma-secretase complex but remains independent of the gamma-secretase catalytic activity.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/biosynthesis , Membrane Proteins/metabolism , Multiprotein Complexes/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis , Caspase 3/metabolism , Cell Line , Endopeptidases , Enzyme Inhibitors/toxicity , Humans , Membrane Glycoproteins/metabolism , Peptide Hydrolases , Phenotype , Poly(ADP-ribose) Polymerases/metabolism , Presenilins/metabolism , Staurosporine/toxicity
18.
J Biol Chem ; 281(14): 9824-31, 2006 Apr 07.
Article in English | MEDLINE | ID: mdl-16464850

ABSTRACT

We established previously that alpha-synuclein displayed a protective anti-apoptotic phenotype in neurons, mainly by down-regulating p53-dependent caspase-3 activation (Alves da Costa, C., Ancolio, K., and Checler, F. (2000) J. Biol. Chem. 275, 24065-24069; Alves da Costa, C., Paitel, E., Vincent, B., and Checler, F. (2002) J. Biol. Chem. 277, 50980-50984). This function was abolished by Parkinson disease-linked pathogenic mutations and by the dopaminergic toxin, 6-hydroxydopamine (6OH-DOPA) (Alves da Costa, C., Paitel, E., Vincent, B., and Checler, F. (2002) J. Biol. Chem. 277, 50980-50984). However, the mechanisms by which 6OH-DOPA interfered with alpha-synuclein function remained unclear. Here we showed that 6OH-DOPA prevents alpha-synuclein-mediated anti-apoptotic function by altering its degradation. Thus, 6OH-DOPA treatment of TSM1 neurons and SH-SY5Y neuroblastoma cells enhances endogenous alpha-synuclein-like immunoreactivity and inhibits the catabolism of endogenous and recombinant alpha-synucleins by purified 20 S proteasome. Furthermore, we demonstrated that 6OH-DOPA directly inhibits endogenous proteasomal activity in TSM1 and SH-SY5Y cells and also blocks purified proteasome activity in vitro. This inhibitory effect can be prevented by the anti-oxidant phenyl-N-butylnitrone. We also established that 6OH-DOPA triggers the aggregation of recombinant alpha-synuclein in vitro. Therefore, we conclude that 6OH-DOPA abolishes alpha-synuclein anti-apoptotic phenotype by inhibiting its proteasomal degradation, thereby increasing its intracellular concentration and potential propensity to aggregation, the latter phenomenon being directly exacerbated by 6OH-DOPA itself. Interestingly, 1-methyl-4-phenylpyridinium (MPP(+)), another toxin inducer of Parkinson disease-like pathology, does not affect alpha-synuclein protective function and fails to trigger aggregation of recombinant alpha-synuclein. Furthermore, MPP(+) does not alter cellular proteasomal activity, and only high concentrations of the toxin affect purified 20 S proteasome by a mechanism that remains insensitive to phenyl-N-butylnitrone. The drastically distinct effects of 6OH-DOPA and MPP(+) on alpha-synuclein function are discussed with respect to Parkinson disease pathology and animal models mimicking this pathology.


Subject(s)
1-Methyl-4-phenylpyridinium/metabolism , Apoptosis/drug effects , Herbicides/metabolism , Oxidopamine/pharmacology , Proteasome Endopeptidase Complex/metabolism , alpha-Synuclein/drug effects , alpha-Synuclein/metabolism , Animals , Disease Models, Animal , Humans , Neuroblastoma/pathology , Neurons/physiology , Parkinson Disease/physiopathology , Phenotype , Tumor Cells, Cultured
19.
Biochem J ; 394(Pt 2): 501-9, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16302845

ABSTRACT

PS (presenilin)-dependent gamma-secretase occurs as a high-molecular-mass complex composed of either PS1 or PS2 associated with Nct (nicastrin), PEN2 (presenilin enhancer 2 homologue) and APH1 (anterior pharynx defective 1 homologue). Numerous reports have documented the very complicated physical and functional cross-talk between these proteins that ultimately governs the biological activity of the gamma-secretase, but very few studies examined the fate of the components of the complex. We show that, in both HEK-293 cells and the TSM1 neuronal cell line, the immunoreactivities of overexpressed myc-tagged-APH1a and -PEN2 were enhanced by the proteasome inhibitors ZIE and lactacystin, whereas a broad range of protease inhibitors had no effect. By contrast, proteasome inhibitors were totally unable to affect the cellular expression of endogenous APH1aL and PEN2 in HEK-293 cells, TSM1 and primary cultured cortical neurons. To explain this apparent discrepancy, we examined the degradation of myc-tagged-APH1a and -PEN2, in vitro, by cell extracts containing endogenous proteasome and by purified 20S proteasome. Strikingly, myc-tagged-APH1a and -PEN2 resist proteolysis by endogenous proteasome and purified 20S proteasome. We also show that endogenous PEN2 expression was drastically higher in wild-type than in PS- and Nct-deficient fibroblasts and was enhanced by proteasome inhibitors only in the two deficient cell systems. However, here again, purified 20S proteasome appeared unable to cleave endogenous PEN2 present in PS-deficient fibroblasts. The levels of endogenous APH1aL-like immunoreactivity were not modified by proteasome inhibitors and were unaffected by PS deficiency. Altogether, our results indicate that endogenous PEN2 and APH1aL do not undergo proteasomal degradation under physiological conditions in HEK-293 cells, TSM1 cells and fibroblasts and that the clearance of PEN2 in PS- and Nct-deficient fibroblasts is not mediated by 20S proteasome. Whether the 26S proteasome participates to PEN2 proteolysis in deficient fibroblasts remains to be established.


Subject(s)
Endopeptidases/metabolism , Membrane Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteins/metabolism , Animals , Cattle , Cell Line , Endopeptidases/genetics , Fibroblasts/metabolism , Gene Deletion , Gene Expression , Humans , Membrane Proteins/genetics , Mice , Neurons/metabolism , Proteasome Inhibitors , Proteins/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...