1
|
Georgiadis D, Skoulikas N, Papakyriakou A, Stratikos E. Phosphinic Peptides as Tool Compounds for the Study of Pharmacologically Relevant Zn-Metalloproteases. ACS Pharmacol Transl Sci 2022; 5:1228-1253. [PMID: 36524013 PMCID: PMC9745897 DOI: 10.1021/acsptsci.2c00183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Indexed: 11/29/2022]
Abstract
Phosphinic peptides constitute an important class of bioactive compounds that have found a wide range of applications in the field of biology and pharmacology of Zn-metalloproteases, the largest family of proteases in humans. They are designed to mimic the structure of natural substrates during their proteolysis, thus acting as mechanism-based, transition state analogue inhibitors. A combination of electrostatic interactions between the phosphinic acid group and the Zn cation as well as optimal noncovalent enzyme-ligand interactions can result in both high binding affinity for the desired target and selectivity against other proteases. Due to these unique properties, phosphinic peptides have been mainly employed as tool compounds for (a) the purposes of rational drug design by serving as ligands in X-ray crystal structures of target enzymes and allowing the identification of crucial interactions that govern optimal molecular recognition, and (b) the delineation of biological pathways where Zn-metalloproteases are key regulators. For the latter objective, inhibitors of the phosphinopeptidic type have been used either unmodified or after being transformed to probes of various types, thus expanding the arsenal of functional tools available to researchers. The aim of this review is to summarize all recent research achievements in which phosphinic peptides have played a central role as tool compounds in the understanding of the mechanism and biological functions of Zn-metalloproteases in both health and disease.
Collapse
Affiliation(s)
- Dimitris Georgiadis
- Department
of Chemistry, National and Kapodistrian
University of Athens, GR-15784 Athens, Greece
| | - Nikolaos Skoulikas
- Department
of Chemistry, National and Kapodistrian
University of Athens, GR-15784 Athens, Greece
| | - Athanasios Papakyriakou
- National
Centre for Scientific Research “Demokritos”, Agia Paraskevi GR-15341 Athens, Greece
| | - Efstratios Stratikos
- Department
of Chemistry, National and Kapodistrian
University of Athens, GR-15784 Athens, Greece
- National
Centre for Scientific Research “Demokritos”, Agia Paraskevi GR-15341 Athens, Greece
| |
Collapse
|
2
|
Bécot A, Pardossi-Piquard R, Bourgeois A, Duplan E, Xiao Q, Diwan A, Lee JM, Lauritzen I, Checler F. The Transcription Factor EB Reduces the Intraneuronal Accumulation of the Beta-Secretase-Derived APP Fragment C99 in Cellular and Mouse Alzheimer’s Disease Models. Cells 2020; 9:cells9051204. [PMID: 32408680 PMCID: PMC7291113 DOI: 10.3390/cells9051204] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 04/29/2020] [Accepted: 05/08/2020] [Indexed: 01/20/2023] Open
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.
Collapse
Affiliation(s)
- Anaïs Bécot
- IPMC UMR 7275 CNRS/UCA, Laboratory of Excellence DistALZ, 660 route des Lucioles, 06650 Valbonne, France; (A.B.); (R.P.-P.); (A.B.); (E.D.); (I.L.)
| | - Raphaëlle Pardossi-Piquard
- IPMC UMR 7275 CNRS/UCA, Laboratory of Excellence DistALZ, 660 route des Lucioles, 06650 Valbonne, France; (A.B.); (R.P.-P.); (A.B.); (E.D.); (I.L.)
| | - Alexandre Bourgeois
- IPMC UMR 7275 CNRS/UCA, Laboratory of Excellence DistALZ, 660 route des Lucioles, 06650 Valbonne, France; (A.B.); (R.P.-P.); (A.B.); (E.D.); (I.L.)
| | - Eric Duplan
- IPMC UMR 7275 CNRS/UCA, Laboratory of Excellence DistALZ, 660 route des Lucioles, 06650 Valbonne, France; (A.B.); (R.P.-P.); (A.B.); (E.D.); (I.L.)
| | - Qingli Xiao
- Department of Neurology and the Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; (Q.X.); (J.-M.L.)
| | - Abhinav Diwan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA;
- John Cochran Veterans Affairs Medical Center, St. Louis, MO 63106, USA
| | - Jin-Moo Lee
- Department of Neurology and the Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; (Q.X.); (J.-M.L.)
| | - Inger Lauritzen
- IPMC UMR 7275 CNRS/UCA, Laboratory of Excellence DistALZ, 660 route des Lucioles, 06650 Valbonne, France; (A.B.); (R.P.-P.); (A.B.); (E.D.); (I.L.)
| | - Frédéric Checler
- IPMC UMR 7275 CNRS/UCA, Laboratory of Excellence DistALZ, 660 route des Lucioles, 06650 Valbonne, France; (A.B.); (R.P.-P.); (A.B.); (E.D.); (I.L.)
- Correspondence: ; Tel.: +33-493-953-460
| |
Collapse
|
3
|
Zhao ZH, Chen ZT, Zhou RL, Zhang X, Ye QY, Wang YZ. Increased DJ-1 and α-Synuclein in Plasma Neural-Derived Exosomes as Potential Markers for Parkinson's Disease. Front Aging Neurosci 2019; 10:438. [PMID: 30692923 PMCID: PMC6339871 DOI: 10.3389/fnagi.2018.00438] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 12/21/2018] [Indexed: 01/12/2023] Open
Abstract
The diagnosis of PD might be in difficulty, especially in the early stages. Therefore, the identification of novel biomarkers is imperative for the diagnosis and monitoring disease progression in PD. DJ-1 and α-synuclein, are two proteins that are critically involved in the pathogenesis of PD, and they have been examined as disease biomarkers in studies. However, no study exists regarding DJ-1 in plasma neural-derived exosomes. In the present study, the levels of DJ-1 and α-synuclein in plasma neural-derived exosomes were studied together in order to investigate novel biomarkers for PD. DJ-1 and α-synuclein in plasma and plasma neural-derived exosomes of the patients with PD and controls were quantified by ELISAs. The data revealed that the levels of DJ-1 and α-synuclein in plasma neural-derived exosomes and the ratio of plasma neural-derived exosomal DJ-1 to total DJ-1 were significantly higher in patients with PD, compared with controls, while levels of the two proteins in plasma exhibited no difference between the patients with PD and controls. However, no relationship was identified between biomarkers and disease progression. In addition, significant positive correlations between DJ-1 and α-synuclein in plasma neural-derived exosomes were found in the patients with PD and in healthy individuals. We hypothesize that DJ-1 in plasma neural-derived exosomes may be used as a potential biomarker as α-synuclein in PD and they might participate in the mechanism of PD together.
Collapse
Affiliation(s)
- Zhen-Hua Zhao
- Department of Neurology, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou, China
| | - Zhi-Ting Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Rui-Ling Zhou
- Department of Neurology, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou, China
| | - Xu Zhang
- Department of Neurology, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou, China
| | - Qin-Yong Ye
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yin-Zhou Wang
- Department of Neurology, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou, China
| |
Collapse
|
4
|
Py NA, Bonnet AE, Bernard A, Marchalant Y, Charrat E, Checler F, Khrestchatisky M, Baranger K, Rivera S. Differential spatio-temporal regulation of MMPs in the 5xFAD mouse model of Alzheimer's disease: evidence for a pro-amyloidogenic role of MT1-MMP. Front Aging Neurosci 2014; 6:247. [PMID: 25278878 PMCID: PMC4166961 DOI: 10.3389/fnagi.2014.00247] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 09/02/2014] [Indexed: 12/03/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are pleiotropic endopeptidases involved in a variety of neurodegenerative/neuroinflammatory processes through their interactions with a large number of substrates. Among those, the amyloid precursor protein (APP) and the beta amyloid peptide (Aβ) are largely associated with the development of Alzheimer’s disease (AD). However, the regulation and potential contribution of MMPs to AD remains unclear. In this study, we investigated the evolution of the expression of MMP-2, MMP-9, and membrane-type 1-MMP (MT1-MMP) in the hippocampus at different stages of the pathology (asymptomatic, prodromal-like and symptomatic) in the 5xFAD transgenic mouse AD model. In parallel we also followed the expression of functionally associated factors. Overall, the expression of MMP-2, MMP-9, and MT1-MMP was upregulated concomitantly with the tissue inhibitor of MMPs-1 (TIMP-1) and several markers of inflammatory/glial response. The three MMPs exhibited age- and cell-dependent upregulation of their expression, with MMP-2 and MMP-9 being primarily located to astrocytes, and MT1-MMP to neurons. MMP-9 and MT1-MMP were also prominently present in amyloid plaques. The levels of active MT1-MMP were highly upregulated in membrane-enriched fractions of hippocampus at 6 months of age (symptomatic phase), when the levels of APP, its metabolites APP C-terminal fragments (CTFs), and Aβ trimers were the highest. Overexpression of MT1-MMP in HEK cells carrying the human APP Swedish mutation (HEKswe) strongly increased β-secretase derived C-terminal APP fragment (C99) and Aβ levels, whereas MMP-2 overexpression nearly abolished Aβ production without affecting C99. Our data consolidate the emerging idea of a regulatory interplay between MMPs and the APP/Aβ system, and demonstrate for the first time the pro-amyloidogenic features of MT1-MMP. Further investigation will be justified to evaluate this MMP as a novel potential therapeutic target in AD.
Collapse
Affiliation(s)
- Nathalie A Py
- Faculty of Medicine, Aix-Marseille Université, CNRS, NICN, UMR7259 Marseille, France
| | - Amandine E Bonnet
- Faculty of Medicine, Aix-Marseille Université, CNRS, NICN, UMR7259 Marseille, France
| | - Anne Bernard
- Faculty of Medicine, Aix-Marseille Université, CNRS, NICN, UMR7259 Marseille, France
| | - Yannick Marchalant
- Faculty of Medicine, Aix-Marseille Université, CNRS, NICN, UMR7259 Marseille, France
| | - Eliane Charrat
- Faculty of Medicine, Aix-Marseille Université, CNRS, NICN, UMR7259 Marseille, France
| | | | - Michel Khrestchatisky
- Faculty of Medicine, Aix-Marseille Université, CNRS, NICN, UMR7259 Marseille, France
| | - Kévin Baranger
- Faculty of Medicine, Aix-Marseille Université, CNRS, NICN, UMR7259 Marseille, France ; Department of Neurology and Neuropsychology, APHM, CHU La Timone Marseille, France
| | - Santiago Rivera
- Faculty of Medicine, Aix-Marseille Université, CNRS, NICN, UMR7259 Marseille, France
| |
Collapse
|
5
|
Caillava C, Ranaldi S, Lauritzen I, Bauer C, Fareh J, Abraham JD, Checler F. Study on Aβ34 biology and detection in transgenic mice brains. Neurobiol Aging 2014; 35:1570-81. [PMID: 24495834 DOI: 10.1016/j.neurobiolaging.2014.01.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 01/02/2014] [Accepted: 01/08/2014] [Indexed: 11/28/2022]
Abstract
The β-amyloid precursor protein undergoes cleavages by β- and γ-secretasses yielding amyloid-β peptides (Aβ) that accumulate in Alzheimer's disease. Subsequently, Aβ peptides are targets of additional truncations or endoproteolytic cleavages explaining the diversity of Aβ-related fragments recovered in cell media or pathologic human fluids. Here, we focused on Aβ1-34 (Aβ34) that has been detected both in vitro and in vivo and that derives from the hydrolysis of Aβ by β-secretase. We have obtained and fully characterized by immunologic and biochemical approaches, a polyclonal antibody that specifically recognizes the C-terminus of Aβx-34. We present immunohistochemical evidence for the presence of Aβx-34 in the brain of 3xTg mice and Alzheimer's disease-affected human brains. Finally, we demonstrate a neprilysin-mediated degradation process of Aβ34 and the ability of synthetic Aβ34 to protect HEK cells overexpressing either wild type or Swedish-mutated β-amyloid precursor protein from apoptosis.
Collapse
Affiliation(s)
- Céline Caillava
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNS, Team "Fondation pour la Recherche Médicale" and "Labex Distalz", Valbonne, France
| | | | - Inger Lauritzen
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNS, Team "Fondation pour la Recherche Médicale" and "Labex Distalz", Valbonne, France
| | - Charlotte Bauer
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNS, Team "Fondation pour la Recherche Médicale" and "Labex Distalz", Valbonne, France
| | - Jeannette Fareh
- SysDiag CNRS-Bio-Rad, UMR3145, SysDiag,, Montpellier, France
| | | | - Frédéric Checler
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNS, Team "Fondation pour la Recherche Médicale" and "Labex Distalz", Valbonne, France.
| |
Collapse
|
6
|
Peuchmaur M, Lacour MA, Sévalle J, Lisowski V, Touati-Jallabe Y, Rodier F, Martinez J, Checler F, Hernandez JF. Further characterization of a putative serine protease contributing to the γ-secretase cleavage of β-amyloid precursor protein. Bioorg Med Chem 2013; 21:1018-29. [DOI: 10.1016/j.bmc.2012.11.045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 11/05/2012] [Accepted: 11/15/2012] [Indexed: 12/11/2022]
|
7
|
Narasingappa RB, Narasingapa RB, Javagal MR, Jargaval MR, Pullabhatla S, Htoo HH, Rao JKS, Hernandez JF, Govitrapong P, Vincent B. Activation of α-secretase by curcumin-aminoacid conjugates. Biochem Biophys Res Commun 2012; 424:691-6. [PMID: 22796219 DOI: 10.1016/j.bbrc.2012.07.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 07/03/2012] [Indexed: 11/16/2022]
Abstract
The extracellular senile plaques observed in Alzheimer's disease (AD) patients are mainly composed of amyloid peptides produced from the β-amyloid precursor protein (βAPP) by β- and γ-secretases. A third non-amyloidogenic α-secretase activity performed by the disintegrins ADAM10 and ADAM17 occurs in the middle of the amyloid-β peptide Aβ and liberates the large sAPPα neuroprotective fragment. Since the activation of α-secretase recently emerged as a promising therapeutic approach to treat AD, the identification of natural compounds able to trigger this cleavage is highly required. Here we describe new curcumin-based modified compounds as α-secretase activators. We established that the aminoacid conjugates curcumin-isoleucine, curcumin-phenylalanine and curcumin-valine promote the constitutive α-secretase activity and increase ADAM10 immunoreactivity. Strickingly, experiments carried out under conditions mimicking the PKC/muscarinic receptor-regulated pathway display different patterns of activation by these compounds. Altogether, our data identified new lead natural compounds for the future development of powerful and stable α-secretase activators and established that some of these molecules are able to discriminate between the constitutive and regulated α-secretase pathways.
Collapse
Affiliation(s)
- Ramesh B Narasingappa
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand
| | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Chami L, Buggia-Prévot V, Duplan E, Delprete D, Chami M, Peyron JF, Checler F. Nuclear factor-κB regulates βAPP and β- and γ-secretases differently at physiological and supraphysiological Aβ concentrations. J Biol Chem 2012; 287:24573-84. [PMID: 22654105 PMCID: PMC3397882 DOI: 10.1074/jbc.m111.333054] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 05/14/2012] [Indexed: 11/06/2022] Open
Abstract
Anatomical lesions in Alzheimer disease-affected brains mainly consist of senile plaques, inflammation stigmata, and oxidative stress. The nuclear factor-κB (NF-κB) is a stress-activated transcription factor that is activated around senile plaques. We have assessed whether NF-κB could be differentially regulated at physiological or supraphysiological levels of amyloid β (Aβ) peptides. Under these experimental conditions, we delineated the putative NF-κB-dependent modulation of all cellular participants in Aβ production, namely its precursor βAPP (β-amyloid precursor protein) and the β- and γ-secretases, the two enzymatic machines involved in Aβ genesis. Under physiological conditions, NF-κB lowers the transcriptional activity of the promoters of βAPP, β-secretase (β-site APP-cleaving enzyme 1, BACE1), and of the four protein components (Aph-1, Pen-2, nicastrin, presenilin-1, or presenilin-2) of the γ-secretase in HEK293 cells. This was accompanied by a reduction of both protein levels and enzymatic activities, thereby ultimately yielding lower amounts of Aβ and AICD (APP intracellular domain). In stably transfected Swedish βAPP-expressing HEK293 cells triggering supraphysiological concentrations of Aβ peptides, NF-κB activates the transcription of βAPP, BACE1, and some of the γ-secretase members and increases protein expression and enzymatic activities, resulting in enhanced Aβ production. Our pharmacological approach using distinct NF-κB kinase modulators indicates that both NF-κB canonical and alternative pathways are involved in the control of Aβ production. Overall, our data demonstrate that under physiological conditions, NF-κB triggers a repressive effect on Aβ production that contributes to maintaining its homeostasis, while NF-κB participates in a degenerative cycle where Aβ would feed its own production under pathological conditions.
Collapse
Affiliation(s)
- Linda Chami
- From the Équipe Labellisée “Fondation pour la Recherche Médicale” and “Excellence Laboratory Distalz”, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNSA, 660 Route des Lucioles, 06560, Valbonne, France and
| | - Virginie Buggia-Prévot
- From the Équipe Labellisée “Fondation pour la Recherche Médicale” and “Excellence Laboratory Distalz”, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNSA, 660 Route des Lucioles, 06560, Valbonne, France and
| | - Eric Duplan
- From the Équipe Labellisée “Fondation pour la Recherche Médicale” and “Excellence Laboratory Distalz”, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNSA, 660 Route des Lucioles, 06560, Valbonne, France and
| | - Dolores Delprete
- From the Équipe Labellisée “Fondation pour la Recherche Médicale” and “Excellence Laboratory Distalz”, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNSA, 660 Route des Lucioles, 06560, Valbonne, France and
| | - Mounia Chami
- From the Équipe Labellisée “Fondation pour la Recherche Médicale” and “Excellence Laboratory Distalz”, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNSA, 660 Route des Lucioles, 06560, Valbonne, France and
| | - Jean-François Peyron
- the Centre Méditerranéen de Médecine Moléculaire, UMR INSERM U895/UNS, 151 route Saint Antoine de Ginestière, BP 2 3194, 06204 Nice Cedex 3, France
| | - Frédéric Checler
- From the Équipe Labellisée “Fondation pour la Recherche Médicale” and “Excellence Laboratory Distalz”, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNSA, 660 Route des Lucioles, 06560, Valbonne, France and
| |
Collapse
|
9
|
Guillot-Sestier MV, Sunyach C, Ferreira ST, Marzolo MP, Bauer C, Thevenet A, Checler F. α-Secretase-derived fragment of cellular prion, N1, protects against monomeric and oligomeric amyloid β (Aβ)-associated cell death. J Biol Chem 2011; 287:5021-32. [PMID: 22184125 DOI: 10.1074/jbc.m111.323626] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
In physiological conditions, both β-amyloid precursor protein (βAPP) 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 β (Aβ)-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 βAPP overexpression in human cells. We designed a CHO cell line overexpressing the London mutated βAPP (APP(LDN)) that yields Aβ 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 Aβ-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.
Collapse
Affiliation(s)
- Marie-Victoire Guillot-Sestier
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR6097 CNRS/Université de Nice-Sophia-Antipolis (UNSA), 660 route des Lucioles, Sophia-Antipolis, 06560 Valbonne, France
| | | | | | | | | | | | | |
Collapse
|
10
|
Sevalle J, Amoyel A, Robert P, Fournié-Zaluski MC, Roques B, Checler F. Aminopeptidase A contributes to the N-terminal truncation of amyloid beta-peptide. J Neurochem 2009; 109:248-56. [PMID: 19187443 DOI: 10.1111/j.1471-4159.2009.05950.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Several lines of data previously indicated that N-terminally truncated forms of amyloid-beta (Abeta) peptides are likely the earliest and more abundant species immunohistochemically detectable in Alzheimer's disease-affected brains. It is noteworthy that the free N-terminal residue of full-length Abeta (fl-Abeta) is an aspartyl residue, suggesting that Abeta could be susceptible to exopeptidasic attack by aminopeptidase A (APA)-like proteases. In this context, we have examined whether APA could target Abeta peptides in both cell-free and cellular models. We first show that the general aminopeptidase inhibitor amastatin as well as two distinct aminopeptidase A inhibitors EC33 and pl302 both significantly increase the recovery of genuine fl-Abeta peptides generated by cells over-expressing Swedish-mutated beta amyloid precursor protein (APP) while the aminopeptidase N blocker pl250 did not modify fl-Abeta recovery. In agreement with this observation, we establish that over-expressed APA drastically reduces, in a calcium dependent manner, fl-Abeta but not APP IntraCellular Domain in a cell-free model of Abeta production. In agreement with the above data, we show that recombinant APA degrades fl-Abeta in a pl302-sensitive manner. Interestingly, we also show that EC33 and pl302 lower staurosporine-stimulated activation of caspase-3 in wild-type fibroblasts but not in betaAPP/beta-amyloid precursor protein-like protein 2 (APLP2) double knockout fibroblasts, suggesting that protecting endogenous fl-Abeta physiological production triggers neuroprotective phenotype. By contrast, EC33 does not modify staurosporine-induced caspase-3 activation in wild-type and Swedish-mutated betaAPP-HEK293 expressing cells that display exacerbated production of Abeta. Overall, our data establish that APA contributes to the N-terminal truncation of Abeta and suggest that this cleavage is likely abrogating a protective function associated with physiological but not supraphysiological levels of genuine fl-Abeta peptides.
Collapse
Affiliation(s)
- Jean Sevalle
- Institut de Pharmacologie Moléculaire et Cellulaire and Institut de NeuroMédecine Moléculaire, UMR6097 CNRS/UNSA, Equipe labellisée Fondation pour la Recherche Médicale, Sophia-Antipolis, Valbonne, France
| | | | | | | | | | | |
Collapse
|
11
|
Dolcini V, Dunys J, Sevalle J, Chen F, Guillot-Sestier MV, St George-Hyslop P, Fraser PE, Checler F. TMP21 regulates Abeta production but does not affect caspase-3, p53, and neprilysin. Biochem Biophys Res Commun 2008; 371:69-74. [PMID: 18405662 DOI: 10.1016/j.bbrc.2008.03.151] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2008] [Accepted: 03/31/2008] [Indexed: 01/09/2023]
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.
Collapse
Affiliation(s)
- Virginia Dolcini
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR 6097, Centre National de la Recherche Scientifique-Université Nice-Sophia-Antipolis, Equipe labellisée Fondation pour la Recherche Médicale, 660 Route des Lucioles, 06560 Valbonne, France
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Buggia-Prevot V, Sevalle J, Rossner S, Checler F. NFκB-dependent Control of BACE1 Promoter Transactivation by Aβ42. J Biol Chem 2008; 283:10037-47. [DOI: 10.1074/jbc.m706579200] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
13
|
Lefranc-Jullien S, Sunyach C, Checler F. APPepsilon, the epsilon-secretase-derived N-terminal product of the beta-amyloid precursor protein, behaves as a type I protein and undergoes alpha-, beta-, and gamma-secretase cleavages. J Neurochem 2006; 97:807-17. [PMID: 16524370 DOI: 10.1111/j.1471-4159.2006.03748.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
beta-Amyloid peptide accumulates in the brain of patients affected by sporadic or familial forms of Alzheimer's disease. It derives from the proteolytic attacks of the beta-amyloid precursor protein (betaAPP) by beta- and gamma-secretase activities. An additional epsilon cleavage taking place a few residues C-terminal to the gamma-site has been reported, leading to the formation of an intracellular fragment referred to as APP intracellular domain C50. This epsilon cleavage received particular attention because it resembles the S3 Notch cleavage generating Notch intracellular domain. Indeed, APP intracellular domain, like its Notch counterpart, appears to mediate important physiological functions. gamma and epsilon cleavages on betaAPP appear spatio-temporally linked but pharmacologically distinct and discriminable by mutagenesis approaches. As these cleavages could be seen as either deleterious (gamma-site) or beneficial (epsilon-site), it appears of most interest to set up models aimed at studying these activities separately, particularly to design specific and bioavailable inhibitors. On the other hand, it is important to respect the topology of the substrates in order to examine physiologically relevant cleavages. Here we describe the obtention of cells overexpressing APPepsilon, the epsilon-secretase-derived N-terminal fragment of betaAPP. Interestingly, this N-terminal fragment of betaAPP was shown by biochemical and immunohistochemical approaches to behave as a genuine membrane-bound protein. APPepsilon undergoes constitutive and protein kinase C-regulated alpha-secretase cleavages. Furthermore, APPepsilon is targeted by the beta-secretase beta-site APP-cleaving enzyme and is subsequently cleaved by gamma-secretase. The resulting beta-amyloid peptide production is fully prevented by various gamma-secretase inhibitors. Altogether, our study shows that APPepsilon is a relevant betaAPP derivative to study gamma-secretase activities and to design specific inhibitors without facing any rate-limiting effect of epsilon-secretase-derived cleavage.
Collapse
Affiliation(s)
- Solveig Lefranc-Jullien
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR6097 CNRS/UNSA, Equipe labellisée Fondation pour la Recherche Médicale, Valbonne, France
| | | | | |
Collapse
|
14
|
Désiré L, Bourdin J, Loiseau N, Peillon H, Picard V, De Oliveira C, Bachelot F, Leblond B, Taverne T, Beausoleil E, Lacombe S, Drouin D, Schweighoffer F. RAC1 inhibition targets amyloid precursor protein processing by gamma-secretase and decreases Abeta production in vitro and in vivo. J Biol Chem 2005; 280:37516-25. [PMID: 16150730 DOI: 10.1074/jbc.m507913200] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
beta-Amyloid peptides (Abeta) that form the senile plaques of Alzheimer disease consist mainly of 40- and 42-amino acid (Abeta 40 and Abeta 42) peptides generated from the cleavage of the amyloid precursor protein (APP). Generation of Abeta involves beta-secretase and gamma-secretase activities and is regulated by membrane trafficking of the proteins involved in Abeta production. Here we describe a new small molecule, EHT 1864, which blocks the Rac1 signaling pathways. In vitro, EHT 1864 blocks Abeta 40 and Abeta 42 production but does not impact sAPPalpha levels and does not inhibit beta-secretase. Rather, EHT 1864 modulates APP processing at the level of gamma-secretase to prevent Abeta 40 and Abeta 42 generation. This effect does not result from a direct inhibition of the gamma-secretase activity and is specific for APP cleavage, since EHT 1864 does not affect Notch cleavage. In vivo, EHT 1864 significantly reduces Abeta 40 and Abeta 42 levels in guinea pig brains at a threshold that is compatible with delaying plaque accumulation and/or clearing the existing plaque in brain. EHT 1864 is the first derivative of a new chemical series that consists of candidates for inhibiting Abeta formation in the brain of AD patients. Our findings represent the first pharmacological validation of Rac1 signaling as a target for developing novel therapies for Alzheimer disease.
Collapse
|
15
|
Lefranc-Jullien S, Lisowski V, Hernandez JF, Martinez J, Checler F. Design and characterization of a new cell-permeant inhibitor of the beta-secretase BACE1. Br J Pharmacol 2005; 145:228-35. [PMID: 15753947 PMCID: PMC1576132 DOI: 10.1038/sj.bjp.0706183] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1 The beta-secretase BACE1 is one of the enzymes that contribute to the production of the Abeta peptide, in vitro and in vivo. JMV1195 was previously shown to inhibit BACE activity in vitro but was unable to block cellular BACE activity. We have designed a new permeable inhibitor, JMV2764 that corresponds to a derivative of JMV1195 to which a penetratin sequence had been added at its N-terminus. We have assessed the ability of JMV2764 to affect BACE1 activity in vitro, and to modify Abeta production in various cell systems. 2 Endogenous beta-secretase or BACE1 activities were monitored in vitro by means of two distinct fluorimetric substrates in HEK293 extracts of cells expressing either wild-type betaAPP, Swedish mutated betaAPP or SPA4CT constructs. Abeta40 recovery was monitored by immunoprecipitation and Western blot analysis. 3 JMV2764 and JMV1195 inhibited endogenous beta-secretase activity of HEK293 cellular homogenates with IC(50)s of 0.8 and 6.6 microM, respectively. Interestingly, JMV2764 also inhibited beta-secretase activity after preincubation with intact cells while JMV1195 was inactive, indicating that unlike JMV1195, JMV2764 could penetrate into the cells. 4 JMV2764 but not JMV1195 also prevented Abeta production by HEK293 cells overexpressing wild-type and Swedish-mutated betaAPP. However, JMV2764 was unable to affect Abeta production from cells expressing SPA4CT, a betaAPP-derived sequence that does not need beta-secretase to produce Abeta. 5 Altogether, we have designed a new cell-permeable BACE1 inhibitor that allows to envision to prevent Abeta production in vivo. Work is in progress to assess the potential of these compounds to prevent Abeta production in transgenic mice overproducing Abeta.
Collapse
Affiliation(s)
| | | | | | | | - Frédéric Checler
- IPMC of CNRS, UMR6097, CNRS/UNSA, 660 Route des Lucioles, 06560 Valbonne, France
- Author for correspondence:
| |
Collapse
|
16
|
Andrau D, Dumanchin-Njock C, Ayral E, Vizzavona J, Farzan M, Boisbrun M, Fulcrand P, Hernandez JF, Martinez J, Lefranc-Jullien S, Checler F. BACE1- and BACE2-expressing human cells: characterization of beta-amyloid precursor protein-derived catabolites, design of a novel fluorimetric assay, and identification of new in vitro inhibitors. J Biol Chem 2003; 278:25859-66. [PMID: 12736275 DOI: 10.1074/jbc.m302622200] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have set up stably transfected HEK293 cells overexpressing the beta-secretases BACE1 and BACE2 either alone or in combination with wild-type beta-amyloid precursor protein (betaAPP). The characterization of the betaAPP-derived catabolites indicates that cells expressing BACEs produce less genuine Abeta1- 40/42 but higher amounts of secreted sAPPbeta and N-terminal-truncated Abeta species. This was accompanied by a concomitant modulation of the C-terminal counterpart products C89 and C79 for BACE1 and BACE2, respectively. These cells were used to set up a novel BACE assay based on two quenched fluorimetric substrates mimicking the wild-type (JMV2235) and Swedish-mutated (JMV2236) betaAPP sequences targeted by BACE activities. We show that BACEs activities are enhanced by the Swedish mutation and maximal at pH 4.5. The specificity of this double assay for genuine beta-secretase activity was demonstrated by means of cathepsin D, a "false positive" BACE candidate. Thus, cathepsin D was unable to cleave preferentially the JMV2236-mutated substrate. The selectivity of the assay was also emphasized by the lack of JMV cleavage triggered by other "secretases" candidates such as ADAM10 (A disintegrin and metalloprotease 10), tumor necrosis alpha-converting enzyme, and presenilins 1 and 2. Finally, the assay was used to screen for putative in vitro BACE inhibitors. We identified a series of statine-derived sequences that dose-dependently inhibited BACE1 and BACE2 activities with IC50 in the micromolar range, some of which displaying selectivity for either BACE1 or BACE2.
Collapse
Affiliation(s)
- David Andrau
- Institut de Pharmacologie Moléculaire et Cellulaire of Centre National de la Recherche Scientifique, UMR6097, 06560 Valbonne, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
Peptidases play a vital and often highly specific role in the physiological and pathological generation and termination of peptide hormone signals. The thermolysin-like family of metalloendopeptidases involved in the extracellular processing of neuroendocrine and cardiovascular peptides are of particular significance, reflecting both their specificity for particular peptide substrates and their utility as therapeutic targets. Although the functions of the membrane-bound members of this family, such as angiotensin-converting enzyme and neutral endopeptidase, are well established, a role for the predominantly soluble family members in peptide metabolism is only just emerging. This review will focus on the biochemistry, cell biology, and physiology of the soluble metalloendopeptidases EC 3.4.24.15 (thimet oligopeptidase) and EC 3.4.24.16 (neurolysin), as well as presenting evidence that both peptidases play an important role in such diverse functions as reproduction, nociception, and cardiovascular homeostasis.
Collapse
|
18
|
Lopez-Perez E, Dumanchin C, Czech C, Campion D, Goud B, Pradier L, Frebourg T, Checler F. Overexpression of Rab11 or constitutively active Rab11 does not affect sAPPalpha and Abeta secretions by wild-type and Swedish mutated betaAPP-expressing HEK293 cells. Biochem Biophys Res Commun 2000; 275:910-5. [PMID: 10973821 DOI: 10.1006/bbrc.2000.3404] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Presenilins 1 and 2 are two homologous proteins which, when mutated, appear responsible for most of the early-onset familial forms of Alzheimer's disease. Among various functional aspects, presenilins appear to behave as chaperoning partners of a series of proteins including the beta-amyloid precursor protein. Recently, presenilins were shown to interact with Rab11, a GTPase involved in intracellular transport. This suggested that Rab11-presenilin interaction could influence the routing of betaAPP and thereby modulate its maturation. In this context, we examined whether overexpression of Rab11 or its constitutively active mutant Rab11Q70L could affect betaAPP maturation in human HEK293 cells. We show here that the overexpression of both Rab11-related proteins does not modify the recovery of secreted sAPPalpha or Abeta in HEK293 cells expressing wild-type betaAPP or betaAPP harboring the Swedish double mutation. These data indicate that Rab11 does not influence betaAPP processing in HEK293 cells. However, it does not preclude the possibility for Rab11 to modulate other presenilin-mediated functions in human cells.
Collapse
Affiliation(s)
- E Lopez-Perez
- IPMC du CNRS, UPR411, 660 Route des Lucioles, Valbonne, 06560, France
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Petit A, Barelli H, Morain P, Checler F. Novel proline endopeptidase inhibitors do not modify Abeta40/42 formation and degradation by human cells expressing wild-type and swedish mutated beta-amyloid precursor protein. Br J Pharmacol 2000; 130:1613-7. [PMID: 10928965 PMCID: PMC1572208 DOI: 10.1038/sj.bjp.0703440] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2000] [Revised: 04/17/2000] [Accepted: 05/02/2000] [Indexed: 11/09/2022] Open
Abstract
Previous studies have suggested that proline endopeptidase (PE) could participate to the catabolism of the beta-amyloid peptide (Abeta) or to the physiopathological maturation of the beta-amyloid protein precursor (betaAPP). We have examined the putative ability of human purified PE to catabolize Abeta40 and Abeta42 and the possible contribution of this enzyme to the generation of Abeta40 and Abeta42 in human HEK293 cells. We show first that purified human PE does not degrade synthetic Abeta40 and Abeta42, in vitro. We establish that HEK293 cell homogenates exhibit a Z-Gly-Pro-7AMC-cleaving enzyme, the activity of which is inhibited by Z-Pro-Prolinal and S17092 and S19825, two novel PE inhibitors, with affinities similar to those displayed on the purified human PE. These inhibitors also penetrate cells and achieve a full inhibition of endogenous proline endopeptidase in human cells. By means of selective antibodies directed towards the C-terminal of Abeta40 and Abeta42, we assessed the effect of PE inhibitors on the recovery of both Abeta species. This was examined in HEK293 cells stably overexpressing the wild-type and the familial Alzheimer's disease-related Swedish mutated beta-APP. We establish that none of these inhibitors affected Abeta40 or Abeta42 production in these transfected cells. Overall, our study indicates that human PE does not degrade Abeta40 and Abeta42. Furthermore, PE does not contribute to Abeta40 and Abeta42 formation in HEK293 cells. Therefore, PE does not appear to contribute to the Abeta-related aetiology of Alzheimer's disease.
Collapse
Affiliation(s)
- Agnès Petit
- Institut de Pharmacologie Moléculaire et Cellulaire, UPR411 du CNRS, 660 route des lucioles, Sophia Antipolis, 06560 Valbonne, France
| | - Hélène Barelli
- Institut de Pharmacologie Moléculaire et Cellulaire, UPR411 du CNRS, 660 route des lucioles, Sophia Antipolis, 06560 Valbonne, France
| | | | - Frédéric Checler
- Institut de Pharmacologie Moléculaire et Cellulaire, UPR411 du CNRS, 660 route des lucioles, Sophia Antipolis, 06560 Valbonne, France
| |
Collapse
|
20
|
Yamin R, Malgeri EG, Sloane JA, McGraw WT, Abraham CR. Metalloendopeptidase EC 3.4.24.15 is necessary for Alzheimer's amyloid-beta peptide degradation. J Biol Chem 1999; 274:18777-84. [PMID: 10373494 DOI: 10.1074/jbc.274.26.18777] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
We have investigated the functional relationship between metalloendopeptidase EC 3.4.24.15 (MP24.15) and the amyloid precursor protein involved in Alzheimer's disease (AD) and discovered that the enzyme promotes Abeta degradation. We show here that conditioned medium (CM) of MP24.15 antisense-transfected SKNMC neuroblastoma has significantly higher levels of Abeta. Furthermore, synthetic-Abeta degradation was increased or decreased following incubation with CM of sense or antisense-transfected cells, respectively. Soluble Abeta1-42 was degraded more slowly than soluble Abeta1-40, while aggregated Abeta1-42 showed almost no degradation. Pretreatment of CM with serine proteinase inhibitors 4-(2-aminoethyl)benzenesulfonyl fluoride and diisopropyl fluorophosphate completely inhibited Abeta degradation. Additionally, alpha1-antichymotrypsin (ACT), a serpin family inhibitor tightly associated with plaques and elevated in AD brain, blocked up to 60% of Abeta degradation. Interestingly, incubation of CM of MP24. 15-overexpressing cells with ACT formed an SDS-resistant ACT complex, suggesting an ACT-serine proteinase interaction. Recombinant MP24. 15 alone did not degrade Abeta. 14C-Diisopropyl fluorophosphate-radiolabeled CM from MP24.15-overexpressing cells contained increased levels of several active serine proteinases, suggesting that MP24.15 activates one or more Abeta-degrading serine proteases. Thus, ACT may cause Abeta accumulation by inhibiting an Abeta-degrading enzyme or by direct binding to Abeta, rendering it degradation-resistant. Identification of the Abeta-degrading enzyme and MP24.15's role in its activation is underway. Pharmacological modulation of either enzyme may provide a means of regulating Abeta in the brain.
Collapse
Affiliation(s)
- R Yamin
- Department of Biochemistry and Medicine, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | | | | | |
Collapse
|
21
|
Barelli H, Petit A, Hirsch E, Wilk S, De Nanteuil G, Morain P, Checler F. S 17092-1, a highly potent, specific and cell permeant inhibitor of human proline endopeptidase. Biochem Biophys Res Commun 1999; 257:657-61. [PMID: 10208839 DOI: 10.1006/bbrc.1999.0366] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Several lines of evidence indicate that proline endopeptidase (PE) could participate to the symptomatology and/or etiology of Alzheimer's disease. Thus, proline endopeptidase appears to contribute to the degradation of neuropeptides involved in learning and memory and could also control the production of the amyloidogenic peptide Abeta. Therefore the design of potent, selective and permeant inhibitors of human PE should lead to potential probes to assess the genuine contribution of this enzyme in Alzheimer's pathology. A novel perhydroindol carboxylic derivative, S17092-1 inhibits the hydrolysis of Z-Gly-Pro-7AMC-hydrolysing activity present in human brain nuclei with a high affinity (Ki = 1 nM) and behaves as a highly potent (Ki = 1.5 nM) inhibitor of partially purified human PE. By contrast, S17092-1 is unable to affect a series of other peptidases including aminopeptidases B and M, dipeptidylaminopeptidase IV, endopeptidases 3.4.24.11, 3.4.24.15, 3.4.24.16, calpains and angiotensin-converting enzyme. Furthermore, we show that the embryonic human kidney 293 cell line displays an intracellular PE-like activity that is blocked after preincubating cells with S17092-1, indicating that this inhibitor penetrates in HEK293 cells and could affect intracellular human PE. Altogether, we establish that S17092-1 behaves as a highly potent, specific and cell permeant inhibitor of human proline endopeptidase and can be seen as a probe to examine PE contribution in Alzheimer's disease.
Collapse
Affiliation(s)
- H Barelli
- IPMC du CNRS, UPR411, 660 Route des Lucioles, Valbonne, 06560, France
| | | | | | | | | | | | | |
Collapse
|
22
|
Marambaud P, Ancolio K, Alves da Costa C, Checler F. Effect of protein kinase A inhibitors on the production of Abeta40 and Abeta42 by human cells expressing normal and Alzheimer's disease-linked mutated betaAPP and presenilin 1. Br J Pharmacol 1999; 126:1186-90. [PMID: 10205007 PMCID: PMC1565882 DOI: 10.1038/sj.bjp.0702406] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/1998] [Revised: 11/27/1998] [Accepted: 12/07/1998] [Indexed: 11/09/2022] Open
Abstract
1. We previously established that the formation of both alpha- and beta/gamma-secretase-derived products generated by human embryonic kidney 293 cells (HEK293) expressing either wild type or mutant betaAPP could be stimulated by agonists of the cyclic AMP/protein kinase A pathways. This cyclic AMP-dependent effect modulates post-translational events since it is not prevented by actinomycin D or cycloheximide. 2. We show here that two protein kinase A inhibitors, H89 and PKI, both trigger dose-dependent inhibition of the basal constitutive production of Abeta40 and Abeta42 by HEK293 cells expressing wild type betaAPP751. 3. H89 also potently inhibits the total Abeta produced by the neocortical neuronal cell line TSM1. 4. These two inhibitors also drastically reduce the recovery of Abeta40 and Abeta42 produced by HEK293 cells expressing the Swedish (Sw) betaAPP and M146V-presenilin 1 (PS1) mutations responsible for cases of the early-onset forms of Familial Alzheimer's disease (FAD). 5. By contrast, H89 and PKI do not significantly affect the recovery of the physiological alpha-secretase-derived fragment APPalpha. 6. Our study indicates that protein kinase A inhibitors selectively lower the formation of Abeta40 and Abeta42 in human cells expressing normal and mutant betaAPP and PS1 without affecting the physiological alpha-secretase pathway in these cells. Selective inhibitors of protein kinase A may be of therapeutic value in both sporadic and Familial Alzheimer's disease, since they may decrease the production of Abeta that is thought to be responsible for the neurodegenerative process.
Collapse
Affiliation(s)
- P Marambaud
- Institut de Pharmacologie Moléculaire et Cellulaire du CNRS, UPR411, 660 Route des Lucioles, Sophia Antipolis, 06560 Valbonne, France
| | - K Ancolio
- Institut de Pharmacologie Moléculaire et Cellulaire du CNRS, UPR411, 660 Route des Lucioles, Sophia Antipolis, 06560 Valbonne, France
| | - C Alves da Costa
- Institut de Pharmacologie Moléculaire et Cellulaire du CNRS, UPR411, 660 Route des Lucioles, Sophia Antipolis, 06560 Valbonne, France
| | - F Checler
- Institut de Pharmacologie Moléculaire et Cellulaire du CNRS, UPR411, 660 Route des Lucioles, Sophia Antipolis, 06560 Valbonne, France
| |
Collapse
|
23
|
Marambaud P, Chevallier N, Ancolio K, Checler F. Post-transcriptional contribution of a cAMP-dependent pathway to the formation of alpha- and beta/gamma-secretases-derived products of beta APP maturation in human cells expressing wild-type and Swedish mutated beta APP. Mol Med 1998; 4:715-23. [PMID: 9932109 PMCID: PMC2230337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND The physiopathological maturation of the beta-amyloid precursor protein can be modulated by effectors targeting a protein kinase C-dependent pathway. These agents increase the recovery of APP alpha, the physiological alpha-secretase-derived product of beta APP processing, and concomittantly lower the production of the pathogenic beta/gamma-secretase-derived A beta fragment. METHODS We set up stably transfected HEK293 cells expressing wild-type or Swedish mutated beta APP. By combined metabolic labeling and/or immunoprecipitation procedures, we assessed the effect of various cAMP effectors on the production of the beta APP maturation products A beta 40, A beta 42, APP alpha, and its C-terminal counterpart. RESULTS We show here that the cAMP-dependent protein kinase (PKA) effectors, dibutyryl-cAMP (dBut-cAMP) and forskolin, but not the inactive analog dideoxyforskolin, enhance the secretion of APP alpha and the intracellular production of its C-terminal counterpart (p10) in stably transfected HEK293 cells. The above agonists also drastically increase both A beta 40 and A beta 42 secretions and intracellular A beta recovery. The same influence was observed with HEK293 cells overexpressing the Swedish mutated beta APP. We attempted to delineate the relative contribution of transcriptional and post-transcriptional events in the cAMP-mediated response. We show here that the dBut-cAMP and forskolin-induced increase of APP alpha and A beta s secretions is not prevented by the transcription inhibitor actinomycin D. CONCLUSION Our data suggest a major contribution of post-transcriptional events in the cAMP-dependent effect on beta APP maturation. It appears likely that cAMP triggers the PKA-dependent phosphorylation of a protein involved in beta APP maturation and occurring upstream to alpha- and beta/gamma-secretase cleavages.
Collapse
Affiliation(s)
- P Marambaud
- Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique, Université de Nice-Sophia Antipolis, Valbonne, France
| | | | | | | |
Collapse
|
24
|
Marambaud P, Chevallier N, Ancolio K, Checler F. Post-transcriptional Contribution of a cAMP-dependent Pathway to the Formation of α- and β/γ-Secretases-Derived Products of βAPP Maturation in Human Cells Expressing Wild-type and Swedish Mutated βAPP. Mol Med 1998. [DOI: 10.1007/bf03401766] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
25
|
Torres MP, Prange C, Lennon G. Human endopeptidase 24.15 (THOP1) is localized on chromosome 19p13.3 and is excluded from the linkage region for late-onset Alzheimer disease. Genomics 1998; 53:239-40. [PMID: 9790774 DOI: 10.1006/geno.1998.5487] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The mapping position of human endopeptidase 24.15 (THOP1) has previously been reported to be within the linkage region for the late-onset Alzheimer disease AD2 locus on chromosome 19q13.3. After localizing THOP1 to the high-resolution cosmid contig map of human chromosome 19, we found that the previous report was incorrect. Results of the hybridization and FISH mapping of positive clones indicated localization of THOP1 to chromosome 19p13.3 and not 19q13. 3. This localization is a correction of wrong chromosomal delegation and excludes THOP1 from the region that shows evidence of linkage to late-onset familial Alzheimer disease.
Collapse
Affiliation(s)
- M P Torres
- Biology and Biotechnology Program, Lawrence Livermore National Laboratory, Livermore, California, 94550, USA.
| | | | | |
Collapse
|
26
|
Marambaud P, Ancolio K, Lopez-Perez E, Checler F. Proteasome inhibitors prevent the degradation of familial Alzheimer's disease-linked presenilin 1 and potentiate A beta 42 recovery from human cells. Mol Med 1998; 4:147-57. [PMID: 9562973 PMCID: PMC2230353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Several lines of evidence suggest that most of the early-onset forms of familial Alzheimer's disease (FAD) are due to inherited mutations borne by a chromosome 14-encoded protein, presenilin 1 (PS1). This is likely related to an increased production of amyloid beta-peptide (A beta) 42, one of the main components of the extracellular deposits called senile plaques that invade human cortical areas during the disease. MATERIALS AND METHODS We set up stably transfected HEK293 cells overexpressing wild-type (wt) and various FAD-linked mutated PS1. By Western blot analysis, we examined the influence of specific proteasome inhibitors on PS1-like immunoreactivities. Furthermore, by means of metabolic labeling and immunoprecipitation with A beta 40 and A beta 42-directed specific antibodies, we assessed the effect of the inhibitors on the production of A beta s by wt and mutated PS1-expressing cells transiently transfected with beta APP751. RESULTS We show that two distinct proteasome inhibitors, Z-IE (Ot-Bu)A-Leucinal and lactacystin, increase in a time- and dose-dependent manner the immunoreactivities of both wt and mutated PS1. Furthermore, we demonstrate that PS1 is polyubiquitinated in these cells. Other inhibitors, ineffective on the proteasome, fail to protect wt and mutated PS1-like immunoreactivities. We also establish that the FAD-linked mutations of PS1 trigger a selective increased formation of A beta 42 as reflected by higher A beta 42 over total A beta ratios when compared with wtPS1-expressing cells. Interestingly, this augmentation was further amplified by proteasome inhibitors in cells expressing mutated but not wtPS1. CONCLUSION Altogether, our data indicate that PS1 undergoes polyubiquitination in HEK293 cells and that the proteasome contributes to the degradation of wt and FAD-linked PS1, thereby directly influencing the A beta production in human cells.
Collapse
Affiliation(s)
- P Marambaud
- Institut de Pharmacologie Moléculaire et Cellulaire, UPR 411 du CNRS, Sophia Antipolis, Valbonne, France
| | | | | | | |
Collapse
|
27
|
Marambaud P, Ancolio K, Lopez-Perez E, Checler F. Proteasome Inhibitors Prevent the Degradation of Familial Alzheimer’s Disease-Linked Presenilin 1 and Potentiate Aβ42 Recovery from Human Cells. Mol Med 1998. [DOI: 10.1007/bf03401912] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
28
|
Barelli H, Lebeau A, Vizzavona J, Delaere P, Chevallier N, Drouot C, Marambaud P, Ancolio K, Buxbaum JD, Khorkova O, Heroux J, Sahasrabudhe S, Martinez J, Warter JM, Mohr M, Checler F. Characterization of new polyclonal antibodies specific for 40 and 42 amino acid-long amyloid beta peptides: their use to examine the cell biology of presenilins and the immunohistochemistry of sporadic Alzheimer's disease and cerebral amyloid angiopathy cases. Mol Med 1997; 3:695-707. [PMID: 9392006 PMCID: PMC2230230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND In Alzheimer's disease (AD), the main histological lesion is a proteinaceous deposit, the senile plaque, which is mainly composed of a peptide called A beta. The aggregation process is thought to occur through enhanced concentration of A beta 40 or increased production of the more readily aggregating 42 amino acid-long A beta 42 species. MATERIALS AND METHODS Specificity of the antibodies was assessed by dot blot, Western blot, ELISA, and immunoprecipitation procedures on synthetic and endogenous A beta produced by secreted HK293 cells. A beta and p3 production by wild-type and mutated presenilin 1-expressing cells transiently transfected with beta APP751 was monitored after metabolic labeling and immunoprecipitation procedures. Immunohistochemical analysis was performed on brains of sporadic and typical cerebrovascular amyloid angiopathy (CAA) cases. RESULTS Dot and Western blot analyses indicate that IgG-purified fractions of antisera recognize native and denaturated A beta s. FCA3340 and FCA 3542 display full specificity for A beta 40 and A beta 42, respectively. Antibodies immunoprecipitate their respective synthetic A beta species but also A beta s and their related p3 counterparts endogenously secreted by transfected human kidney 293 cells. This allowed us to show that mutations on presenilin 1 triggered similar increased ratios of A beta 42 and its p 342 counterpart over total A beta and p3. ELISA assays allow detection of about 25-50 pg/ml of A beta s and remain linear up to 750 to 1500 pg/ml without any cross-reactivity. FCA18 and FCA3542 label diffuse and mature plaques of a sporadic AD case whereas FCA3340 only reveals the mature lesions and particularly labels their central dense core. In a CAA case, FCA18 and FCA3340 reveal leptomeningeal and cortical arterioles whereas FCA3542 only faintly labels such structures. CONCLUSIONS Polyclonal antibodies exclusively recognizing A beta 40 (FCA 3340) or A beta 42 (FCA3542) were obtained. These demonstrated that FAD-linked presenilins similarly affect both p342 and A beta 42, suggesting that these mutations misroute the beta APP to a compartment where gamma-secretase, but not alpha-secretase, cleavages are modified. Overall, these antibodies should prove useful for fundamental and diagnostic approaches, as suggested by their usefulness for biochemical, cell biological, and immunohistochemical techniques.
Collapse
Affiliation(s)
- H Barelli
- IPMC du CNRS, UPR411, Valbonne, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Barelli H, Lebeau A, Vizzavona J, Delaere P, Chevallier N, Drouot C, Marambaud P, Ancolio K, Buxbaum JD, Khorkova O, Heroux J, Sahasrabudhe S, Martinez J, Warter JM, Mohr M, Checler F. Characterization of New Polyclonal Antibodies Specific for 40 and 42 Amino Acid-Long Amyloid β Peptides: Their Use to Examine the Cell Biology of Presenilins and the Immunohistochemistry of Sporadic Alzheimer’s Disease and Cerebral Amyloid Angiopathy Cases. Mol Med 1997. [DOI: 10.1007/bf03401708] [Citation(s) in RCA: 110] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|