Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 23
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Neurotherapeutics ; 18(2): 1137-1150, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-33533011

RÉSUMÉ

Prion diseases are caused by the propagation of PrPSc, the pathological conformation of the PrPC prion protein. The molecular mechanisms underlying PrPSc propagation are still unsolved and no therapeutic solution is currently available. We thus sought to identify new anti-prion molecules and found that flunarizine inhibited PrPSc propagation in cell culture and significantly prolonged survival of prion-infected mice. Using an in silico therapeutic repositioning approach based on similarities with flunarizine chemical structure, we tested azelastine, duloxetine, ebastine, loperamide and metixene and showed that they all have an anti-prion activity. Like flunarizine, these marketed drugs reduced PrPSc propagation in cell culture and in mouse cerebellum organotypic slice culture, and inhibited the protein folding activity of the ribosome (PFAR). Strikingly, some of these drugs were also able to alleviate phenotypes due to PABPN1 nuclear aggregation in cell and Drosophila models of oculopharyngeal muscular dystrophy (OPMD). These data emphasize the therapeutic potential of anti-PFAR drugs for neurodegenerative and neuromuscular proteinopathies.


Sujet(s)
Systèmes de délivrance de médicaments/méthodes , Flunarizine/administration et posologie , Protéine-1 de liaison au poly(A)/métabolisme , Maladies à prions/métabolisme , Agrégats de protéines/effets des médicaments et des substances chimiques , Pliage des protéines/effets des médicaments et des substances chimiques , Animaux , Inhibiteurs des canaux calciques/administration et posologie , Lignée cellulaire , Bases de données factuelles , Drosophila , Femelle , Souris , Souris transgéniques , Techniques de culture d'organes , Protéine-1 de liaison au poly(A)/antagonistes et inhibiteurs , Protéine-1 de liaison au poly(A)/génétique , Maladies à prions/traitement médicamenteux , Maladies à prions/génétique , Protéines prion/antagonistes et inhibiteurs , Protéines prion/génétique , Protéines prion/métabolisme , Agrégats de protéines/physiologie , Ovis
2.
Int J Mol Sci ; 21(23)2020 Dec 02.
Article de Anglais | MEDLINE | ID: mdl-33276687

RÉSUMÉ

Studies on the cellular prion protein (PrPC) have been actively conducted because misfolded PrPC is known to cause transmissible spongiform encephalopathies or prion disease. PrPC is a glycophosphatidylinositol-anchored cell surface glycoprotein that has been reported to affect several cellular functions such as stress protection, cellular differentiation, mitochondrial homeostasis, circadian rhythm, myelin homeostasis, and immune modulation. Recently, it has also been reported that PrPC mediates tumor progression by enhancing the proliferation, metastasis, and drug resistance of cancer cells. In addition, PrPC regulates cancer stem cell properties by interacting with cancer stem cell marker proteins. In this review, we summarize how PrPC promotes tumor progression in terms of proliferation, metastasis, drug resistance, and cancer stem cell properties. In addition, we discuss strategies to treat tumors by modulating the function and expression of PrPC via the regulation of HSPA1L/HIF-1α expression and using an anti-prion antibody.


Sujet(s)
Tumeurs/métabolisme , Protéines prion/métabolisme , Antinéoplasiques/administration et posologie , Antinéoplasiques/effets indésirables , Antinéoplasiques/usage thérapeutique , Marqueurs biologiques tumoraux , Prolifération cellulaire , Prise en charge de la maladie , Résistance aux médicaments antinéoplasiques , Espace intracellulaire/métabolisme , Thérapie moléculaire ciblée , Métastase tumorale , Stadification tumorale , Tumeurs/traitement médicamenteux , Tumeurs/étiologie , Tumeurs/anatomopathologie , Cellules souches tumorales/métabolisme , Protéines PrPC/métabolisme , Protéines prion/antagonistes et inhibiteurs
3.
J Biol Chem ; 295(39): 13516-13531, 2020 09 25.
Article de Anglais | MEDLINE | ID: mdl-32723867

RÉSUMÉ

Prion disease is a rapidly progressive neurodegenerative disorder caused by misfolding and aggregation of the prion protein (PrP), and there are currently no therapeutic options. PrP ligands could theoretically antagonize prion formation by protecting the native protein from misfolding or by targeting it for degradation, but no validated small-molecule binders have been discovered to date. We deployed a variety of screening methods in an effort to discover binders of PrP, including 19F-observed and saturation transfer difference (STD) NMR spectroscopy, differential scanning fluorimetry (DSF), DNA-encoded library selection, and in silico screening. A single benzimidazole compound was confirmed in concentration-response, but affinity was very weak (Kd > 1 mm), and it could not be advanced further. The exceptionally low hit rate observed here suggests that PrP is a difficult target for small-molecule binders. Whereas orthogonal binder discovery methods could yield high-affinity compounds, non-small-molecule modalities may offer independent paths forward against prion disease.


Sujet(s)
Benzimidazoles/pharmacologie , Maladies à prions/traitement médicamenteux , Protéines prion/antagonistes et inhibiteurs , Bibliothèques de petites molécules/pharmacologie , Benzimidazoles/composition chimique , Découverte de médicament , Évaluation préclinique de médicament , Humains , Spectroscopie par résonance magnétique , Maladies à prions/métabolisme , Protéines prion/métabolisme , Bibliothèques de petites molécules/composition chimique
4.
Eur J Med Chem ; 196: 112295, 2020 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-32325366

RÉSUMÉ

Prion diseases or transmissible spongiform encephalopathies (TSEs) are a group of rare neurodegenerative disorders. TSEs are characterized by the accumulation of prions (PrPSc) that represent pathological isoforms of the physiological cellular prion protein PrPC. Although the conversion of PrPC to PrPSc is still not completely understood, blocking this process may lead to develop new therapies. Here, we have generated a pharmacophore model, based on anti-prion molecules reported in literature to be effective in phenotypic assay. The model was used to conduct a virtual screen of commercial compound databases that selected a small library of ten compounds. These molecules were then screened in mouse neuroblastoma cell line chronically infected with prions (ScN2a) after excluding neurotoxicity. 1 has been identified as the therapeutic hit on the basis of the following evidence: chronic treatments of ScN2a cells using 1 eliminate PrPSc loaded in both Western blotting analysis and Real-Time Quaking-Induced Conversion (RT-QuIC) assay. We also proposed the mechanism of action of 1 by which it has the ability to bind PrPC and consequentially blocks prion conversion. Herein we describe the results of these efforts.


Sujet(s)
Phénothiazines/pharmacologie , Protéines prion/antagonistes et inhibiteurs , Animaux , Lignée cellulaire , Évaluation préclinique de médicament , Souris , Modèles moléculaires , Structure moléculaire , Phénothiazines/composition chimique , Protéines prion/isolement et purification , Protéines prion/métabolisme , Relation quantitative structure-activité
5.
Int J Mol Sci ; 20(20)2019 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-31618844

RÉSUMÉ

The cellular prion protein (PrPc) is an evolutionarily conserved cell surface protein encoded by the PRNP gene. PrPc is ubiquitously expressed within nearly all mammalian cells, though most abundantly within the CNS. Besides being implicated in the pathogenesis and transmission of prion diseases, recent studies have demonstrated that PrPc contributes to tumorigenesis by regulating tumor growth, differentiation, and resistance to conventional therapies. In particular, PrPc over-expression has been related to the acquisition of a malignant phenotype of cancer stem cells (CSCs) in a variety of solid tumors, encompassing pancreatic ductal adenocarcinoma (PDAC), osteosarcoma, breast cancer, gastric cancer, and primary brain tumors, mostly glioblastoma multiforme (GBM). Thus, PrPc is emerging as a key in maintaining glioblastoma cancer stem cells' (GSCs) phenotype, thereby strongly affecting GBM infiltration and relapse. In fact, PrPc contributes to GSCs niche's maintenance by modulating GSCs' stem cell-like properties while restraining them from differentiation. This is the first review that discusses the role of PrPc in GBM. The manuscript focuses on how PrPc may act on GSCs to modify their expression and translational profile while making the micro-environment surrounding the GSCs niche more favorable to GBM growth and infiltration.


Sujet(s)
Prédisposition aux maladies , Glioblastome/étiologie , Glioblastome/métabolisme , Protéines prion/génétique , Protéines prion/métabolisme , Animaux , Autophagie , Communication cellulaire , Différenciation cellulaire , Exosomes/métabolisme , Glioblastome/traitement médicamenteux , Glioblastome/anatomopathologie , Humains , Thérapie moléculaire ciblée , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Protéines prion/antagonistes et inhibiteurs , Protéines prion/composition chimique , Microenvironnement tumoral
6.
Protein Sci ; 28(9): 1690-1702, 2019 09.
Article de Anglais | MEDLINE | ID: mdl-31306510

RÉSUMÉ

Aggregation of the disordered protein α-synuclein into amyloid fibrils is a central feature of synucleinopathies, neurodegenerative disorders that include Parkinson's disease. Small, pre-fibrillar oligomers of misfolded α-synuclein are thought to be the key toxic entities, and α-synuclein misfolding can propagate in a prion-like way. We explored whether a compound with anti-prion activity that can bind to unfolded parts of the protein PrP, the cyclic tetrapyrrole Fe-TMPyP, was also active against α-synuclein aggregation. Observing the initial stages of aggregation via fluorescence cross-correlation spectroscopy, we found that Fe-TMPyP inhibited small oligomer formation in a dose-dependent manner. Fe-TMPyP also inhibited the formation of mature amyloid fibrils in vitro, as detected by thioflavin T fluorescence. Isothermal titration calorimetry indicated Fe-TMPyP bound to monomeric α-synuclein with a stoichiometry of 2, and two-dimensional heteronuclear single quantum coherence NMR spectra revealed significant interactions between Fe-TMPyP and the C-terminus of the protein. These results suggest commonalities among aggregation mechanisms for α-synuclein and the prion protein may exist that can be exploited as therapeutic targets.


Sujet(s)
Métalloporphyrines/pharmacologie , alpha-Synucléine/composition chimique , alpha-Synucléine/métabolisme , Amyloïde/effets des médicaments et des substances chimiques , Sites de fixation , Relation dose-effet des médicaments , Humains , Protéines prion/antagonistes et inhibiteurs , Protéines prion/composition chimique , Multimérisation de protéines/effets des médicaments et des substances chimiques , alpha-Synucléine/antagonistes et inhibiteurs
7.
Molecules ; 24(12)2019 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-31208037

RÉSUMÉ

BACKGROUND: While phase III clinical trials for the treatment of Alzheimer's disease (AD) keep failing regardless of the target, more and more data suggest that the toxic protein assemblies of amyloid-beta protein (Aß) and tubulin binding protein (TAU) behave like prions. Irrespective of the question of whether AD is theoretically or practically contagious, the presence of a self-replicating toxic etiologic agent in the brains of AD patients must have decisive consequences for drug development programs and clinical trial designs. OBJECTIVES: We intend to challenge the hypothesis that the underlying etiologic agent of AD is behaving prion-like. We want to discuss whether the outcome of clinical trials could have been predicted based on this hypothesis, and whether compounds that directly disassemble the toxic prion could be more beneficial for AD treatment. METHOD: We collected publicly accessible pre-clinical efficacy data of Aß targeting compounds that failed or still are in phase III clinical trials. We describe the desired properties of an anti-prion compound and compare it the properties of past and current phase III drug candidates. RESULTS: We could not find convincing and reproducible pre-clinical efficacy data of past and current phase III drug candidates on cognition other than in preventive treatment settings. The desired properties of an anti-Aß-prionic compound are fulfilled by the drug candidate RD2, which has been developed to directly disassemble toxic Aß oligomers. CONCLUSION: RD2 is the first anti-prion drug candidate. It is able to enhance cognition and impede neurodegeneration in three different transgenic AD mouse models, even under truly non-preventive conditions and even when applied orally. In addition, it is safe in humans.


Sujet(s)
Maladie d'Alzheimer/traitement médicamenteux , Peptides/usage thérapeutique , Protéines prion/antagonistes et inhibiteurs , Maladie d'Alzheimer/étiologie , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Peptides bêta-amyloïdes/antagonistes et inhibiteurs , Peptides bêta-amyloïdes/composition chimique , Peptides bêta-amyloïdes/métabolisme , Animaux , Essais cliniques comme sujet , Humains , Peptides/composition chimique , Peptides/pharmacologie , Protéines prion/métabolisme , Agrégats de protéines , Agrégation pathologique de protéines , Multimérisation de protéines , Résultat thérapeutique
8.
Molecules ; 24(2)2019 Jan 11.
Article de Anglais | MEDLINE | ID: mdl-30641935

RÉSUMÉ

Renal carcinoma cells express Membrane Type 1-Matrix Metalloproteinase (MT1-MMP, MMP-14) to degrade extracellular matrix components and a range of bioactive molecules to allow metastasis and cell proliferation. The activity of MT1-MMP is modulated by the endogenous inhibitors, Tissue Inhibitor of Metalloproteinases (TIMPs). In this study, we describe a novel strategy that would enable a "designer" TIMP-1 tailored specifically for MT1-MMP inhibition (V4A/P6V/T98L; Kiapp 1.66 nM) to be targeted to the plasma membrane for more effective MT1-MMP inhibition. To achieve this, we fuse the designer TIMP-1 to the glycosyl-phosphatidyl inositol (GPI) anchor of the prion protein to create a membrane-tethered, high-affinity TIMP variant named "T1Pr αMT1" that is predominantly located on the cell surface and co-localised with MT1-MMP. Confocal microscopy shows that T1Pr αMT1 is found throughout the cell surface in particular the membrane ruffles where MT1-MMP is most abundant. Expression of T1Pr αMT1 brings about a complete abrogation of the gelatinolytic activity of cellular MT1-MMP in HT1080 fibrosarcoma cells whilst in renal carcinoma cells CaKi-1, the GPI-TIMP causes a disruption in MMP-mediated proteolysis of ECM components such as fibronectin, collagen I and laminin that consequently triggers a downstream senescence response. Moreover, the transduced cells also suffer from an impairment in proliferation and survival in vitro as well as in NOD/SCID mouse xenograft. Taken together, our findings demonstrate that the GPI anchor of prion could be exploited as a targeting device in TIMP engineering for MT1-MMP inhibition with a potential in renal carcinoma therapy.


Sujet(s)
Néphrocarcinome/métabolisme , Membrane cellulaire/métabolisme , Matrix metalloproteinase 14/métabolisme , Inhibiteurs de métalloprotéinases matricielles/pharmacologie , Protéines prion/antagonistes et inhibiteurs , Inhibiteur tissulaire de métalloprotéinase-1/antagonistes et inhibiteurs , Inhibiteur tissulaire de métalloprotéinase-1/métabolisme , Animaux , Néphrocarcinome/traitement médicamenteux , Néphrocarcinome/génétique , Néphrocarcinome/anatomopathologie , Lignée cellulaire tumorale , Vieillissement de la cellule , Modèles animaux de maladie humaine , Expression des gènes , Humains , Matrix metalloproteinase 14/composition chimique , Matrix metalloproteinase 14/génétique , Inhibiteurs de métalloprotéinases matricielles/composition chimique , Souris , Inhibiteur tissulaire de métalloprotéinase-1/composition chimique , Tests d'activité antitumorale sur modèle de xénogreffe
9.
Cell Rep ; 26(1): 145-158.e8, 2019 01 02.
Article de Anglais | MEDLINE | ID: mdl-30605671

RÉSUMÉ

Cellular prion protein (PrPC) binds the scrapie conformation of PrP (PrPSc) and oligomeric ß-amyloid peptide (Aßo) to mediate transmissible spongiform encephalopathy (TSE) and Alzheimer's disease (AD), respectively. We conducted cellular and biochemical screens for compounds blocking PrPC interaction with Aßo. A polymeric degradant of an antibiotic targets Aßo binding sites on PrPC with low nanomolar affinity and prevents Aßo-induced pathophysiology. We then identified a range of negatively charged polymers with specific PrPC affinity in the low to sub-nanomolar range, from both biological (melanin) and synthetic (poly [4-styrenesulfonic acid-co-maleic acid], PSCMA) origin. Association of PSCMA with PrPC prevents Aßo/PrPC-hydrogel formation, blocks Aßo binding to neurons, and abrogates PrPSc production by ScN2a cells. We show that oral PSCMA yields effective brain concentrations and rescues APPswe/PS1ΔE9 transgenic mice from AD-related synapse loss and memory deficits. Thus, an orally active PrPC-directed polymeric agent provides a potential therapeutic approach to address neurodegeneration in AD and TSE.


Sujet(s)
Maladie d'Alzheimer/physiopathologie , Protéines prion/antagonistes et inhibiteurs , Animaux , Souris , Souris transgéniques , Transduction du signal
10.
Mol Neurobiol ; 55(3): 2384-2396, 2018 03.
Article de Anglais | MEDLINE | ID: mdl-28357807

RÉSUMÉ

Prion diseases are fatal transmissible neurodegenerative disorders affecting humans and various mammals. In spite of intensive efforts, there is no effective cure or treatment for prion diseases. Cellular forms of prion protein (PrPC) is essential for propagation of abnormal isoforms of prion protein (PrPSc) and pathogenesis. The effect of an artificial dual microRNA (DmiR) on PrPC suppression and resultant inhibition of prion replication was determined using prion-infectible cell cultures: differentiated C2C12 culture and primary mixed neuronal and glial cells culture (MNGC). Processing of DmiR by prion-susceptible myotubes, but not by reserve cells, in differentiated C2C12 culture slowed prion replication, implying an importance of cell type-specific PrPC targeting. In MNGC, reduction of PrPC with DmiR was effective for suppressing prion replication. MNGC lentivirally transduced with non-targeting control miRNAs (scrambled) reduced prion replication at a level similar to that with a synthetic analogue of viral RNA, poly I:C. The results suggest that a synergistic combination of the immunostimulatory RNA duplexes (miRNA) and PrPC silencing with DmiR might augment a therapeutic potential of RNA interference.


Sujet(s)
Survie cellulaire/physiologie , microARN/génétique , microARN/métabolisme , Protéines prion/génétique , Protéines prion/métabolisme , Animaux , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Cellules HEK293 , Humains , Lipopolysaccharides/toxicité , Souris , Protéines prion/antagonistes et inhibiteurs
11.
Handb Exp Pharmacol ; 245: 85-110, 2018.
Article de Anglais | MEDLINE | ID: mdl-28965171

RÉSUMÉ

The formation of protein aggregates and inclusions in the brain and spinal cord is a common neuropathological feature of a number of neurodegenerative diseases including Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and many others. These are commonly referred as neurodegenerative proteinopathies or protein-misfolding diseases. The main characteristic of protein aggregates in these disorders is the fact that they are enriched in amyloid fibrils. Since protein aggregation is considered to play a central role for the onset of neurodegenerative proteinopathies, research is ongoing to develop strategies aimed at preventing or removing protein aggregation in the brain of affected patients. Numerous studies have shown that small molecule-based approaches may be potentially the most promising for halting protein aggregation in neurodegenerative diseases. Indeed, several of these compounds have been found to interact with intrinsically disordered proteins and promote their clearing in experimental models. This notwithstanding, at present small molecule inhibitors still awaits achievements for clinical translation. Hopefully, if we determine whether the formation of insoluble inclusions is effectively neurotoxic and find a valid biomarker to assess their protein aggregation-inhibitory activity in the human central nervous system, the use of small molecule inhibitors will be considered as a cure for neurodegenerative protein-misfolding diseases.


Sujet(s)
Maladies neurodégénératives/traitement médicamenteux , Agrégation pathologique de protéines/traitement médicamenteux , Peptides bêta-amyloïdes/antagonistes et inhibiteurs , Peptides bêta-amyloïdes/composition chimique , Humains , Maladies neurodégénératives/étiologie , Protéines prion/antagonistes et inhibiteurs , Protéines prion/composition chimique , Superoxide dismutase-1/antagonistes et inhibiteurs , Superoxide dismutase-1/composition chimique , alpha-Synucléine/antagonistes et inhibiteurs , alpha-Synucléine/composition chimique
12.
Article de Anglais | MEDLINE | ID: mdl-29133563

RÉSUMÉ

The search for antiprion compounds has been encouraged by the fact that transmissible spongiform encephalopathies (TSEs) share molecular mechanisms with more prevalent neurodegenerative pathologies, such as Parkinson's and Alzheimer's diseases. Cellular prion protein (PrPC) conversion into protease-resistant forms (protease-resistant PrP [PrPRes] or the scrapie form of PrP [PrPSc]) is a critical step in the development of TSEs and is thus one of the main targets in the screening for antiprion compounds. In this work, three trimethoxychalcones (compounds J1, J8, and J20) and one oxadiazole (compound Y17), previously identified in vitro to be potential antiprion compounds, were evaluated through different approaches in order to gain inferences about their mechanisms of action. None of them changed PrPC mRNA levels in N2a cells, as shown by reverse transcription-quantitative real-time PCR. Among them, J8 and Y17 were effective in real-time quaking-induced conversion reactions using rodent recombinant PrP (rPrP) from residues 23 to 231 (rPrP23-231) as the substrate and PrPSc seeds from hamster and human brain. However, when rPrP from residues 90 to 231 (rPrP90-231), which lacks the N-terminal domain, was used as the substrate, only J8 remained effective, indicating that this region is important for Y17 activity, while J8 seems to interact with the PrPC globular domain. J8 also reduced the fibrillation of mouse rPrP23-231 seeded with in vitro-produced fibrils. Furthermore, most of the compounds decreased the amount of PrPC on the N2a cell surface by trapping this protein in the endoplasmic reticulum. On the basis of these results, we hypothesize that J8, a nontoxic compound previously shown to be a promising antiprion agent, may act by different mechanisms, since its efficacy is attributable not only to PrP conversion inhibition but also to a reduction of the PrPC content on the cell surface.


Sujet(s)
Chalcones/pharmacologie , Médicaments en essais cliniques/pharmacologie , Neurones/effets des médicaments et des substances chimiques , Oxadiazoles/pharmacologie , Protéines prion/antagonistes et inhibiteurs , Animaux , Sites de fixation , Lignée cellulaire tumorale , Chalcones/synthèse chimique , Clonage moléculaire , Médicaments en essais cliniques/synthèse chimique , Réticulum endoplasmique/effets des médicaments et des substances chimiques , Réticulum endoplasmique/métabolisme , Réticulum endoplasmique/ultrastructure , Escherichia coli/génétique , Escherichia coli/métabolisme , Expression des gènes , Vecteurs génétiques/composition chimique , Vecteurs génétiques/métabolisme , Cinétique , Souris , Simulation de docking moléculaire , Neurones/métabolisme , Neurones/anatomopathologie , Oxadiazoles/synthèse chimique , Protéines prion/composition chimique , Protéines prion/génétique , Protéines prion/métabolisme , Liaison aux protéines , Structure en hélice alpha , Motifs et domaines d'intéraction protéique , Protéines recombinantes/composition chimique , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme , Relation structure-activité , Thermodynamique
13.
PLoS One ; 12(9): e0185357, 2017.
Article de Anglais | MEDLINE | ID: mdl-28934337

RÉSUMÉ

Our previous study on prion-infected rodents revealed that hydroxypropyl methylcellulose compounds (HPMCs) with different molecular weights but similar composition and degree of substitution have different levels of long-lasting anti-prion activity. In this study, we searched these HPMCs for a parameter specifically associated with in vivo anti-prion activity by analyzing in vitro chemical properties and in vivo tissue distributions. Infrared spectroscopic and thermal analyses revealed no differences among HPMCs, whereas pyrene conjugation and spectroscopic analysis revealed that the fluorescence intensity ratio of peak III/peak I correlated with anti-prion activity. This correlation was more clearly demonstrated in the anti-prion activity of the 1-year pre-infection treatment than that of the immediate post-infection treatment. In addition, the intensity ratio of peak III/peak I negatively correlated with the macrophage uptake level of HPMCs in our previous study. However, the in vivo distribution pattern was apparently not associated with anti-prion activity and was different in the representative tissues. These findings suggest that pyrene conjugation and spectroscopic analysis are powerful methods to successfully demonstrate local dielectric differences in HPMCs and provide a feasible parameter denoting the long-lasting anti-prion activity of HPMCs in vivo.


Sujet(s)
Dérivés de l'hypromellose/composition chimique , Dérivés de l'hypromellose/pharmacologie , Protéines prion/antagonistes et inhibiteurs , Pyrènes/composition chimique , Animaux , Transport biologique , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Impédance électrique , Dérivés de l'hypromellose/métabolisme , Dérivés de l'hypromellose/pharmacocinétique , Macrophages/métabolisme , Souris , Masse moléculaire , Protéines prion/métabolisme , Spectroscopie infrarouge à transformée de Fourier , Rate/effets des médicaments et des substances chimiques , Rate/métabolisme , Relation structure-activité , Distribution tissulaire
14.
Prog Mol Biol Transl Sci ; 150: 35-56, 2017.
Article de Anglais | MEDLINE | ID: mdl-28838668

RÉSUMÉ

The function of the cellular prion protein (PrPC), while still poorly understood, is increasingly linked to its ability to bind physiological metal ions at the cell surface. PrPC binds divalent forms of both copper and zinc through its unstructured N-terminal domain, modulating interactions between PrPC and various receptors at the cell surface and ultimately tuning downstream cellular processes. In this chapter, we briefly discuss the molecular features of copper and zinc uptake by PrPC and summarize evidence implicating these metal ions in PrP-mediated physiology. We then focus our review on recent biophysical evidence revealing a physical interaction between the flexible N-terminal and globular C-terminal domains of PrPC. This interdomain cis interaction is electrostatic in nature and is promoted by the binding of Cu2+ and Zn2+ to the N-terminal octarepeat domain. These findings, along with recent cellular studies, suggest a mechanism whereby NC interactions serve to regulate the activity and/or toxicity of the PrPC N-terminus. We discuss this potential mechanism in relation to familial prion disease mutations, lethal deletions of the PrPC central region, and neurotoxicity induced by certain globular domain ligands, including bona fide prions and toxic amyloid-ß oligomers.


Sujet(s)
Cuivre/pharmacologie , Neurotoxines/antagonistes et inhibiteurs , Neurotoxines/composition chimique , Protéines prion/antagonistes et inhibiteurs , Protéines prion/composition chimique , Zinc/pharmacologie , Animaux , Humains , Modèles moléculaires , Domaines protéiques
15.
Prion ; 11(4): 265-276, 2017 07 04.
Article de Anglais | MEDLINE | ID: mdl-28665745

RÉSUMÉ

Prion diseases are fatal and incurable neurodegenerative diseases of humans and animals. Despite years of research, no therapeutic agents have been developed that can effectively manage or reverse disease progression. Recently it has been identified that recombinant prion proteins (rPrP) expressed in bacteria can act as inhibitors of prion replication within the in vitro prion replication system protein misfolding cyclic amplification (PMCA). Here, within PMCA reactions amplifying a range of ruminant prions including distinct Prnp genotypes/host species and distinct prion strains, recombinant ovine VRQ PrP displayed consistent inhibition of prion replication and produced IC50 values of 122 and 171 nM for ovine scrapie and bovine BSE replication, respectively. These findings illustrate the therapeutic potential of rPrPs with distinct TSE diseases.


Sujet(s)
Maladies à prions/traitement médicamenteux , Maladies à prions/médecine vétérinaire , Protéines prion/antagonistes et inhibiteurs , Protéines recombinantes/métabolisme , Animaux , Encéphale/métabolisme , Encéphale/anatomopathologie , Bovins , Encéphalopathie spongiforme bovine/traitement médicamenteux , Encéphalopathie spongiforme bovine/métabolisme , Encéphalopathie spongiforme bovine/transmission , Régulation de l'expression des gènes bactériens , Techniques in vitro , Maladies à prions/métabolisme , Protéines prion/métabolisme , Pliage des protéines , Tremblante/traitement médicamenteux , Tremblante/métabolisme , Ovis
16.
ChemMedChem ; 12(16): 1286-1292, 2017 08 22.
Article de Anglais | MEDLINE | ID: mdl-28722340

RÉSUMÉ

Into the fold: Prion diseases are neurodegenerative disorders characterized by the accumulation in the brain of a self-replicating, misfolded isoform (PrPSc ) of the cellular prion protein (PrPC ). No therapies are available for these pathologies. We capitalized on previously described cell-based assays to screen a library of small molecules, and identified 55, a compound capable of counteracting both prion replication and toxicity. Compound 55 may represent the starting point for the development of a completely new class of therapeutics for prion diseases.


Sujet(s)
Protéines prion/métabolisme , Bibliothèques de petites molécules/métabolisme , Animaux , Encéphale/métabolisme , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules HEK293 , Humains , Concentration inhibitrice 50 , Mutagenèse , Protéines PrPSc/antagonistes et inhibiteurs , Protéines PrPSc/génétique , Protéines PrPSc/métabolisme , Maladies à prions/métabolisme , Maladies à prions/anatomopathologie , Protéines prion/antagonistes et inhibiteurs , Protéines prion/génétique , Liaison aux protéines , Bibliothèques de petites molécules/composition chimique , Bibliothèques de petites molécules/toxicité
17.
Phytother Res ; 31(7): 1046-1055, 2017 Jul.
Article de Anglais | MEDLINE | ID: mdl-28509424

RÉSUMÉ

Neurodegenerative diseases are associated with accumulation of amyloid-type protein misfolding products. Prion protein (PrP) is known for its ability to aggregate into soluble oligomers that in turn associate into amyloid fibrils. Preventing the formation of these infective and neurotoxic entities represents a viable strategy to control prion diseases. Numerous attempts to find dietary compounds with anti-prion properties have been made; however, the most promising agent found so far was curcumin, which is poorly soluble and merely bioavailable. In the present work, we identify 3,4-dimethoxycinnamic acid (DMCA) which is a bioavailable coffee component as a perspective anti-prion compound. 3,4-Dimethoxycinnamic acid was found to bind potently to prion protein with a Kd of 405 nM. An in vitro study of DMCA effect on PrP oligomerization and fibrillization was undertaken using isothermal titration calorimetry (ITC), dynamic light scattering (DLS) and circular dichroism (CD) methodologies. We demonstrated that DMCA affects PrP oligomer formation reducing the oligomer content by 30-40%, and enhancing SH-SY5Y cell viability treated with prion oligomers. Molecular docking studies allowed to suggest a site where DMCA is able to bind stabilizing PrP tertiary structure. We suggest that DMCA is a perspective dietary compound for prophylaxis of neurodegenerative diseases that needs further research. Copyright © 2017 John Wiley & Sons, Ltd.


Sujet(s)
Cinnamates/composition chimique , Protéines prion/antagonistes et inhibiteurs , Prions/antagonistes et inhibiteurs , Sites de fixation , Simulation de docking moléculaire , Maladies à prions
18.
Int J Mol Sci ; 18(5)2017 May 17.
Article de Anglais | MEDLINE | ID: mdl-28513534

RÉSUMÉ

Transmissible spongiform encephalopathies (TSEs) are a group of neurodegenerative disorders that affect humans and other mammals. The etiologic agents common to these diseases are misfolded conformations of the prion protein (PrP). The molecular mechanisms that trigger the structural conversion of the normal cellular PrP (PrPC) into the pathogenic conformer (PrPSc) are still poorly understood. It is proposed that a molecular cofactor would act as a catalyst, lowering the activation energy of the conversion process, therefore favoring the transition of PrPC to PrPSc. Several in vitro studies have described physical interactions between PrP and different classes of molecules, which might play a role in either PrP physiology or pathology. Among these molecules, nucleic acids (NAs) are highlighted as potential PrP molecular partners. In this context, the SELEX (Systematic Evolution of Ligands by Exponential Enrichment) methodology has proven extremely valuable to investigate PrP-NA interactions, due to its ability to select small nucleic acids, also termed aptamers, that bind PrP with high affinity and specificity. Aptamers are single-stranded DNA or RNA oligonucleotides that can be folded into a wide range of structures (from harpins to G-quadruplexes). They are selected from a nucleic acid pool containing a large number (1014-1016) of random sequences of the same size (~20-100 bases). Aptamers stand out because of their potential ability to bind with different affinities to distinct conformations of the same protein target. Therefore, the identification of high-affinity and selective PrP ligands may aid the development of new therapies and diagnostic tools for TSEs. This review will focus on the selection of aptamers targeted against either full-length or truncated forms of PrP, discussing the implications that result from interactions of PrP with NAs, and their potential advances in the studies of prions. We will also provide a critical evaluation, assuming the advantages and drawbacks of the SELEX (Systematic Evolution of Ligands by Exponential Enrichment) technique in the general field of amyloidogenic proteins.


Sujet(s)
Aptamères nucléotidiques/métabolisme , Protéines de transport/métabolisme , Protéines prion/métabolisme , Protéines amyloïdogènes/métabolisme , Animaux , ADN simple brin/métabolisme , Humains , Acides nucléiques/métabolisme , Maladies à prions/métabolisme , Protéines prion/antagonistes et inhibiteurs , Liaison aux protéines , Technique SELEX
19.
BMC Genomics ; 18(1): 319, 2017 04 22.
Article de Anglais | MEDLINE | ID: mdl-28431525

RÉSUMÉ

BACKGROUND: The function of the prion protein, involved in the so-called prion diseases, remains a subject of intense debate and the possibility that it works as a pleiotropic protein through the interaction with multiple membrane proteins is somehow supported by recent reports. Therefore, the use of proteomic and bioinformatics combined to uncover cellular processes occurring together with changes in the expression of the prion protein may provide further insight into the putative pleiotropic role of the prion protein. RESULTS: This study assessed the membrane-enriched proteome changes accompanying alterations in the expression of the prion protein. A 2D-DIGE approach was applied to two cell lines after prefractionation towards the membrane protein subset: an embryonic stem cell line and the PK1 subline of neuroblastoma cells which efficiently propagates prion infection. Several proteins were differentially abundant with the increased expression of the prion protein during neural differentiation of embryonic stem cells and with the knockdown of the prion protein in PK1 cells. The identity of around 20% of the differentially abundant proteins was obtained by tandem MS. The catalytic subunit A of succinate dehydrogenase, a key enzyme for the aerobic energy metabolism and redox homeostasis, showed a similar abundance trend as the prion protein in both proteomic experiments. A gene ontology analysis revealed "myelin sheath", "organelle membrane" and "focal adhesion" associated proteins as the main cellular components, and "protein folding" and "ATPase activity" as the biological processes enriched in the first set of differentially abundant proteins. The known interactome of these differentially abundant proteins was customized to reveal four interactors with the prion protein, including two heat shock proteins and a protein disulfide isomerase. CONCLUSIONS: Overall, our study shows that expression of the prion protein occurs concomitantly with changes in chaperone activity and cell-redox homeostasis, emphasizing the functional link between these cellular processes and the prion protein.


Sujet(s)
Protéines membranaires/métabolisme , Protéines prion/métabolisme , Protéome/métabolisme , Animaux , Différenciation cellulaire , Lignée cellulaire tumorale , Électrophorèse bidimensionnelle sur gel , Protéines membranaires/analyse , Souris , Cellules souches embryonnaires de souris/cytologie , Cellules souches embryonnaires de souris/métabolisme , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Protéines prion/antagonistes et inhibiteurs , Protéines prion/génétique , Protéome/analyse , Interférence par ARN , Petit ARN interférent/métabolisme , Spectrométrie de masse MALDI
20.
Prion ; 10(4): 305-18, 2016 07 03.
Article de Anglais | MEDLINE | ID: mdl-27565564

RÉSUMÉ

Prion-infected cells have been used for analyzing the effect of compounds on the formation of abnormal isoform of prion protein (PrP(Sc)). PrP(Sc) is usually detected using anti-prion protein (PrP) antibodies after the removal of the cellular isoform of prion protein (PrP(C)) by proteinase K (PK) treatment. However, it is expected that the PK-sensitive PrP(Sc) (PrP(Sc)-sen), which possesses higher infectivity and conversion activity than the PK-resistant PrP(Sc) (PrP(Sc)-res), is also digested through PK treatment. To overcome this problem, we established a novel cell-based ELISA in which PrP(Sc) can be directly detected from cells persistently infected with prions using anti-PrP monoclonal antibody (mAb) 132 that recognizes epitope consisting of mouse PrP amino acids 119-127. The novel cell-based ELISA could distinguish prion-infected cells from prion-uninfected cells without cell lysis and PK treatment. MAb 132 could detect both PrP(Sc)-sen and PrP(Sc)-res even if all PrP(Sc) molecules were not detected. The analytical dynamic range for PrP(Sc) detection was approximately 1 log. The coefficient of variation and signal-to-background ratio were 7%-11% and 2.5-3.3, respectively, demonstrating the reproducibility of this assay. The addition of a cytotoxicity assay immediately before PrP(Sc) detection did not affect the following PrP(Sc) detection. Thus, all the procedures including cell culture, cytotoxicity assay, and PrP(Sc) detection were completed in the same plate. The simplicity and non-requirement for cell lysis or PK treatment are advantages for the high throughput screening of anti-prion compounds.


Sujet(s)
Endopeptidase K/pharmacologie , Test ELISA/méthodes , Protéines PrPSc/analyse , Maladies à prions/diagnostic , Protéines prion/métabolisme , Animaux , Anticorps monoclonaux/pharmacologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Résistance aux substances , Souris , Maladies à prions/immunologie , Maladies à prions/métabolisme , Protéines prion/antagonistes et inhibiteurs , Protéines prion/immunologie , Isoformes de protéines
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...