Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 404
Filtrer
1.
BMB Rep ; 57(6): 263-272, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38835114

RÉSUMÉ

Amyloid-ß (Aß) is one of the amyloidogenic intrinsically disordered proteins (IDPs) that self-assemble to protein aggregates, incurring cell malfunction and cytotoxicity. While Aß has been known to regulate multiple physiological functions, such as enhancing synaptic functions, aiding in the recovery of the blood-brain barrier/brain injury, and exhibiting tumor suppression/antimicrobial activities, the hydrophobicity of the primary structure promotes pathological aggregations that are closely associated with the onset of Alzheimer's disease (AD). Aß proteins consist of multiple isoforms with 37-43 amino acid residues that are produced by the cleavage of amyloid-ß precursor protein (APP). The hydrolytic products of APP are secreted to the extracellular regions of neuronal cells. Aß 1-42 (Aß42) and Aß 1-40 (Aß40) are dominant isoforms whose significance in AD pathogenesis has been highlighted in numerous studies to understand the molecular mechanism and develop AD diagnosis and therapeutic strategies. In this review, we focus on the differences between Aß42 and Aß40 in the molecular mechanism of amyloid aggregations mediated by the two additional residues (Ile41 and Ala42) of Aß42. The current comprehension of Aß42 and Aß40 in AD progression is outlined, together with the structural features of Aß42/Aß40 amyloid fibrils, and the aggregation mechanisms of Aß42/Aß40. Furthermore, the impact of the heterogeneous distribution of Aß isoforms during amyloid aggregations is discussed in the system mimicking the coexistence of Aß42 and Aß40 in human cerebrospinal fluid (CSF) and plasma. [BMB Reports 2024; 57(6): 263-272].


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Isoformes de protéines , Animaux , Humains , Maladie d'Alzheimer/métabolisme , Peptides bêta-amyloïdes/métabolisme , Précurseur de la protéine bêta-amyloïde/métabolisme , Fragments peptidiques/métabolisme , Fragments peptidiques/composition chimique , Agrégats de protéines/physiologie , Agrégation pathologique de protéines/métabolisme , Isoformes de protéines/métabolisme
2.
Acta Neuropathol Commun ; 12(1): 84, 2024 05 31.
Article de Anglais | MEDLINE | ID: mdl-38822421

RÉSUMÉ

Alpha-synuclein (αsyn) is an intrinsically disordered protein that aggregates in the brain in several neurodegenerative diseases collectively called synucleinopathies. Phosphorylation of αsyn at serine 129 (PSER129) was considered rare in the healthy human brain but is enriched in pathological αsyn aggregates and is used as a specific marker for disease inclusions. However, recent observations challenge this assumption by demonstrating that PSER129 results from neuronal activity and can be readily detected in the non-diseased mammalian brain. Here, we investigated experimental conditions under which two distinct PSER129 pools, namely endogenous-PSER129 and aggregated-PSER129, could be detected and differentiated in the mammalian brain. Results showed that in the wild-type (WT) mouse brain, perfusion fixation conditions greatly influenced the detection of endogenous-PSER129, with endogenous-PSER129 being nearly undetectable after delayed perfusion fixation (30-min and 1-h postmortem interval). Exposure to anesthetics (e.g., Ketamine or xylazine) before perfusion did not significantly influence endogenous-PSER129 detection or levels. In situ, non-specific phosphatase calf alkaline phosphatase (CIAP) selectively dephosphorylated endogenous-PSER129 while αsyn preformed fibril (PFF)-seeded aggregates and genuine disease aggregates (Lewy pathology and Papp-Lantos bodies in Parkinson's disease and multiple systems atrophy brain, respectively) were resistant to CIAP-mediated dephosphorylation. The phosphatase resistance of aggregates was abolished by sample denaturation, and CIAP-resistant PSER129 was closely associated with proteinase K (PK)-resistant αsyn (i.e., a marker of aggregation). CIAP pretreatment allowed for highly specific detection of seeded αsyn aggregates in a mouse model that accumulates non-aggregated-PSER129. We conclude that αsyn aggregates are impervious to phosphatases, and CIAP pretreatment increases detection specificity for aggregated-PSER129, particularly in well-preserved biological samples (e.g., perfusion fixed or flash-frozen mammalian tissues) where there is a high probability of interference from endogenous-PSER129. Our findings have important implications for the mechanism of PSER129-accumulation in the synucleinopathy brain and provide a simple experimental method to differentiate endogenous-from aggregated PSER129.


Sujet(s)
Encéphale , Souris de lignée C57BL , alpha-Synucléine , Animaux , Humains , Mâle , Souris , Phosphatase alcaline/métabolisme , alpha-Synucléine/métabolisme , Encéphale/métabolisme , Encéphale/anatomopathologie , Souris transgéniques , Phosphoric monoester hydrolases/métabolisme , Phosphorylation , Agrégats de protéines/physiologie , Agrégation pathologique de protéines/métabolisme , Agrégation pathologique de protéines/anatomopathologie , Synucléinopathies/métabolisme , Synucléinopathies/anatomopathologie
3.
Gac Med Mex ; 160(1): 1-8, 2024.
Article de Anglais | MEDLINE | ID: mdl-38753562

RÉSUMÉ

BACKGROUND: Protein interactions participate in many molecular mechanisms involved in cellular processes. The human TATA box binding protein (hTBP) interacts with Antennapedia (Antp) through its N-terminal region, specifically via its glutamine homopeptides. This PolyQ region acts as a binding site for other transcription factors under normal conditions, but when it expands, it generates spinocerebellar ataxia 17 (SCA17), whose protein aggregates in the brain prevent its correct functioning. OBJECTIVE: To determine whether the hTBP glutamine-rich region is involved in its interaction with homeoproteins and the role it plays in the formation of protein aggregates in SCA17. MATERIAL AND METHODS: We characterized hTBP interaction with other homeoproteins using BiFC, and modeled SCA17 in Drosophila melanogaster by targeting hTBPQ80 to the fly brain using UAS/GAL4. RESULTS: There was hTBP interaction with homeoproteins through its glutamine-rich region, and hTBP protein aggregates with expanded glutamines were found to affect the locomotor capacity of flies. CONCLUSIONS: The study of hTBP interactions opens the possibility for the search for new therapeutic strategies in neurodegenerative pathologies such as SCA17.


ANTECEDENTES: Las interacciones proteicas participan en una gran cantidad de mecanismos moleculares que rigen los procesos celulares. La proteína de unión a la caja TATA humana (hTBP) interacciona con Antennapedia (Antp) a través de su extremo N-terminal, específicamente a través de sus homopéptidos de glutaminas. Esta región PolyQ sirve como sitio de unión a factores de transcripción en condiciones normales, pero cuando se expande genera la ataxia espinal cerebelosa 17 (SCA17), cuyos agregados proteicos en el cerebro impiden su funcionamiento correcto. OBJETIVO: Determinar si la región rica en glutaminas de hTBP interviene en su interacción con homeoproteínas y el papel que tiene en la formación de agregados proteicos en SCA17. MATERIAL Y MÉTODOS: Se caracterizó la interacción de hTBP con otras homeoproteínas usando BiFC y se modeló SCA17 en Drosophila melanogaster dirigiendo hTBPQ80 al cerebro de las moscas usando UAS/GAL4. RESULTADOS: Existió interacción de hTBP con homeoproteínas a través de su región rica en glutaminas. Los agregados proteicos de hTBP con las glutaminas expandidas afectaron la capacidad locomotriz de las moscas. CONCLUSIONES: El estudio de las interacciones de hTBP abre la posibilidad para la búsqueda de nuevas estrategias terapéuticas en patologías neurodegenerativas como SCA17.


Sujet(s)
Modèles animaux de maladie humaine , Drosophila melanogaster , Ataxies spinocérébelleuses , Protéine de liaison à la boite TATA , Animaux , Drosophila melanogaster/métabolisme , Ataxies spinocérébelleuses/métabolisme , Ataxies spinocérébelleuses/génétique , Protéine de liaison à la boite TATA/métabolisme , Protéine de liaison à la boite TATA/génétique , Humains , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Glutamine/métabolisme , Agrégats de protéines/physiologie , Peptides/métabolisme , Encéphale/métabolisme
4.
Neurochem Int ; 177: 105762, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38723901

RÉSUMÉ

Linalool is a neuroprotective monoterpene found in essential oils from aromatic plants. Linalool's effectiveness in AD animal models has been established previously, but its mechanisms of action remain unclear. Therefore, this study aims to investigate whether linalool binds directly to the amyloid beta (Aß) fibrils to understand it's role in preventing neurodegeneration. The anti-aggregation ability of Linalool was determined using Dithiothreitol (DTT), and thermal aggregation assays followed by Thioflavin T (ThT) binding assay. AD animals were treated with Linalool, and Thioflavin T staining was used to check the binding of linalool to Aß fibrils in rat brain tissue sections. Preliminary studies revealed the anti-aggregation potential of linalool under the thermal and chemical stimulus. Further, in ThT binding assay Linalool inhibited Aß aggregation, binding directly to Aß fibrils. The reduced fluorescence intensity of ThT in AD brain tissues following linalool administration, highlights its neuroprotective potential as a therapeutic agent for AD.


Sujet(s)
Monoterpènes acycliques , Peptides bêta-amyloïdes , Peptides bêta-amyloïdes/métabolisme , Peptides bêta-amyloïdes/antagonistes et inhibiteurs , Monoterpènes acycliques/pharmacologie , Animaux , Rats , Mâle , Monoterpènes/pharmacologie , Monoterpènes/usage thérapeutique , Monoterpènes/composition chimique , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Neuroprotecteurs/pharmacologie , Encéphale/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Rat Wistar , Agrégats de protéines/effets des médicaments et des substances chimiques , Agrégats de protéines/physiologie , Rat Sprague-Dawley , Agrégation pathologique de protéines/traitement médicamenteux , Agrégation pathologique de protéines/métabolisme , Agrégation pathologique de protéines/prévention et contrôle
5.
ACS Chem Neurosci ; 15(9): 1915-1925, 2024 05 01.
Article de Anglais | MEDLINE | ID: mdl-38634811

RÉSUMÉ

Calcium-binding S100A8 and S100A9 proteins play a significant role in various disorders due to their pro-inflammatory functions. Substantially, they are also relevant in neurodegenerative disorders via the delivery of signals for the immune response. However, at the same time, they can aggregate and accelerate the progression of diseases. Natively, S100A8 and S100A9 exist as homo- and heterodimers, but upon aggregation, they form amyloid-like oligomers, fibrils, or amorphous aggregates. In this study, we aimed to elucidate the aggregation propensities of S100A8, S100A9, and their heterodimer calprotectin by investigating aggregation kinetics, secondary structures, and morphologies of the aggregates. For the first time, we followed the in vitro aggregation of S100A8, which formed spherical aggregates, unlike the fibrillar structures of S100A9 under the same conditions. The aggregates were sensitive to amyloid-specific ThT and ThS dyes and had a secondary structure composed of ß-sheets. Similarly to S100A9, S100A8 protein was stabilized by calcium ions, resulting in aggregation inhibition. Finally, the formation of S100A8 and S100A9 heterodimers stabilized the proteins in the absence of calcium ions and prevented their aggregation.


Sujet(s)
Amyloïde , Calgranuline A , Calgranuline B , Complexe antigénique L1 leucocytaire , Calgranuline B/métabolisme , Calgranuline A/métabolisme , Complexe antigénique L1 leucocytaire/métabolisme , Amyloïde/métabolisme , Humains , Agrégats de protéines/physiologie , Agrégats de protéines/effets des médicaments et des substances chimiques , Calcium/métabolisme , Structure secondaire des protéines
6.
ACS Chem Neurosci ; 15(9): 1770-1786, 2024 05 01.
Article de Anglais | MEDLINE | ID: mdl-38637513

RÉSUMÉ

Parkinson's disease arises from protein misfolding, aggregation, and fibrillation and is characterized by LB (Lewy body) deposits, which contain the protein α-synuclein (α-syn) as their major component. Another synuclein, γ-synuclein (γ-syn), coexists with α-syn in Lewy bodies and is also implicated in various types of cancers, especially breast cancer. It is known to seed α-syn fibrillation after its oxidation at methionine residue, thereby contributing in synucleinopathy. Despite its involvement in synucleinopathy, the search for small molecule inhibitors and modulators of γ-syn fibrillation remains largely unexplored. This work reveals the modulatory properties of cyclic-nordihydroguaiaretic acid (cNDGA), a natural polyphenol, on the structural and aggregational properties of human γ-syn employing various biophysical and structural tools, namely, thioflavin T (ThT) fluorescence, Rayleigh light scattering, 8-anilinonaphthalene-1-sulfonic acid binding, far-UV circular dichroism (CD), Fourier transform infrared spectroscopy (FTIR) spectroscopy, atomic force microscopy, ITC, molecular docking, and MTT-toxicity assay. cNDGA was observed to modulate the fibrillation of γ-syn to form off-pathway amorphous species that are nontoxic in nature at as low as 75 µM concentration. The modulation is dependent on oxidizing conditions, with cNDGA weakly interacting (Kd ∼10-5 M) with the residues at the N-terminal of γ-syn protein as investigated by isothermal titration calorimetry and molecular docking, respectively. Increasing cNDGA concentration results in an increased recovery of monomeric γ-syn as shown by sodium dodecyl sulfate and native-polyacrylamide gel electrophoresis. The retention of native structural properties of γ-syn in the presence of cNDGA was further confirmed by far-UV CD and FTIR. In addition, cNDGA is most effective in suppression of fibrillation when added at the beginning of the fibrillation kinetics and is also capable of disintegrating the preformed mature fibrils. These findings could, therefore, pave the ways for further exploring cNDGA as a potential therapeutic against γ-synucleinopathies.


Sujet(s)
Masoprocol , gamma-Synucléine , Humains , gamma-Synucléine/métabolisme , Masoprocol/pharmacologie , Agrégats de protéines/effets des médicaments et des substances chimiques , Agrégats de protéines/physiologie , Spectroscopie infrarouge à transformée de Fourier , Agrégation pathologique de protéines/métabolisme , Agrégation pathologique de protéines/traitement médicamenteux
7.
J Chem Neuroanat ; 138: 102420, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38626816

RÉSUMÉ

Protein aggregation is a pathological feature in various neurodegenerative diseases and is thought to play a crucial role in the onset and progression of neurological disorders. This pathological phenomenon has attracted increasing attention from researchers, but the underlying mechanism has not been fully elucidated yet. Researchers are increasingly interested in identifying chemicals or methods that can effectively detect protein aggregation or maintain protein stability to prevent aggregation formation. To date, several methods are available for detecting protein aggregates, including fluorescence correlation spectroscopy, electron microscopy, and molecular detection methods. Unfortunately, there is still a lack of methods to observe protein aggregation in situ under a microscope. This article reviews the two main aspects of protein aggregation: the mechanisms and detection methods of protein aggregation. The aim is to provide clues for the development of new methods to study this pathological phenomenon.


Sujet(s)
Agrégation pathologique de protéines , Humains , Animaux , Agrégation pathologique de protéines/métabolisme , Agrégats de protéines/physiologie , Maladies du système nerveux/métabolisme , Maladies neurodégénératives/métabolisme
8.
Cells ; 13(3)2024 Jan 29.
Article de Anglais | MEDLINE | ID: mdl-38334646

RÉSUMÉ

Protein aggregation is a predominant feature of many neurodegenerative diseases, including synucleinopathies, which are characterized by cellular inclusions containing α-Synuclein (αSyn) phosphorylated at serine 129 (pSer129). In the present study, we characterized the development of αSyn pre-formed fibril (PFF)-induced pSer129-αSyn pathology in F28tg mice overexpressing human wild-type αSyn, as well as in ex vivo organotypic cultures and in vitro primary cultures from the same mouse model. Concurrently, we collected cerebrospinal fluid (CSF) from mice and conditioned media from ex vivo and in vitro cultures and quantified the levels of neurofilament light chain (NFL), a biomarker of neurodegeneration. We found that the intra-striatal injection of PFFs induces the progressive spread of pSer129-αSyn pathology and microglial activation in vivo, as well as modest increases in NFL levels in the CSF. Similarly, PFF-induced αSyn pathology occurs progressively in ex vivo organotypic slice cultures and is accompanied by significant increases in NFL release into the media. Using in vitro primary hippocampal cultures, we further confirmed that pSer129-αSyn pathology and NFL release occur in a manner that correlates with the fibril dose and the level of the αSyn protein. Overall, we demonstrate that αSyn pathology is associated with NFL release across preclinical models of seeded αSyn aggregation and that the pharmacological inhibition of αSyn aggregation in vitro also significantly reduces NFL release.


Sujet(s)
Maladies neurodégénératives , Synucléinopathies , Animaux , Humains , Souris , alpha-Synucléine/métabolisme , Filaments intermédiaires/métabolisme , Maladies neurodégénératives/anatomopathologie , Agrégats de protéines/physiologie
9.
Sci Adv ; 9(37): eadi1057, 2023 09 15.
Article de Anglais | MEDLINE | ID: mdl-37713485

RÉSUMÉ

Insulin is a hormone responsible for maintaining normal glucose levels by activating insulin receptor (IR) and is the primary treatment for diabetes. However, insulin is prone to unfolding and forming cross-ß fibers. Fibrillation complicates insulin storage and therapeutic application. Molecular details of insulin fibrillation remain unclear, hindering efforts to prevent fibrillation process. Here, we characterized insulin fibrils using cryo-electron microscopy (cryo-EM), showing multiple forms that contain one or more of the protofilaments containing both the A and B chains of insulin linked by disulfide bonds. We solved the cryo-EM structure of one of the fibril forms composed of two protofilaments at 3.2-Å resolution, which reveals both the ß sheet conformation of the protofilament and the packing interaction between them that underlie the fibrillation. On the basis of this structure, we designed several insulin mutants that display reduced fibrillation while maintaining native IR signaling activity. These designed insulin analogs may be developed into more effective therapeutics for type 1 diabetes.


Sujet(s)
Diabète de type 1 , Insuline , Agrégats de protéines , Humains , Cryomicroscopie électronique , Diabète de type 1/traitement médicamenteux , Insuline/composition chimique , Insuline/physiologie , Agrégats de protéines/physiologie
10.
Bioorg Med Chem Lett ; 86: 129257, 2023 04 15.
Article de Anglais | MEDLINE | ID: mdl-36966976

RÉSUMÉ

The formation of aggregates due to protein misfolding is encountered in various neurodegenerative diseases. α-Synuclein (α-Syn) aggregation is linked to Parkinson's disease (PD). It is one of the most prevalent neurodegenerative disorders after Alzheimer's disease. Aggregation of α-Syn is associated with Lewy body formation and degeneration of the dopaminergic neurons in the brain. These are the pathological hallmarks of PD progression. α-Syn aggregates in a multi-step process. The native unstructured α-Syn monomers combine to form oligomers, followed by amyloid fibrils, and finally Lewy bodies. Recent evidence suggests that α-Syn oligomerization and fibrils formation play major roles in PD development. α-Syn oligomeric species is the main contributor to neurotoxicity. Therefore, the detection of α-Syn oligomers and fibrils has drawn significant attention for potential diagnostic and therapeutic development. In this regard, the fluorescence strategy has become the most popular approach for following the protein aggregation process. Thioflavin T (ThT) is the most frequently used probe for monitoring amyloid kinetics. Unfortunately, it suffers from several significant drawbacks including the inability to detect neurotoxic oligomers. Researchers developed several small molecule-based advanced fluorescent probes compared to ThT for the detection/monitoring of α-Syn aggregates states. These are summarized here.


Sujet(s)
Maladie d'Alzheimer , Maladie de Parkinson , Humains , alpha-Synucléine/métabolisme , Colorants fluorescents , Maladie de Parkinson/métabolisme , Agrégats de protéines/physiologie , Maladie d'Alzheimer/métabolisme , Amyloïde/métabolisme , Agrégation pathologique de protéines/métabolisme
11.
Nat Commun ; 14(1): 947, 2023 02 28.
Article de Anglais | MEDLINE | ID: mdl-36854675

RÉSUMÉ

The ability of cells to manage consequences of exogenous proteotoxicity is key to cellular homeostasis. While a plethora of well-characterised machinery aids intracellular proteostasis, mechanisms involved in the response to denaturation of extracellular proteins remain elusive. Here we show that aggregation of protein ectodomains triggers their endocytosis via a macroendocytic route, and subsequent lysosomal degradation. Using ERBB2/HER2-specific antibodies we reveal that their cross-linking ability triggers specific and fast endocytosis of the receptor, independent of clathrin and dynamin. Upon aggregation, canonical clathrin-dependent cargoes are redirected into the aggregation-dependent endocytosis (ADE) pathway. ADE is an actin-driven process, which morphologically resembles macropinocytosis. Physical and chemical stress-induced aggregation of surface proteins also triggers ADE, facilitating their degradation in the lysosome. This study pinpoints aggregation of extracellular domains as a trigger for rapid uptake and lysosomal clearance which besides its proteostatic function has potential implications for the uptake of pathological protein aggregates and antibody-based therapies.


Sujet(s)
Membrane cellulaire , Protéines membranaires , Agrégats de protéines , Homéostasie protéique , Anticorps , Membrane cellulaire/métabolisme , Clathrine , Endocytose/physiologie , Protéines membranaires/métabolisme , Protéines membranaires/physiologie , Agrégats de protéines/physiologie
12.
FASEB J ; 37(1): e22702, 2023 01.
Article de Anglais | MEDLINE | ID: mdl-36520044

RÉSUMÉ

Neurodegenerative diseases result from the interplay of abnormal gene expression and various pathological factors. Therefore, a disease-specific integrative genetic approach is required to understand the complexities and causes of target diseases. Recent studies have identified the correlation between genes encoding several transmembrane proteins, such as the cluster of differentiation (CD) and Alzheimer's disease (AD) pathogenesis. In this study, CD48 and CD40 gene expression in AD, a neurodegenerative disease, was analyzed to infer this link. Total RNA sequencing was performed using an Alzheimer's disease mouse model brain and blood, and gene expression was determined using a genome-wide association study (GWAS). We observed a marked elevation of CD48 and CD40 genes in Alzheimer's disease. Indeed, the upregulation of both CD48 and CD40 genes was significantly increased in the severe Alzheimer's disease group. With the elevation of CD48 and CD40 genes in Alzheimer's disease, associations of protein levels were also markedly increased in tissues. In addition, overexpression of CD48 and CD40 genes triggered tau aggregation, and co-expression of these genes accelerated aggregation. The nuclear factor kappa B (NF-ĸB) signaling pathway was enriched by CD48 and CD40 gene expression: it was also associated with tau pathology. Our data suggested that the CD48 and CD40 genes are novel AD-related genes, and this approach may be useful as a diagnostic or therapeutic target for the disease.


Sujet(s)
Maladie d'Alzheimer , Antigènes CD40 , Antigène CD48 , Agrégats de protéines , Protéines tau , Animaux , Souris , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/métabolisme , Antigènes CD40/génétique , Antigènes CD40/métabolisme , Antigène CD48/génétique , Antigène CD48/métabolisme , Expression des gènes , Étude d'association pangénomique , Maladies neurodégénératives/génétique , Maladies neurodégénératives/métabolisme , Agrégats de protéines/génétique , Agrégats de protéines/physiologie , Protéines tau/génétique , Protéines tau/métabolisme
13.
Nat Chem ; 14(9): 1045-1053, 2022 09.
Article de Anglais | MEDLINE | ID: mdl-35798951

RÉSUMÉ

The composition of soluble toxic protein aggregates formed in vivo is currently unknown in neurodegenerative diseases, due to their ultra-low concentration in human biofluids and their high degree of heterogeneity. Here we report a method to capture amyloid-containing aggregates in human biofluids in an unbiased way, a process we name amyloid precipitation. We use a structure-specific chemical dimer, a Y-shaped, bio-inspired small molecule with two capture groups, for amyloid precipitation to increase affinity. Our capture molecule for amyloid precipitation (CAP-1) consists of a derivative of Pittsburgh Compound B (dimer) to target the cross ß-sheets of amyloids and a biotin moiety for surface immobilization. By coupling CAP-1 to magnetic beads, we demonstrate that we can target the amyloid structure of all protein aggregates present in human cerebrospinal fluid, isolate them for analysis and then characterize them using single-molecule fluorescence imaging and mass spectrometry. Amyloid precipitation enables unbiased determination of the molecular composition and structural features of the in vivo aggregates formed in neurodegenerative diseases.


Sujet(s)
Amyloïde , Sécrétions corporelles , Agrégats de protéines , Amyloïde/composition chimique , Peptides bêta-amyloïdes , Sécrétions corporelles/composition chimique , Humains , Agrégats de protéines/physiologie
14.
ACS Chem Neurosci ; 13(16): 2380-2385, 2022 08 17.
Article de Anglais | MEDLINE | ID: mdl-35904551

RÉSUMÉ

Abrupt aggregation of α-synuclein (α-Syn) leads to a formation of highly toxic protein oligomers. These aggregates are the underlying molecular cause of an onset of the irreversible degeneration of dopaminergic neurons in midbrain, hypothalamus, and thalamus, a pathology known as Parkinson's disease. The transient nature of oligomers, as well as their structural and morphological heterogeneity, limits the use of cryo-electron microscopy and solid-state NMR, classical tools of structural biology, for elucidation of their secondary structure. Despite this limitation, numerous pieces of experimental evidence suggest that phospholipids can uniquely alter the structure and toxicity of oligomers. In this study, we utilize an innovative nano-infrared imaging technique, also known as atomic force microscopy infrared (AFM-IR) spectroscopy, to examine the structure of individual α-Syn oligomers grown in the presence of phosphatidylcholine (α-Syn:PC) and phosphatidylserine (α-Syn:PS). We determined the amount of the parallel and the antiparallel ß-sheets, as well as the amount the α-helix and the unordered protein, in the secondary structure of α-Syn:PC and α-Syn:PS formed at day 2 (D2), 8 (D8), and 15 (D15) after initiation of protein aggregation. We found a gradual decrease in the amount of the parallel ß-sheet in both α-Syn:PC and α-Syn:PS from D2 to D15 together with an increase in the α-helix and the unordered protein secondary structure. We infer that this is due to the presence of lipids in the structure of oligomers that prevent an expansion of the parallel ß-sheet upon interaction of the oligomers with monomeric α-Syn.


Sujet(s)
Agrégats de protéines , alpha-Synucléine , Cryomicroscopie électronique , Phosphatidylcholines , Phosphatidylsérine , Agrégats de protéines/physiologie , alpha-Synucléine/métabolisme
15.
Biomolecules ; 12(5)2022 04 30.
Article de Anglais | MEDLINE | ID: mdl-35625583

RÉSUMÉ

Alpha-synuclein (α-syn) is a small protein composed of 140 amino acids and belongs to the group of intrinsically disordered proteins. It is a soluble protein that is highly expressed in neurons and expressed at low levels in glial cells. The monomeric protein aggregation process induces the formation of oligomeric intermediates and proceeds towards fibrillar species. These α-syn conformational species have been detected in the extracellular space and mediate consequences on surrounding neurons and glial cells. In particular, higher-ordered α-syn aggregates are involved in microglial and oligodendrocyte activation, as well as in the induction of astrogliosis. These phenomena lead to mitochondrial dysfunction, reactive oxygen and nitrogen species formation, and the induction of an inflammatory response, associated with neuronal cell death. Several receptors participate in cell activation and/or in the uptake of α-syn, which can vary depending on the α-syn aggregated state and cell types. The receptors involved in this process are of outstanding relevance because they may constitute potential therapeutic targets for the treatment of PD and related synucleinopathies. This review article focuses on the mechanism associated with extracellular α-syn uptake in glial cells and the consequent glial cell activation that contributes to the neuronal death associated with synucleinopathies.


Sujet(s)
Maladie de Parkinson , Synucléinopathies , Humains , Névroglie/métabolisme , Maladie de Parkinson/métabolisme , Agrégats de protéines/physiologie , alpha-Synucléine/métabolisme
16.
Nat Commun ; 13(1): 904, 2022 02 16.
Article de Anglais | MEDLINE | ID: mdl-35173167

RÉSUMÉ

Targeted protein degradation allows targeting undruggable proteins for therapeutic applications as well as eliminating proteins of interest for research purposes. While several degraders that harness the proteasome or the lysosome have been developed, a technology that simultaneously degrades targets and accelerates cellular autophagic flux is still missing. In this study, we develop a general chemical tool and platform technology termed AUTOphagy-TArgeting Chimera (AUTOTAC), which employs bifunctional molecules composed of target-binding ligands linked to autophagy-targeting ligands. AUTOTACs bind the ZZ domain of the otherwise dormant autophagy receptor p62/Sequestosome-1/SQSTM1, which is activated into oligomeric bodies in complex with targets for their sequestration and degradation. We use AUTOTACs to degrade various oncoproteins and degradation-resistant aggregates in neurodegeneration at nanomolar DC50 values in vitro and in vivo. AUTOTAC provides a platform for selective proteolysis in basic research and drug development.


Sujet(s)
Autophagie/physiologie , Lysosomes/métabolisme , Protéines oncogènes/métabolisme , Agrégats de protéines/physiologie , Protéolyse , Lignée cellulaire tumorale , Cellules HeLa , Humains , Liaison aux protéines/physiologie , Pliage des protéines , Homéostasie protéique/physiologie , Transduction du signal
17.
Int J Biol Macromol ; 195: 237-245, 2022 Jan 15.
Article de Anglais | MEDLINE | ID: mdl-34896474

RÉSUMÉ

In present study, a novel glutathione functionalized MoS2 quantum dots (GSH-MoS2 QDs) was synthesized from sodium molybdate dehydrate and glutathione by using a one-pot hydrothermal method. After they were characterized, the influence of GSH-MoS2 QDs on amyloid aggregation of bovine serum albumin (BSA) was investigated by various analytical methods including thioflavin T fluorescence assay, circular dichroism and transmission electron microscope. Moreover, the effect of GSH-MoS2 QDs on cytotoxicity induced by BSA amyloid fibrils and cell penetration were evaluated by MTT assay and confocal fluorescence imaging, respectively. The results indicated that the GSH-MoS2 QDs not only had good water solubility, excellent biocompatibility and low cytotoxicity, but also could obviously inhibit the aggregation of BSA and depolymerize the formed BSA aggregates. The data obtained from this work demonstrated that the GSH-MoS2 QDs is expected to become a candidate drug for the treatment of amyloid-related diseases.


Sujet(s)
Disulfures/composition chimique , Glutathion/composition chimique , Molybdène/composition chimique , Boîtes quantiques/composition chimique , Colorants fluorescents , Glutathion/sang , Glutathion/métabolisme , Imagerie optique/méthodes , Agrégats de protéines/physiologie , Sérumalbumine bovine/composition chimique , Spectrométrie de fluorescence/méthodes
18.
Proc Natl Acad Sci U S A ; 118(49)2021 12 07.
Article de Anglais | MEDLINE | ID: mdl-34873058

RÉSUMÉ

Protein homeostasis is constantly being challenged with protein misfolding that leads to aggregation. Hsp70 is one of the versatile chaperones that interact with misfolded proteins and actively support their folding. Multifunctional Hsp70s are harnessed to specific roles by J-domain proteins (JDPs, also known as Hsp40s). Interaction with the J-domain of these cochaperones stimulates ATP hydrolysis in Hsp70, which stabilizes substrate binding. In eukaryotes, two classes of JDPs, Class A and Class B, engage Hsp70 in the reactivation of aggregated proteins. In most species, excluding metazoans, protein recovery also relies on an Hsp100 disaggregase. Although intensely studied, many mechanistic details of how the two JDP classes regulate protein disaggregation are still unknown. Here, we explore functional differences between the yeast Class A (Ydj1) and Class B (Sis1) JDPs at the individual stages of protein disaggregation. With real-time biochemical tools, we show that Ydj1 alone is superior to Sis1 in aggregate binding, yet it is Sis1 that recruits more Ssa1 molecules to the substrate. This advantage of Sis1 depends on its ability to bind to the EEVD motif of Hsp70, a quality specific to most of Class B JDPs. This second interaction also conditions the Hsp70-induced aggregate modification that boosts its subsequent dissolution by the Hsp104 disaggregase. Our results suggest that the Sis1-mediated chaperone assembly at the aggregate surface potentiates the entropic pulling, driven polypeptide disentanglement, while Ydj1 binding favors the refolding of the solubilized proteins. Such subspecialization of the JDPs across protein reactivation improves the robustness and efficiency of the disaggregation machinery.


Sujet(s)
Protéines du choc thermique HSP40/métabolisme , Protéines du choc thermique HSP70/métabolisme , Agrégats de protéines/physiologie , Protéines de Saccharomyces cerevisiae/métabolisme , Protéines du choc thermique HSP40/génétique , Protéines du choc thermique/métabolisme , Chaperons moléculaires/métabolisme , Liaison aux protéines/physiologie , Domaines protéiques/physiologie , Pliage des protéines , Homéostasie protéique/physiologie , Troubles de l'homéostasie des protéines/métabolisme , Troubles de l'homéostasie des protéines/physiopathologie , Saccharomyces cerevisiae/métabolisme , Protéines de Saccharomyces cerevisiae/génétique , Spécificité du substrat
19.
Invest Ophthalmol Vis Sci ; 62(15): 27, 2021 12 01.
Article de Anglais | MEDLINE | ID: mdl-34964803

RÉSUMÉ

Purpose: Exfoliation syndrome (XFS) is a condition characterized by the production of insoluble fibrillar aggregates (exfoliation material; XFM) in the eye and elsewhere. Many patients with XFS progress to exfoliation glaucoma (XFG), a significant cause of global blindness. We used quantitative mass spectrometry to analyze the composition of XFM in lens capsule specimens and in aqueous humor (AH) samples from patients with XFS, patients with XFG and unaffected individuals. Methods: Pieces of lens capsule and samples of AH were obtained with consent from patients undergoing cataract surgery. Tryptic digests of capsule or AH were analyzed by high-performance liquid chromatography-mass spectrometry and relative differences between samples were quantified using the tandem mass tag technique. The distribution of XFM on the capsular surface was visualized by SEM and super-resolution light microscopy. Results: A small set of proteins was consistently upregulated in capsule samples from patients with XFS and patients with XFG, including microfibril components fibrillin-1, latent transforming growth factor-ß-binding protein-2 and latent transforming growth factor-ß-binding protein-3. Lysyl oxidase-like 1, a cross-linking enzyme associated with XFS in genetic studies, was an abundant XFM constituent. Ligands of the transforming growth factor-ß superfamily were prominent, including LEFTY2, a protein best known for its role in establishing the embryonic body axis. Elevated levels of LEFTY2 were also detected in AH from patients with XFG, a finding confirmed subsequently by ELISA. Conclusions: This analysis verified the presence of suspected XFM proteins and identified novel components. Quantitative comparisons between patient samples revealed a consistent XFM proteome characterized by strong expression of fibrillin-1, lysyl oxidase-like-1, and LEFTY2. Elevated levels of LEFTY2 in the AH of patients with XFG may serve as a biomarker for the disease.


Sujet(s)
Humeur aqueuse/métabolisme , Cristallines/métabolisme , Glaucome capsulaire/métabolisme , Glaucome à angle ouvert/métabolisme , Capsule du cristallin/métabolisme , Agrégats de protéines/physiologie , Sujet âgé , Sujet âgé de 80 ans ou plus , Amino-acid oxidoreductases/métabolisme , Chromatographie en phase liquide à haute performance , Cristallines/ultrastructure , Test ELISA , Femelle , Fibrilline-1/métabolisme , Technique d'immunofluorescence indirecte , Humains , Protéines de liaison au TGF-bêta latent/métabolisme , Facteurs de détermination de l'asymétrie droite-gauche/métabolisme , Capsule du cristallin/ultrastructure , Mâle , Spectrométrie de masse , Microscopie électronique à balayage , Adulte d'âge moyen
20.
Int J Mol Sci ; 22(24)2021 Dec 13.
Article de Anglais | MEDLINE | ID: mdl-34948195

RÉSUMÉ

Parkinson's disease (PD) is a neurodegenerative disease characterized by the loss of dopamine neurons and the deposition of misfolded proteins known as Lewy bodies (LBs), which contain α-synuclein (α-syn). The causes and molecular mechanisms of PD are not clearly understood to date. However, misfolded proteins, oxidative stress, and impaired autophagy are believed to play important roles in the pathogenesis of PD. Importantly, α-syn is considered a key player in the development of PD. The present study aimed to assess the role of Ellagic acid (EA), a polyphenol found in many fruits, on α-syn aggregation and toxicity. Using thioflavin and seeding polymerization assays, in addition to electron microscopy, we found that EA could dramatically reduce α-syn aggregation. Moreover, EA significantly mitigated the aggregated α-syn-induced toxicity in SH-SY5Y cells and thus enhanced their viability. Mechanistically, these cytoprotective effects of EA are mediated by the suppression of apoptotic proteins BAX and p53 and a concomitant increase in the anti-apoptotic protein, BCL-2. Interestingly, EA was able to activate autophagy in SH-SY5Y cells, as evidenced by normalized/enhanced expression of LC3-II, p62, and pAKT. Together, our findings suggest that EA may attenuate α-syn toxicity by preventing aggregation and improving viability by restoring autophagy and suppressing apoptosis.


Sujet(s)
Acide ellagique/pharmacologie , Agrégation pathologique de protéines/prévention et contrôle , alpha-Synucléine/métabolisme , Apoptose/physiologie , Autophagie/physiologie , Lignée cellulaire tumorale , Neurones dopaminergiques/métabolisme , Acide ellagique/métabolisme , Humains , Corps de Lewy/métabolisme , Maladies neurodégénératives/métabolisme , Maladie de Parkinson/métabolisme , Agrégats de protéines/physiologie , Agrégation pathologique de protéines/métabolisme , alpha-Synucléine/physiologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...