Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Neurologia (Engl Ed) ; 39(4): 321-328, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38616059

RESUMO

INTRODUCTION: The aim of this study was to compare the effect of five types of PEGlated nanoliposomes (PNLs) on α-synuclein (α-syn) fibrillization, attenuation of microglial activation, and silence of the SNCA gene, which encodes α-syn. METHODS: To evaluate the inhibition of α-syn fibrillization, we used standard in vitro assay based on Thioflavin T (ThT) fluorescence. Next, to evaluate the attenuation of microglial activation, the concentration of TNF-a and IL-6 was quantified by ELISA assay in BV2 microglia cells treated with 100nM A53T α-syn and PNLs. In order to determine the silencing of the SNCA, real-time PCR and Western blot analysis was used. Finally, the efficacy of PNLs was confirmed in a transgenic mouse model expressing human α-syn. RESULTS: ThT assay showed both PNL1 and PNL2 significantly inhibited a-syn fibrillization. ELISA test also showed the production of TNF-a and IL-6 was significantly attenuated when microglial cells treated with PNL1 or PNL2. We also found that SNCA gene, at both mRNA and protein levels, was significantly silenced when BV2 microglia cells were treated with PNL1 or PNL2. Importantly, the efficacy of PNL1 and PNL2 was finally confirmed in vivo in a transgenic mouse model. CONCLUSIONS: In conclusion, the novel multifunctional nanoliposomes tested in our study inhibit α-syn fibrillization, attenuate microglial activation, and silence SNCA gene. Our findings suggest the therapeutic potential of PNL1 and PNL2 for treating synucleinopathies.


Assuntos
Microglia , alfa-Sinucleína , Humanos , Animais , Camundongos , alfa-Sinucleína/genética , Interleucina-6 , Modelos Animais de Doenças , Camundongos Transgênicos
2.
Macromol Biosci ; : e2300579, 2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38552257

RESUMO

Throughout the past decades, amphipathic peptide-based hydrogels have proven to be promising materials for biomedical applications. Amphipathic peptides are known to adopt ß-sheet configurations that self-assemble into fibers that then interact to form a hydrogel network. A fundamental understanding of how the peptide sequence alters the structural properties of the hydrogels would allow for a more rational design of novel peptides for a variety of biomedical applications in the future. Therefore, the current work investigates how changing the type of amino acid, the amphipathic pattern, and the peptide length affects the secondary structure, fiber characteristics, and stiffness of peptide-based hydrogels. Hereto, seven amphipathic peptides of different sequence and length, four of which have not been previously reported, based on and including the hexapeptide H-Phe-Gln-Phe-Gln-Phe-Lys-NH2, are synthesized and thoroughly characterized by circular dichroism (CD), Fourier Transform Infrared (FTIR) spectroscopy, Wide Angle X-ray Scattering (WAXS), Small Angle X-ray Scattering (SAXS), Transmission Electron Microscopy (TEM), and Thioflavin T (ThT) fibrillization assays. The results show that a high amount of regularly spaced ß-sheets, a high amount of fibers, and fiber bundling contribute to the stiffness of the hydrogel. Furthermore, a study of the time-dependent fibril formation process reveals complex transient dynamics. The peptide strands structure through an intermediate helical state prior to ß-sheet formation, which is found to be concentration- and time-dependent.

3.
Molecules ; 28(18)2023 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-37764331

RESUMO

Molecular crowding environments play a crucial role in understanding the mechanisms of biological reactions. Inside living cells, a diverse array of molecules coexists within a volume fraction ranging from 10% to 30% v/v. However, conventional spectroscopic methods often face difficulties in selectively observing the structures of particular proteins or membranes within such molecularly crowded environments due to the presence of high background signals. Therefore, it is crucial to establish in vitro measurement conditions that closely resemble the intracellular environment. Meanwhile, the neutron scattering method offers a significant advantage in selectively observing target biological components, even within crowded environments. Recently, we have demonstrated a novel scattering method capable of selectively detecting the structures of targeted proteins or membranes in a closely mimicking intracellular milieu achieved utilizing whole-cell contents (deuterated-cell debris). This method relies on the inverse contrast matching technique in neutron scattering. By employing this method, we successfully observed the fibrillization process of human amyloid beta-protein (Aß 1-40) under a molecular crowding environment (13.1% w/v cell debris, Aß/cell debris = ~1/25 w/w) that closely mimics the interior of living cells. Aß protein is well known as a major pathogenic component of Alzheimer's disease. The present results combining model simulation analyses clearly show that the intracellular environment facilitates the potential formation of even more intricate higher-order aggregates of Aß proteins than those previously reported.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Humanos , Simulação por Computador , Nêutrons
4.
ASN Neuro ; 15: 17590914231184086, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37428128

RESUMO

There is a critical need for small molecules capable of rescuing pathophysiological phenotypes induced by alpha-synuclein (aSyn) misfolding and oligomerization. Building upon our previous aSyn cellular fluorescence lifetime (FLT)-Förster resonance energy transfer (FRET) biosensors, we have developed an inducible cell model incorporating the red-shifted mCyRFP1/mMaroon1 (OFP/MFP) FRET pair. This new aSyn FRET biosensor improves the signal-to-noise ratio, reduces nonspecific background FRET, and results in a 4-fold increase (transient transfection) and 2-fold increase (stable, inducible cell lines) in FRET signal relative to our previous GFP/RFP aSyn biosensors. The inducible system institutes greater temporal control and scalability, allowing for fine-tuning of biosensor expression and minimizes cellular cytotoxicity due to overexpression of aSyn. Using these inducible aSyn-OFP/MFP biosensors, we screened the Selleck library of 2684 commercially available, FDA-approved compounds and identified proanthocyanidins and casanthranol as novel hits. Secondary assays validated the ability of these compounds to modulate aSyn FLT-FRET. Functional assays probing cellular cytotoxicity and aSyn fibrillization demonstrated their capability to inhibit seeded aSyn fibrillization. Proanthocyanidins completely rescued aSyn fibril-induced cellular toxicity with EC50 of 200 nM and casanthranol supported a 85.5% rescue with a projected EC50 of 34.2 µM. Furthermore, proanthocyanidins provide a valuable tool compound to validate our aSyn biosensor performance in future high-throughput screening campaigns of industrial-scale (million-compound) chemical libraries.


Assuntos
Técnicas Biossensoriais , Emodina , Proantocianidinas , alfa-Sinucleína/metabolismo , Transferência Ressonante de Energia de Fluorescência/métodos , Ensaios de Triagem em Larga Escala
5.
Front Chem ; 11: 1145877, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37304685

RESUMO

Parkinson's disease is an age-related neurological disorder, and the pathology of the disease is linked to different types of aggregates of α-synuclein or alpha-synuclein (aS), which is an intrinsically disordered protein. The C-terminal domain (residues 96-140) of the protein is highly fluctuating and possesses random/disordered coil conformation. Thus, the region plays a significant role in the protein's solubility and stability by an interaction with other parts of the protein. In the current investigation, we examined the structure and aggregation behavior of two artificial single point mutations at a C-terminal residue at position 129 that represent a serine residue in the wild-type human aS (wt aS). Circular Dichroism (CD) and Raman spectroscopy were performed to analyse the secondary structure of the mutated proteins and compare it to the wt aS. Thioflavin T assay and atomic force microscopy imaging helped in understanding the aggregation kinetics and type of aggregates formed. Finally, the cytotoxicity assay gave an idea about the toxicity of the aggregates formed at different stages of incubation due to mutations. Compared to wt aS, the mutants S129A and S129W imparted structural stability and showed enhanced propensity toward the α-helical secondary structure. CD analysis showed proclivity of the mutant proteins toward α-helical conformation. The enhancement of α-helical propensity lengthened the lag phase of fibril formation. The growth rate of ß-sheet-rich fibrillation was also reduced. Cytotoxicity tests on SH-SY5Y neuronal cell lines established that the S129A and S129W mutants and their aggregates were potentially less toxic than wt aS. The average survivability rate was ∼40% for cells treated with oligomers (presumably formed after 24 h of incubation of the freshly prepared monomeric protein solution) produced from wt aS and ∼80% for cells treated with oligomers obtained from mutant proteins. The relative structural stability with α-helical propensity of the mutants could be a plausible reason for their slow rate of oligomerization and fibrillation, and this was also the possible reason for reduced toxicity to neuronal cells.

6.
Int J Biol Macromol ; 239: 124197, 2023 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-36972817

RESUMO

Misfolding and protein aggregation have been linked to numerous human neurodegenerative disorders such as Alzheimer's, prion, and Parkinson's diseases. Ruthenium (Ru) complexes have received considerable attention in studying protein aggregation due to their interesting photophysical and photo properties. In this study, we have synthesized the novel Ru complexes ([Ru(p-cymene)Cl(L-1)][PF6](Ru-1), and [Ru(p-cymene)Cl(L-2)][PF6](Ru-2)) and investigated their inhibitory activity against the bovine serum albumin (BSA) aggregation and the Aß1-42 peptides amyloid formation. Several spectroscopic methods were used to characterize these complexes, and the molecular structure of the complex was determined by X-ray crystallography. Amyloid aggregation and inhibition activities were examined using the Thioflavin-T (ThT) assay, and the secondary structures of the protein were analyzed by circular dichroism (CD) spectroscopy and transmission electron microscopy (TEM). The cell viability assay was carried out on the neuroblastoma cell line, revealing that the complex Ru-2 showed better protective effects against Aß1-42 peptide toxicity on neuro-2a cells than the complex Ru-1. Molecular docking studies elucidate the binding sites and interactions between the Ru-complexes and Aß1-42 peptides. The experimental studies revealed that these complexes significantly inhibited the BSA aggregation and Aß1-42 amyloid fibril formation at 1:3 and 1:1 molar concentrations, respectively. Antioxidant assays demonstrated that these complexes act as antioxidants, protecting from amyloid-induced oxidative stress. Molecular docking studies with the monomeric Aß1-42 (PDB: 1IYT) show hydrophobic interaction, and both complexes bind preferably in the central region of the peptide and coordinate with two binding sites of the peptide. Hence, we suggest that the Ru-based complexes could be applied as a potential agent in metallopharmaceutical research against Alzheimer's disease.


Assuntos
Complexos de Coordenação , Rutênio , Humanos , Peptídeos beta-Amiloides/metabolismo , Agregados Proteicos , Rutênio/farmacologia , Rutênio/química , Simulação de Acoplamento Molecular , Proteínas Amiloidogênicas , Amiloide/metabolismo , Complexos de Coordenação/química
7.
Protein Sci ; 32(4): e4610, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36851846

RESUMO

Transthyretin (TTR)-related amyloidosis (ATTR) is a syndrome of diseases characterized by the extracellular deposition of fibrillar materials containing TTR variants. Ala97Ser (A97S) is the major mutation reported in Taiwanese ATTR patients. Here, we combine atomic resolution structural information together with the biochemical data to demonstrate that substitution of polar Ser for a small hydrophobic side chain of Ala at residue 97 of TTR largely influences the local packing density of the FG-loop, thus leading to the conformational instability of native tetramer, the increased monomeric species, and thus the enhanced amyloidogenicity of apo-A97S. Based on calorimetric studies, the tetramer destabilization of A97S can be substantially altered by interacting with native stabilizers via similarly energetic patterns compared to that of wild-type (WT) TTR; however, stabilizer binding partially rearranges the networks of hydrogen bonding in TTR variants while FG-loops of tetrameric A97S still remain relatively flexible. Moreover, TTR in complexed with holo-retinol binding protein 4 is slightly influenced by the structural and dynamic changes of FG-loop caused by A97S substitution with an approximately five-fold difference in binding affinity. Collectively, our findings suggest that the amyloidogenic A97S mutation destabilizes TTR by increasing the flexibility of the FG-loop in the monomer, thus modulating the rate of amyloid fibrillization.


Assuntos
Amiloide , Pré-Albumina , Humanos , Amiloide/química , Proteínas Amiloidogênicas/genética , Calorimetria , Mutação , Pré-Albumina/genética , Pré-Albumina/química
8.
Adv Mater ; 35(6): e2208211, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36418914

RESUMO

The structural order and aggregation of non-fullerene acceptors (NFA) are critical toward light absorption, phase separation, and charge transport properties of their photovoltaic blends with electron donors, and determine the power conversion efficiency (PCE) of the corresponding organic solar cells (OSCs). In this work, the fibrillization of small molecular NFA L8-BO with the assistance of fused-ring solvent additive 1-fluoronaphthalene (FN) to substantially improve device PCE is demonstrated. Molecular dynamics simulations show that FN attaches to the backbone of L8-BO as the molecular bridge to enhance the intermolecular packing , inducing 1D self-assembly of L8-BO into fine fibrils with a compact polycrystal structure. The L8-BO fibrils are incorporated into a pseudo-bulk heterojunction (P-BHJ) active layer with D18 as a donor, and show enhanced light absorption, charge transport, and collection properties, leading to enhanced PCE from 16.0% to an unprecedented 19.0% in the D18/L8-BO binary P-BHJ OSC, featuring a high fill factor of 80%. This work demonstrates a strategy for fibrillating NFAs toward the enhanced performance of OSCs.

9.
Methods Mol Biol ; 2551: 63-77, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36310197

RESUMO

We employ time-resolved in situ atomic force microcopy to monitor the growth of individual Aß40 fibrils and thereby directly measure the fibril growth rates. We describe procedures to express and purify the Aß peptide and verify its identity, prepare solutions and seeds, quantify the displacements of the growing tips of individual fibrils, and determine their respective growth rates. We discuss approaches to evaluate and minimize the impact of the scanning tip on the monitored processes. We use the distribution of fibril thickness to characterize approximately the fibril structure. The ability to quantify faithfully the growth kinetics of amyloid fibrils empowers exploration of the molecular-level processes of fibril growth that relate to behaviors of amyloid species of laboratory and clinical interest.


Assuntos
Peptídeos beta-Amiloides , Amiloide , Peptídeos beta-Amiloides/química , Amiloide/química , Cinética , Fragmentos de Peptídeos/química
10.
J Biol Chem ; 298(12): 102662, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36334629

RESUMO

Fibrillization of the protein amyloid ß is assumed to trigger Alzheimer's pathology. Approaches that target amyloid plaques, however, have garnered limited clinical success, and their failures may relate to the scarce understanding of the impact of potential drugs on the intertwined stages of fibrillization. Here, we demonstrate that bexarotene, a T-cell lymphoma medication with known antiamyloid activity both in vitro and in vivo, suppresses amyloid fibrillization by promoting an alternative fibril structure. We employ time-resolved in situ atomic force microscopy to quantify the kinetics of growth of individual fibrils and supplement it with structure characterization by cryo-EM. We show that fibrils with structure engineered by the drug nucleate and grow substantially slower than "normal" fibrils; remarkably, growth remains stunted even in drug-free solutions. We find that the suppression of fibril growth by bexarotene is not because of the drug binding to the fibril tips or to the peptides in the solution. Kinetic analyses attribute the slow growth of drug-enforced fibril polymorph to the distinctive dynamics of peptide chain association to their tips. As an additional benefit, the bexarotene fibrils kill primary rat hippocampal neurons less efficiently than normal fibrils. In conclusion, the suggested drug-driven polymorph transformation presents a mode of action to irreversibly suppress toxic aggregates not only in Alzheimer's but also potentially in myriad diverse pathologies that originate with protein condensation.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Animais , Ratos , Peptídeos beta-Amiloides/metabolismo , Doença de Alzheimer/metabolismo , Bexaroteno/farmacologia , Amiloide/química , Placa Amiloide , Fragmentos de Peptídeos/química
11.
Biomedicines ; 10(7)2022 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-35884817

RESUMO

We here report a novel strategy to control the bioavailability of the fibrillizing parathyroid hormone (PTH)-derived peptides, where the concentration of the bioactive form is controlled by an reversible, photoswitchable peptide. PTH1-84, a human hormone secreted by the parathyroid glands, is important for the maintenance of extracellular fluid calcium and phosphorus homeostasis. Controlling fibrillization of PTH1-84 represents an important approach for in vivo applications, in view of the pharmaceutical applications for this protein. We embed the azobenzene derivate 3-{[(4-aminomethyl)phenyl]diazenyl}benzoic acid (3,4'-AMPB) into the PTH-derived peptide PTH25-37 to generate the artificial peptide AzoPTH25-37 via solid-phase synthesis. AzoPTH25-37 shows excellent photostability (more than 20 h in the dark) and can be reversibly photoswitched between its cis/trans forms. As investigated by ThT-monitored fibrillization assays, the trans-form of AzoPTH25-37 fibrillizes similar to PTH25-37, while the cis-form of AzoPTH25-37 generates only amorphous aggregates. Additionally, cis-AzoPTH25-37 catalytically inhibits the fibrillization of PTH25-37 in ratios of up to one-fifth. The approach reported here is designed to control the concentration of PTH-peptides, where the bioactive form can be catalytically controlled by an added photoswitchable peptide.

12.
Int J Biol Macromol ; 216: 414-425, 2022 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-35803407

RESUMO

Even though amyloid aggregates were discovered many years ago the mechanism of their formation is still a mystery. Because of their connection to many of untreatable neurodegenerative diseases the motivation for finding a common aggregation path is high. We report a new high heat induced fibrillization path of a model protein ß-lactoglobulin (BLG) when incubated in glycine instead of water at pH 2. By combining atomic force microscopy (AFM), transmission emission microscopy (TEM), dynamic light scattering (DLS) and circular dichroism (CD) we predict that the basic building blocks of fibrils made in glycine are not peptides, but rather spheroid oligomers of different height that form by stacking of ring-like structures. Spheroid oligomers linearly align to form fibrils by opening up and combining. We suspect that glycine acts as an hydrolysation inhibitor which consequently promotes a different fibrillization path. By combining the known data on fibrillization in water with our experimental conclusions we come up with a new fibrillization scheme for BLG. We show that by changing the fibrillization conditions just by small changes in buffer composition can dramatically change the aggregation pathway and the effect of buffer shouldn't be neglected. Fibrils seen in our study are also gaining more and more attention because of their pore-like structure and a possible cytotoxic mechanism by forming pernicious ion-channels. By preparing them in a simple model system as BLG we opened a new way to study their formation.


Assuntos
Amiloide , Lactoglobulinas , Amiloide/química , Glicina/farmacologia , Lactoglobulinas/química , Microscopia de Força Atômica/métodos , Água
13.
Colloids Surf B Biointerfaces ; 217: 112670, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35779329

RESUMO

BACKGROUND: Poly(3-caprolactone) (PCL)/ß-tricalcium phosphate (ß-TCP) composite scaffolds fabricated by three-dimensional (3D) printing are one of the common scaffolds for bone tissue regeneration. However, the main challenge of these 3D printed PCL/ß-TCP scaffolds is the fact that many cells pass from porosities during in vitro cell seeding, leading to poor initial cell attachment. This study aimed to demonstrate the fabrication of a new collagen coating process for optimizing the hydrophilic property and cell-substrate interactions. This method may be used for coating collagen on any relevant biomedical constructs made of synthetic polymers to increase their biocompatibility and cell attachment. MATERIALS AND METHODS: Porous composite scaffolds fabricated by 3D printing were coated with collagen by a novel method and compared to traditional methods. After plasma treatment, samples were inverted in a homogenized collagen solution, freeze-dried, stabilized by crosslinking, freeze-dried again, and fibrillated using a defined salt concentration. Samples were characterized by a 3D laser microscope, cytocompatibility assay, attenuated total reflectance Fourier-transform infrared (ATR-FTIR) spectroscopy, water absorption, protein absorption, and bioactivity assay. RESULTS: Homogenized collagen at pH= 7 resulted in a very uniform layer on the surface of scaffolds with significantly higher cell proliferation (p < 0.05). Collagen-coated scaffolds showed significantly higher water absorption, protein absorption, and bioactivity compared to non-coated samples (p < 0.05). CONCLUSION: The results demonstrate that both the pH and collagen structure influence the coating of scaffolds, while the concentrations used in this study do not have a significant difference in this aspect. The combination of homogenization and fibrillization makes scaffolds more biocompatible and desirable for bone tissue engineering.


Assuntos
Poliésteres , Alicerces Teciduais , Colágeno/química , Colágeno/farmacologia , Poliésteres/química , Poliésteres/farmacologia , Impressão Tridimensional , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Água
14.
J Biol Chem ; 298(7): 102071, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35643314

RESUMO

Alzheimer's disease (AD) involves the neurotoxic self-assembly of a 40 and 42 residue peptide, Amyloid-ß (Aß). Inherited early-onset AD can be caused by single point mutations within the Aß sequence, including Arctic (E22G) and Italian (E22K) familial mutants. These mutations are heterozygous, resulting in an equal proportion of the WT and mutant Aß isoform expression. It is therefore important to understand how these mixtures of Aß isoforms interact with each other and influence the kinetics and morphology of their assembly into oligomers and fibrils. Using small amounts of nucleating fibril seeds, here, we systematically monitored the kinetics of fibril formation, comparing self-seeding with cross-seeding behavior of a range of isoform mixtures of Aß42 and Aß40. We confirm that Aß40(WT) does not readily cross-seed Aß42(WT) fibril formation. In contrast, fibril formation of Aß40(Arctic) is hugely accelerated by Aß42(WT) fibrils, causing an eight-fold reduction in the lag-time to fibrillization. We propose that cross-seeding between the more abundant Aß40(Arctic) and Aß42(WT) may be important for driving early-onset AD and will propagate fibril morphology as indicated by fibril twist periodicity. This kinetic behavior is not emulated by the Italian mutant, where minimal cross-seeding is observed. In addition, we studied the cross-seeding behavior of a C-terminal-amidated Aß42 analog to probe the coulombic charge interplay between Glu22/Asp23/Lys28 and the C-terminal carboxylate. Overall, these studies highlight the role of cross-seeding between WT and mutant Aß40/42 isoforms, which can impact the rate and structure of fibril assembly.


Assuntos
Doença de Alzheimer , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Amiloide/genética , Peptídeos beta-Amiloides/metabolismo , Humanos , Cinética , Mutação , Fragmentos de Peptídeos/metabolismo
15.
Spectrochim Acta A Mol Biomol Spectrosc ; 278: 121372, 2022 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-35588606

RESUMO

In this study, the acceleratory effect of magnesium oxide nanoparticles (MgO NPs) on the amyloid fibrillization of human tau protein, a major protein involved in the onset of Alzheimer's disease (AD) was investigated. The MgO NPs were fabricated through laser ablation synthesis in solution (LASiS), well-characterized, and explored further for tau aggregation and relevant neurotoxicity by different assays. The results showed that the MgO NPs have a size of around 30 nm, a hydrodynamic radius of 57.09 nm, and a zeta potential of -18.06 mV. The data from ThT and ANS fluorescence-based assays along with circular dichroism (CD) spectroscopy clearly indicated that MgO NPs could significantly promote tau fibrillization, concentration-dependently. Considering the acceleratory effect of MgO NPs against tau fibrillization, cellular assays including cell viability, reactive oxygen species (ROS), and caspase-3 assays indicated that the neurotoxicity of tau amyloid fibrils formed with MgO NPs was higher than that of tau samples aged alone against N2a neuron-like cells. Therefore, it was concluded that the interaction of MgO NPs with tau can lead to acceleration of tau aggregation and underlying neurotoxicity. This study, then can provide useful information about the direct effect of MgO NPs against memory proteins and subsequent adverse effects.


Assuntos
Terapia a Laser , Nanopartículas , Idoso , Amiloide , Humanos , Óxido de Magnésio/química , Nanopartículas/química , Nanopartículas/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Proteínas tau
16.
Proc Natl Acad Sci U S A ; 119(5)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35082148

RESUMO

Triggering receptor expressed on myeloid cells 2 (TREM2) is a single-pass transmembrane receptor of the immunoglobulin superfamily that is secreted in a soluble (sTREM2) form. Mutations in TREM2 have been linked to increased risk of Alzheimer's disease (AD). A prominent neuropathological component of AD is deposition of the amyloid-ß (Aß) into plaques, particularly Aß40 and Aß42. While the membrane-bound form of TREM2 is known to facilitate uptake of Aß fibrils and the polarization of microglial processes toward amyloid plaques, the role of its soluble ectodomain, particularly in interactions with monomeric or fibrillar Aß, has been less clear. Our results demonstrate that sTREM2 does not bind to monomeric Aß40 and Aß42, even at a high micromolar concentration, while it does bind to fibrillar Aß42 and Aß40 with equal affinities (2.6 ± 0.3 µM and 2.3 ± 0.4 µM). Kinetic analysis shows that sTREM2 inhibits the secondary nucleation step in the fibrillization of Aß, while having little effect on the primary nucleation pathway. Furthermore, binding of sTREM2 to fibrils markedly enhanced uptake of fibrils into human microglial and neuroglioma derived cell lines. The disease-associated sTREM2 mutant, R47H, displayed little to no effect on fibril nucleation and binding, but it decreased uptake and functional responses markedly. We also probed the structure of the WT sTREM2-Aß fibril complex using integrative molecular modeling based primarily on the cross-linking mass spectrometry data. The model shows that sTREM2 binds fibrils along one face of the structure, leaving a second, mutation-sensitive site free to mediate cellular binding and uptake.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Amiloide/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Imunológicos/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Amiloide/genética , Peptídeos beta-Amiloides/genética , Animais , Humanos , Cinética , Glicoproteínas de Membrana/genética , Camundongos , Microglia/metabolismo , Mutação/genética , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/genética , Placa Amiloide/metabolismo , Receptores Imunológicos/genética , Proteínas tau/genética , Proteínas tau/metabolismo
17.
Int J Biol Macromol ; 192: 491-497, 2021 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-34599991

RESUMO

There are several studies reporting that different plant-based metabolites are potential inhibitors of protein amyloid fibrillation. As chemical features of metabolites can regulate protein aggregation process, in the present in vitro investigation, tau protein was selected as a model of Alzheimer's disease to elaborate the inhibitory effect of syringic acid (SA) on its assembly and associated neurotoxicity in aggregation conditions. Extrinsic fluorescence, Congo red adsorption, and CD spectroscopic studies, TEM, size-exclusion chromatography, and MALDI-TOF mass spectrometry analysis along with MTT and qRT-PCR assays were performed to assess the inhibitory effects of SA against tau aggregation and neurotoxicity. It was shown that SA has the tendency to control the aggregation of the tau proteins through modulating the amyloid kinetic parameters, exposure of hydrophobic residues, and structural changes. Moreover, the structures formed in the presence of SA recovered the viability of neuron-like cells (SH-SY5Y) through regulation of endoplasmic reticulum stress signaling pathway by downregulation of ATF-6, caspase-8 and caspase-3 mRNA. In conclusion, it can be suggested that SA may be used as a potential small molecule in the development of therapeutic platforms against Alzheimer's disease.


Assuntos
Amiloide/antagonistas & inibidores , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Ácido Gálico/análogos & derivados , Fármacos Neuroprotetores/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteínas tau/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Amiloide/metabolismo , Apoptose/efeitos dos fármacos , Ácido Gálico/farmacologia , Humanos , Cinética , Agregados Proteicos/efeitos dos fármacos , Agregação Patológica de Proteínas , Conformação Proteica , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
18.
Neurobiol Dis ; 161: 105536, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34718129

RESUMO

The accumulation of hyperphosphorylated fibrillar Tau aggregates in the brain is one of the defining hallmarks of Tauopathy diseases, including Alzheimer's disease. However, the primary events or molecules responsible for initiation of the pathological Tau aggregation and spreading remain unknown. The discovery of heparin as an effective inducer of Tau aggregation in vitro was instrumental to enabling different lines of research into the role of Tau aggregation in the pathogenesis of Tauopathies. However, recent proteomics and cryogenic electron microscopy (cryo-EM) studies have revealed that heparin-induced Tau fibrils generated in vitro do not reproduce the biochemical and ultrastructural properties of disease-associated brain-derived Tau fibrils. These observations demand that we reassess our current approaches for investigating the mechanisms underpinning Tau aggregation and pathology formation. Our review article presents an up-to-date survey and analyses of 1) the evolution of our understanding of the interactions between Tau and heparin, 2) the various structural and mechanistic models of the heparin-induced Tau aggregation, 3) the similarities and differences between brain-derived and heparin-induced Tau fibrils; and 4) emerging concepts on the biochemical and structural determinants underpinning Tau pathological heterogeneity in Tauopathies. Our analyses identify specific knowledge gaps and call for 1) embracing the complexities of Tau pathologies; 2) reassessment of current approaches to investigate, model and reproduce pathological Tau aggregation as it occurs in the brain; 3) more research towards a better understanding of the naturally-occurring cofactor molecules that are associated with Tau brain pathology initiation and propagation; and 4) developing improved approaches for in vitro production of the Tau aggregates and fibrils that recapitulate and/or amplify the biochemical and structural complexity and diversity of pathological Tau in Tauopathies. This will result in better and more relevant tools, assays, and mechanistic models, which could significantly improve translational research and the development of drugs and antibodies that have higher chances for success in the clinic.


Assuntos
Doença de Alzheimer , Tauopatias , Doença de Alzheimer/patologia , Encéfalo/metabolismo , Humanos , Tauopatias/patologia , Proteínas tau/metabolismo
19.
Proc Natl Acad Sci U S A ; 118(38)2021 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-34518234

RESUMO

Amyloid fibrillization is an exceedingly complex process in which incoming peptide chains bind to the fibril while concertedly folding. The coupling between folding and binding is not fully understood. We explore the molecular pathways of association of Aß40 monomers to fibril tips by combining time-resolved in situ scanning probe microscopy with molecular modeling. The comparison between experimental and simulation results shows that a complex supported by nonnative contacts is present in the equilibrium structure of the fibril tip and impedes fibril growth in a supersaturated solution. The unraveling of this frustrated state determines the rate of fibril growth. The kinetics of growth of freshly cut fibrils, in which the bulk fibril structure persists at the tip, complemented by molecular simulations, indicate that this frustrated complex comprises three or four monomers in nonnative conformations and likely is contained on the top of a single stack of peptide chains in the fibril structure. This pathway of fibril growth strongly deviates from the common view that the conformational transformation of each captured peptide chain is templated by the previously arrived peptide. The insights into the ensemble structure of the frustrated complex may guide the search for suppressors of Aß fibrillization. The uncovered dynamics of coupled structuring and assembly during fibril growth are more complex than during the folding of most globular proteins, as they involve the collective motions of several peptide chains that are not guided by a funneled energy landscape.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Amiloide/metabolismo , Fragmentos de Peptídeos/metabolismo , Cinética , Simulação de Dinâmica Molecular , Dobramento de Proteína
20.
Bioorg Med Chem ; 43: 116247, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34157569

RESUMO

Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized, in part, by the misfolding, oligomerization and fibrillization of amyloid-ß (Aß). Evidence suggests that the mechanisms underpinning Aß oligomerization and subsequent fibrillization are distinct, and may therefore require equally distinct therapeutic approaches. Prior studies have suggested that amide derivatives of ferulic acid, a natural polyphenol, may combat multiple AD pathologies, though its impact on Aß aggregation is controversial. We designed and synthesized a systematic library of amide derivatives of ferulic acid and evaluated their anti-oligomeric and anti-fibrillary capacities independently. Azetidine tethered, triphenyl derivatives were the most potent anti-oligomeric agents (compound 2i: IC50 = 1.8 µM ± 0.73 µM); notably these were only modest anti-fibrillary agents (20.57% inhibition of fibrillization), and exemplify the poor correlation between anti-oligomeric/fibrillary activities. These data were subsequently codified in an in silico QSAR model, which yielded a strong predictive model of anti-Aß oligomeric activity (κ = 0.919 for test set; κ = 0.737 for validation set).


Assuntos
Doença de Alzheimer/tratamento farmacológico , Amiloide/antagonistas & inibidores , Ácidos Cumáricos/farmacologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Amidas , Amiloide/metabolismo , Ácidos Cumáricos/química , Relação Dose-Resposta a Droga , Humanos , Estrutura Molecular , Agregados Proteicos/efeitos dos fármacos , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA