Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.234
Filter
1.
Molecules ; 29(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38731472

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by the accumulation of amyloid beta (Aß) plaques in the brain. Aß1-42 is the main component of Aß plaque, which is toxic to neuronal cells. Si nanowires (Si NWs) have the advantages of small particle size, high specific surface area, and good biocompatibility, and have potential application prospects in suppressing Aß aggregation. In this study, we employed the vapor-liquid-solid (VLS) growth mechanism to grow Si NWs using Au nanoparticles as catalysts in a plasma-enhanced chemical vapor deposition (PECVD) system. Subsequently, these Si NWs were transferred to a phosphoric acid buffer solution (PBS). We found that Si NWs significantly reduced cell death in PC12 cells (rat adrenal pheochromocytoma cells) induced by Aß1-42 oligomers via double staining with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and fluorescein diacetate/propyl iodide (FDA/PI). Most importantly, pre-incubated Si NWs largely prevented Aß1-42 oligomer-induced PC12 cell death, suggesting that Si NWs exerts an anti-Aß neuroprotective effect by inhibiting Aß aggregation. The analysis of Fourier Transform Infrared (FTIR) results demonstrates that Si NWs reduce the toxicity of fibrils and oligomers by intervening in the formation of ß-sheet structures, thereby protecting the viability of nerve cells. Our findings suggest that Si NWs may be a potential therapeutic agent for AD by protecting neuronal cells from the toxicity of Aß1-42.


Subject(s)
Amyloid beta-Peptides , Nanowires , Silicon , Amyloid beta-Peptides/toxicity , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Nanowires/chemistry , Animals , PC12 Cells , Rats , Silicon/chemistry , Peptide Fragments/chemistry , Peptide Fragments/toxicity , Peptide Fragments/pharmacology , Cell Survival/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Protein Aggregates/drug effects , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism
2.
Int J Mol Sci ; 25(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38731881

ABSTRACT

Aging and age-related diseases are associated with a decline in the capacity of protein turnover. Intrinsically disordered proteins, as well as proteins misfolded and oxidatively damaged, prone to aggregation, are preferentially digested by the ubiquitin-independent proteasome system (UIPS), a major component of which is the 20S proteasome. Therefore, boosting 20S activity constitutes a promising strategy to counteract a decrease in total proteasome activity during aging. One way to enhance the proteolytic removal of unwanted proteins appears to be the use of peptide-based activators of the 20S. In this study, we synthesized a series of peptides and peptidomimetics based on the C-terminus of the Rpt5 subunit of the 19S regulatory particle. Some of them efficiently stimulated human 20S proteasome activity. The attachment of the cell-penetrating peptide TAT allowed them to penetrate the cell membrane and stimulate proteasome activity in HEK293T cells, which was demonstrated using a cell-permeable substrate of the proteasome, TAS3. Furthermore, the best activator enhanced the degradation of aggregation-prone α-synuclein and Tau-441. The obtained compounds may therefore have the potential to compensate for the unbalanced proteostasis found in aging and age-related diseases.


Subject(s)
Aging , Proteasome Endopeptidase Complex , Humans , Proteasome Endopeptidase Complex/metabolism , HEK293 Cells , Aging/metabolism , Protein Aggregates/drug effects , Proteolysis/drug effects , alpha-Synuclein/metabolism , Peptides/pharmacology , Peptides/chemistry , Peptides/metabolism , tau Proteins/metabolism , Protein Aggregation, Pathological/metabolism , Peptidomimetics/pharmacology , Peptidomimetics/chemistry
3.
Colloids Surf B Biointerfaces ; 238: 113928, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38692175

ABSTRACT

In this research, four water-insoluble flavonoid compounds were utilized and reacted with arginine to prepare four carbonized polymer dots with good water-solubility in a hydrothermal reactor. Structural characterization demonstrated that the prepared carbonized polymer dots were classic core-shell structure. Effect of the prepared carbonized polymer dots on protein amyloid aggregation was further investigated using hen egg white lysozyme and human lysozyme as model protein in aqueous solution. All of the prepared carbonized polymer dots could retard the amyloid aggregation of hen egg white lysozyme and human lysozyme in a dose-depended manner. All measurements displayed that the inhibition ratio of luteolin-derived carbonized polymer dots (CPDs-1) was higher than that of the other three carbonized polymer dots under the same dosage. This result may be interpreted by the highest content of phenolic hydroxyl groups on the periphery. The inhibition ratio of CPDs-1 on hen egg white lysozyme and human lysozyme reached 88 % and 83 % at the concentration of 0.5 mg/mL, respectively. CPDs-1 also could disaggregate the formed mature amyloid fibrils into short aggregates.


Subject(s)
Amyloid , Flavonoids , Muramidase , Polymers , Protein Aggregates , Muramidase/chemistry , Muramidase/metabolism , Humans , Polymers/chemistry , Polymers/pharmacology , Amyloid/chemistry , Amyloid/antagonists & inhibitors , Flavonoids/chemistry , Flavonoids/pharmacology , Protein Aggregates/drug effects , Animals , Chickens , Carbon/chemistry
4.
Int J Nanomedicine ; 19: 4299-4317, 2024.
Article in English | MEDLINE | ID: mdl-38766654

ABSTRACT

Background: Inhibition of amyloid ß protein fragment (Aß) aggregation is considered to be one of the most effective strategies for the treatment of Alzheimer's disease. (-)-Epigallocatechin-3-gallate (EGCG) has been found to be effective in this regard; however, owing to its low bioavailability, nanodelivery is recommended for practical applications. Compared to chemical reduction methods, biosynthesis avoids possible biotoxicity and cumbersome preparation processes. Materials and Methods: The interaction between EGCG and Aß42 was simulated by molecular docking, and green tea-conjugated gold nanoparticles (GT-Au NPs) and EGCG-Au NPs were synthesized using EGCG-enriched green tea and EGCG solutions, respectively. Surface active molecules of the particles were identified and analyzed using various liquid chromatography-tandem triple quadrupole mass spectrometry methods. ThT fluorescence assay, circular dichroism, and TEM were used to investigate the effect of synthesized particles on the inhibition of Aß42 aggregation. Results: EGCG as well as apigenin, quercetin, baicalin, and glutathione were identified as capping ligands stabilized on the surface of GT-Au NPs. They more or less inhibited Aß42 aggregation or promoted fibril disaggregation, with EGCG being the most effective, which bound to Aß42 through hydrogen bonding, hydrophobic interactions, etc. resulting in 39.86% and 88.50% inhibition of aggregation and disaggregation effects, respectively. EGCG-Au NPs were not as effective as free EGCG, whereas multiple thiols and polyphenols in green tea accelerated and optimized heavy metal detoxification. The synthesized GT-Au NPs conferred the efficacy of diverse ligands to the particles, with inhibition of aggregation and disaggregation effects of 54.69% and 88.75%, respectively, while increasing the yield, enhancing water solubility, and decreasing cost. Conclusion: Biosynthesis of nanoparticles using green tea is a promising simple and economical drug-carrying approach to confer multiple pharmacophore molecules to Au NPs. This could be used to design new drug candidates to treat Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides , Catechin , Gold , Metal Nanoparticles , Molecular Docking Simulation , Peptide Fragments , Tea , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Catechin/chemistry , Catechin/pharmacology , Catechin/analogs & derivatives , Tea/chemistry , Metal Nanoparticles/chemistry , Metal Nanoparticles/administration & dosage , Gold/chemistry , Ligands , Peptide Fragments/chemistry , Peptide Fragments/antagonists & inhibitors , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Protein Aggregates/drug effects
5.
Nat Commun ; 15(1): 3835, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714700

ABSTRACT

Aggregated forms of α-synuclein constitute the major component of Lewy bodies, the proteinaceous aggregates characteristic of Parkinson's disease. Emerging evidence suggests that α-synuclein aggregation may occur within liquid condensates formed through phase separation. This mechanism of aggregation creates new challenges and opportunities for drug discovery for Parkinson's disease, which is otherwise still incurable. Here we show that the condensation-driven aggregation pathway of α-synuclein can be inhibited using small molecules. We report that the aminosterol claramine stabilizes α-synuclein condensates and inhibits α-synuclein aggregation within the condensates both in vitro and in a Caenorhabditis elegans model of Parkinson's disease. By using a chemical kinetics approach, we show that the mechanism of action of claramine is to inhibit primary nucleation within the condensates. These results illustrate a possible therapeutic route based on the inhibition of protein aggregation within condensates, a phenomenon likely to be relevant in other neurodegenerative disorders.


Subject(s)
Caenorhabditis elegans , Parkinson Disease , Protein Aggregates , alpha-Synuclein , alpha-Synuclein/metabolism , alpha-Synuclein/chemistry , Caenorhabditis elegans/metabolism , Animals , Parkinson Disease/metabolism , Parkinson Disease/drug therapy , Humans , Protein Aggregates/drug effects , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/drug therapy , Disease Models, Animal , Lewy Bodies/metabolism , Kinetics
6.
Int J Biol Macromol ; 267(Pt 2): 131423, 2024 May.
Article in English | MEDLINE | ID: mdl-38583832

ABSTRACT

This article reveals the binding mechanism between glycyrrhizic acid (GA) and α-synuclein to may provide further information for the modulation of synucleinopathies using bioactive compounds. Therefore, the inhibitory activities of GA against α-synuclein aggregation and induced neurotoxicity were evaluated using different assays. Results showed that α-synuclein-GA binding was mediated by intermolecular hydrogen bonds leading to the formation of a slightly folded complex. Theoretical studies revealed that GA binds to the N-terminal domain of α-synuclein and triggers a compact structure around a major part of the N-terminal and the NAC regions along with fluctuations in the C-terminal domain, which are prerequisites for the inhibition of α-synuclein aggregation. Then, the cellular assays showed that GA as a potential small molecule can inhibit the oligomerization of α-synuclein and relevant neurotoxicity through modulation of neural viability, membrane leakage, and ROS formation in a concentration-dependent manner. As a result, the primary mechanism of GA's anti-aggregation and neuroprotective activities is the reorganized α-synuclein structure and fluctuating C-terminal domain, which promotes long-range transient intramolecular contacts between the N-terminal and the C-terminal domain.


Subject(s)
Glycyrrhizic Acid , Protein Aggregates , Synucleinopathies , alpha-Synuclein , Humans , alpha-Synuclein/metabolism , alpha-Synuclein/chemistry , Cell Survival/drug effects , Glycyrrhizic Acid/pharmacology , Glycyrrhizic Acid/chemistry , Hydrogen Bonding , Molecular Docking Simulation , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Protein Aggregates/drug effects , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/drug therapy , Protein Binding , Reactive Oxygen Species/metabolism , Synucleinopathies/metabolism , Synucleinopathies/pathology
7.
Biophys Chem ; 310: 107236, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38615538

ABSTRACT

A novel inhibitor, carboxyphenylboronic acid-modified chitosan oligosaccharide (COS-CPBA), was developed by coupling carboxyphenylboronic acid (CPBA) with chitosan oligosaccharide (COS) to inhibit insulin fibrillation. Extensive biophysical assays indicated that COS-CPBA could decelerate insulin aggregation, hinder the conformational transition from α-helix to ß-sheet structure, change the morphology of insulin aggregates and alter fibrillation pathway. A mechanism for the inhibition of insulin fibrillation by COS-CPBA was proposed. It considers that insulin molecules bind to COS-CPBA via hydrophobic interactions, while the positively charged groups in COS-CPBA exert electrostatic repulsion on the bound insulin molecules. These two opposite forces cause the insulin molecules to display extended conformations and hinder the conformational transition of insulin from α-helix to ß-sheet structure necessary for fibrillation, thus decelerating aggregation and altering the fibrillation pathway of insulin. The studies provide novel ideas for the development of more effective inhibitors of amyloid fibrillation.


Subject(s)
Boronic Acids , Chitosan , Hydrophobic and Hydrophilic Interactions , Insulin , Oligosaccharides , Static Electricity , Insulin/chemistry , Insulin/metabolism , Chitosan/chemistry , Boronic Acids/chemistry , Oligosaccharides/chemistry , Oligosaccharides/pharmacology , Protein Aggregates/drug effects , Animals , Humans
8.
Bioorg Chem ; 147: 107373, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38653149

ABSTRACT

The escalating prevalence of Alzheimer's disease (AD) has prompted extensive research into potential therapeutic interventions, with a specific focus on molecular targets such as amyloid beta (Aß) and tau protein aggregation. In this study, a series of α-ketoamide derivatives was synthesized from ß,γ-unsaturated α-keto thioesters, achieving high purity and good yield. Thioflavin T based Aß aggregation assay identified four promising compounds (BD19, BD23, BD24, and BD27) that demonstrated significant inhibitory effects on Aß aggregation. BD23, selected for its better solubility (0.045 ± 0.0012 mg/ml), was further subjected to in vitro Parallel Artificial Membrane Permeability Assay to determine the Blood-Brain-Barrier permeability and emerged as BBB permeable with permeability rate (Pe) of 10.66 ± 8.11 × 10-6 cm/s. In addition to its Aß inhibitory properties, BD23 exhibited significant inhibition of heparin-induced tau aggregation and demonstrated non-toxicity in SHSY5Y cell lines. Subsequent in vivo assays were conducted, administering compound BD23 to an Aß induced mouse model of AD at various doses (1, 2, & 5 mg/kg). The results revealed a noteworthy enhancement in cognitive functions, particularly when BD23 was administered at a dosage of 5 mg/kg, comparable to the effects observed with the standard dose of Donepezil (DNP). In silico investigations, including molecular docking, molecular dynamics simulations, and Density Functional Theory calculations provided insights into BD23's interactions with the targets and electronic properties. These analyses contribute to the understanding of the therapeutic potential of the lead compounds BD23 which further pave the way for further exploration of its therapeutic potential in the context of AD.


Subject(s)
Alzheimer Disease , Amides , Amyloid beta-Peptides , Dose-Response Relationship, Drug , Protein Aggregates , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Humans , Animals , Mice , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amides/chemistry , Amides/pharmacology , Amides/chemical synthesis , Protein Aggregates/drug effects , Structure-Activity Relationship , Molecular Structure , Molecular Docking Simulation , tau Proteins/metabolism , tau Proteins/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Male
9.
Chem Biol Drug Des ; 103(4): e14529, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38670598

ABSTRACT

With the increasing aging population, rational design of drugs for Alzheimer's disease (AD) treatment has become an important research area. Based on the multifunctional design strategy, four diosmetin derivatives (1-4) were designed, synthesized, and characterized by 1H NMR, 13C NMR, and MS. Docking study was firstly applied to substantiate the design strategies and then the biological activities including cholinesterase inhibition, metal chelation, antioxidation and ß-amyloid (Aß) aggregation inhibition in vitro were evaluated. The results showed that 1-4 had good acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) inhibition, metal chelation (selective chelation of Cu2+ ions), antioxidation, self-induced, Cu2+-induced, and AChE-induced Aß aggregation inhibition activities, and suitable blood-brain barrier (BBB) permeability. Especially, compound 3 had the strongest inhibitory effect on AChE (10-8 M magnitude) and BuChE (10-7 M magnitude) and showed the best inhibition on AChE-induced Aß aggregation with 66.14% inhibition ratio. Furthermore, compound 3 could also reduce intracellular reactive oxygen species (ROS) levels in Caenorhabditis elegans and had lower cytotoxicity. In summary, 3 might be considered as a potential multifunctional anti-AD ligand.


Subject(s)
Acetylcholinesterase , Alzheimer Disease , Amyloid beta-Peptides , Blood-Brain Barrier , Butyrylcholinesterase , Caenorhabditis elegans , Cholinesterase Inhibitors , Drug Design , Flavonoids , Molecular Docking Simulation , Reactive Oxygen Species , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/metabolism , Acetylcholinesterase/metabolism , Animals , Butyrylcholinesterase/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/metabolism , Ligands , Blood-Brain Barrier/metabolism , Humans , Reactive Oxygen Species/metabolism , Flavonoids/chemistry , Flavonoids/pharmacology , Flavonoids/chemical synthesis , Antioxidants/pharmacology , Antioxidants/chemistry , Antioxidants/chemical synthesis , Structure-Activity Relationship , Protein Aggregates/drug effects
10.
ACS Chem Neurosci ; 15(9): 1915-1925, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38634811

ABSTRACT

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.


Subject(s)
Amyloid , Calgranulin A , Calgranulin B , Leukocyte L1 Antigen Complex , Calgranulin B/metabolism , Calgranulin A/metabolism , Leukocyte L1 Antigen Complex/metabolism , Amyloid/metabolism , Humans , Protein Aggregates/physiology , Protein Aggregates/drug effects , Calcium/metabolism , Protein Structure, Secondary
11.
Nat Commun ; 15(1): 3658, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38688913

ABSTRACT

Abberent protein-protein interactions potentiate many diseases and one example is the toxic, self-assembly of α-Synuclein in the dopaminergic neurons of patients with Parkinson's disease; therefore, a potential therapeutic strategy is the small molecule modulation of α-Synuclein aggregation. In this work, we develop an Oligopyridylamide based 2-dimensional Fragment-Assisted Structure-based Technique to identify antagonists of α-Synuclein aggregation. The technique utilizes a fragment-based screening of an extensive array of non-proteinogenic side chains in Oligopyridylamides, leading to the identification of NS132 as an antagonist of the multiple facets of α-Synuclein aggregation. We further identify a more cell permeable analog (NS163) without sacrificing activity. Oligopyridylamides rescue α-Synuclein aggregation mediated Parkinson's disease phenotypes in dopaminergic neurons in early and post disease Caenorhabditis elegans models. We forsee tremendous potential in our technique to identify lead therapeutics for Parkinson's disease and other diseases as it is expandable to other oligoamide scaffolds and a larger array of side chains.


Subject(s)
Caenorhabditis elegans , Dopaminergic Neurons , Parkinson Disease , alpha-Synuclein , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Caenorhabditis elegans/metabolism , Parkinson Disease/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/pathology , Animals , Humans , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Phenotype , Protein Aggregates/drug effects , Disease Models, Animal , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/drug therapy , Pyridines/pharmacology , Pyridines/chemistry , Amides/pharmacology , Amides/chemistry
12.
Bioorg Med Chem Lett ; 105: 129752, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38631541

ABSTRACT

The misfolding and aggregation of α-Syn play a pivotal role in connecting diverse pathological pathways in Parkinson's disease (PD). Preserving α-Syn proteostasis and functionality by inhibiting its aggregation or disaggregating existing aggregates using suitable inhibitors represents a promising strategy for PD prevention and treatment. In this study, a series of benzothiazole-polyphenol hybrids was designed and synthesized. Three identified compounds exhibited notable inhibitory activities against α-Syn aggregation in vitro, with IC50 values in the low micromolar range. These inhibitors demonstrated sustained inhibitory effects throughout the entire aggregation process, stabilizing α-Syn proteostasis conformation. Moreover, the compounds effectively disintegrated preformed α-Syn oligomers and fibers, potentially by binding to specific domains within the fibers, inducing fibril instability, collapse, and ultimately resulting in smaller-sized aggregates and monomers. These findings offer valuable insights into the therapeutic potential of polyphenol hybrids with 2-conjugated benzothiazole targeting α-Syn aggregation in the treatment of PD.


Subject(s)
Benzothiazoles , Polyphenols , Protein Aggregates , alpha-Synuclein , Benzothiazoles/chemistry , Benzothiazoles/pharmacology , Benzothiazoles/chemical synthesis , alpha-Synuclein/antagonists & inhibitors , alpha-Synuclein/metabolism , Polyphenols/chemistry , Polyphenols/pharmacology , Polyphenols/chemical synthesis , Humans , Protein Aggregates/drug effects , Molecular Structure , Structure-Activity Relationship , Dose-Response Relationship, Drug , Parkinson Disease/drug therapy , Parkinson Disease/metabolism
13.
Biochemistry ; 63(9): 1162-1169, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38668883

ABSTRACT

Parkinson's disease (PD) is characterized by the toxic oligomeric and fibrillar phases formed by monomeric alpha-synuclein (α-syn). Certain nanoparticles have been demonstrated to promote protein aggregation, while other nanomaterials have been found to prevent the process. In the current work, we use nuclear magnetic resonance spectroscopy in conjunction with isothermal titration calorimetry to investigate the cause and mechanism of these opposing effects at the amino acid protein level. The interaction of α-syn with two types of nanomaterials was considered: citrate-capped gold nanoparticles (AuNPs) and graphene oxide (GO). In the presence of AuNPs, α-syn aggregation is accelerated, whereas in the presence of GO, aggregation is prevented. The study indicates that GO sequesters the NAC region of α-syn monomers through electrostatic and hydrophobic interactions, leading to a reduced elongation rate, and AuNPs leave the NAC region exposed while binding the N-terminus, leading to higher aggregation. The protein's inclination toward quicker aggregation is explained by the binding of the N-terminus of α-syn with the gold nanoparticles. Conversely, a comparatively stronger interaction with GO causes the nucleation and growth phases to be postponed and inhibits intermolecular interactions. Our finding offers novel experimental insights at the residue level regarding the aggregation of α-syn in the presence of various nanomaterials and creates new opportunities for the development of suitably functionalized nanomaterial-based therapeutic reagents against Parkinson's and other neurodegenerative diseases.


Subject(s)
Gold , Graphite , Metal Nanoparticles , Protein Aggregates , alpha-Synuclein , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , Gold/chemistry , Metal Nanoparticles/chemistry , Graphite/chemistry , Humans , Protein Aggregates/drug effects , Parkinson Disease/metabolism , Nanostructures/chemistry , Citric Acid/chemistry , Citric Acid/metabolism , Hydrophobic and Hydrophilic Interactions
14.
J Phys Chem B ; 128(18): 4283-4300, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38683125

ABSTRACT

Kidney-associated human lysozyme amyloidosis leads to renal impairments;thus, patients are often prescribed furosemide. Based on this fact, the effect of furosemide on induced human lysozyme fibrillation, in vitro, is evaluated by spectroscopic, calorimetric, computational, and cellular-based assays/methods. Results show that furosemide increases the lag phase and decreases the apparent rate of aggregation of human lysozyme, thereby decelerating the nucleation phase and amyloid fibril formation, as confirmed by the decrease in the level of Thioflavin-T fluorescence. Fewer entities of hydrodynamic radii of ∼171 nm instead of amyloid fibrils (∼412 nm) are detected in human lysozyme in the presence of furosemide by dynamic light scattering. Moreover, furosemide decreases the extent of conversion of the α/ß structure of human lysozyme into a predominant ß-sheet. The isothermal titration calorimetry established that furosemide forms a complex with human lysozyme, which was also confirmed through fluorescence quenching and computational studies. Also, human lysozyme lytic activity is inhibited competitively by furosemide due to the involvement of amino acid residues of the active site in catalysis, as well as complex formation. Conclusively, furosemide interacts with Gln58, Ile59, Asn60, Ala108, and Trp109 of aggregation-prone regions 2 and 4 of human lysozyme, thereby masking its sites of aggregation and generating only lower-order entities that are less toxic to red blood cells than the fibrils. Thus, furosemide slows the progression of amyloid fibrillation in human lysozyme.


Subject(s)
Furosemide , Muramidase , Muramidase/chemistry , Muramidase/metabolism , Humans , Furosemide/chemistry , Furosemide/pharmacology , Protein Aggregates/drug effects , Amyloid/metabolism , Amyloid/chemistry , Molecular Dynamics Simulation
15.
Nanoscale ; 16(16): 8074-8089, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38563405

ABSTRACT

Amyloid aggregation is implicated in the pathogenesis of various neurodegenerative disorders, such as Alzheimer's disease (AD) and Parkinson's disease (PD). It is critical to develop high-performance drugs to combat amyloid-related diseases. Most identified nanomaterials exhibit limited biocompatibility and therapeutic efficacy. In this work, we used a solvent-free carbonization process to prepare new photo-responsive carbon nanodots (CNDs). The surface of the CNDs is densely packed with chemical groups. CNDs with large, conjugated domains can interact with proteins through π-π stacking and hydrophobic interactions. Furthermore, CNDs possess the ability to generate singlet oxygen species (1O2) and can be used to oxidize amyloid. The hydrophobic interaction and photo-oxidation can both influence amyloid aggregation and disaggregation. Thioflavin T (ThT) fluorescence analysis and circular dichroism (CD) spectroscopy indicate that CNDs can block the transition of amyloid from an α-helix structure to a ß-sheet structure. CNDs demonstrate efficacy in alleviating cytotoxicity induced by Aß42 and exhibit promising blood-brain barrier (BBB) permeability. CNDs have small size, low biotoxicity, good fluorescence and photocatalytic properties, and provide new ideas for the diagnosis and treatment of amyloid-related diseases.


Subject(s)
Amyloid beta-Peptides , Carbon , Carbon/chemistry , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/chemistry , Humans , Catalysis , Singlet Oxygen/metabolism , Singlet Oxygen/chemistry , Blood-Brain Barrier/metabolism , Animals , Protein Aggregates/drug effects , Quantum Dots/chemistry , Amyloid/chemistry , Amyloid/metabolism , Cell Survival/drug effects , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Hydrophobic and Hydrophilic Interactions
16.
J Phys Chem Lett ; 15(17): 4761-4766, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38661515

ABSTRACT

Heart tissue can experience a progressive accumulation of transthyretin (TTR), a small four subunit protein that transports holoretinol binding protein and thyroxine. This severe pathology is known as transthyretin amyloid cardiomyopathy. Numerous experimental studies indicated that the aggregation rate and toxicity of TTR fibrils could be altered by the presence of lipids; however, the role of plasmalogens in this process remains unknown. In this study, we investigate the effect of choline plasmalogens (CPs) with different lengths and saturations of fatty acids (FAs) on TTR aggregation. We found that CPs with saturated and unsaturated FAs strongly suppressed TTR aggregation. We also found that CPs with saturated FAs did not change the morphology of TTR fibrils; however, much thicker fibrillar species were formed in the presence of CPs with unsaturated FAs. Finally, we found that CPs with C16:0, C18:0, and C18:1 FAs substantially lowered the cytotoxicity of TTR fibrils that were formed in their presence.


Subject(s)
Plasmalogens , Prealbumin , Prealbumin/chemistry , Prealbumin/metabolism , Plasmalogens/metabolism , Plasmalogens/chemistry , Humans , Amyloid/chemistry , Amyloid/metabolism , Protein Aggregates/drug effects , Fatty Acids/chemistry , Fatty Acids/metabolism
17.
Nat Chem Biol ; 20(5): 634-645, 2024 May.
Article in English | MEDLINE | ID: mdl-38632492

ABSTRACT

Machine learning methods hold the promise to reduce the costs and the failure rates of conventional drug discovery pipelines. This issue is especially pressing for neurodegenerative diseases, where the development of disease-modifying drugs has been particularly challenging. To address this problem, we describe here a machine learning approach to identify small molecule inhibitors of α-synuclein aggregation, a process implicated in Parkinson's disease and other synucleinopathies. Because the proliferation of α-synuclein aggregates takes place through autocatalytic secondary nucleation, we aim to identify compounds that bind the catalytic sites on the surface of the aggregates. To achieve this goal, we use structure-based machine learning in an iterative manner to first identify and then progressively optimize secondary nucleation inhibitors. Our results demonstrate that this approach leads to the facile identification of compounds two orders of magnitude more potent than previously reported ones.


Subject(s)
Drug Discovery , Machine Learning , Protein Aggregates , alpha-Synuclein , alpha-Synuclein/antagonists & inhibitors , alpha-Synuclein/metabolism , alpha-Synuclein/chemistry , Humans , Drug Discovery/methods , Protein Aggregates/drug effects , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemistry , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Structure-Activity Relationship
18.
ACS Chem Neurosci ; 15(9): 1770-1786, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38637513

ABSTRACT

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.


Subject(s)
Masoprocol , gamma-Synuclein , Humans , gamma-Synuclein/metabolism , Masoprocol/pharmacology , Protein Aggregates/drug effects , Protein Aggregates/physiology , Spectroscopy, Fourier Transform Infrared , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/drug therapy
19.
Food Funct ; 15(8): 4233-4245, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38517352

ABSTRACT

The aggregation of ß-amyloid (Aß) peptides to form amyloid plaques is one of the primary hallmarks for Alzheimer's disease (AD). Dietary flavonoid supplements containing hesperetin have an ability to decline the risk of developing AD, but the molecular mechanism is still unclear. In this work, hesperetin, a flavanone abundant in citrus fruits, has been proven to prevent the formation of Aß aggregates and depolymerized preformed fibrils in a concentration-dependent fashion. Hesperetin inhibited the conformational conversion from the natural structure to a ß-sheet-rich conformation. It was found that hesperetin significantly reduced the cytotoxicity and relieved oxidative stress eventuated by Aß aggregates in a concentration-dependent manner. Additionally, the beneficial effects of hesperetin were confirmed in Caenorhabditis elegans, including the inhibition of the formation and deposition of Aß aggregates and extension of their lifespan. Finally, the results of molecular dynamics simulations showed that hesperetin directly interacted with an Aß42 pentamer mainly through strong non-polar and electrostatic interactions, which destroyed the structural stability of the preformed pentamer. To summarize, hesperetin exhibits great potential as a prospective dietary supplement for preventing and improving AD.


Subject(s)
Amyloid beta-Peptides , Caenorhabditis elegans , Hesperidin , Hesperidin/pharmacology , Hesperidin/chemistry , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Amyloid beta-Peptides/chemistry , Animals , Caenorhabditis elegans/drug effects , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/prevention & control , Amyloid/metabolism , Molecular Dynamics Simulation , Oxidative Stress/drug effects , Protein Aggregates/drug effects
20.
Chem Biol Interact ; 394: 110978, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38552766

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by the accumulation of amyloid-ß (Aß) protein aggregates, leading to synaptic dysfunction and neuronal cell death. In this study, we used a comprehensive approach encompassing in vitro assays, computational analyses, and an in vivo Caenorhabditis elegans model to evaluate the inhibitory effects of various xanthones, focusing on Garcinone D (GD), on Aß42 oligomer formation. Dot blot analysis revealed concentration-dependent responses among xanthones, with GD consistently inhibiting Aß42 oligomer formation at low concentrations (0.1 and 0.5 µM, inhibitions of 84.66 ± 2.25% and 85.06 ± 6.57%, respectively). Molecular docking and dynamics simulations provided insights into the molecular interactions between xanthones and Aß42, highlighting the disruption of key residues involved in Aß42 aggregation. The neuroprotective potential of GD was established using transgenic C. elegans GMC101, with substantial delays in paralysis reported at higher concentrations. Our findings show that GD is a potent suppressor of Aß42 oligomer formation, suggesting its potential as a therapeutic candidate for AD. The concentration-dependent effects observed in both in vitro and in vivo models underscore the need for nuanced dose-response assessments. These findings contribute novel insights into the therapeutic landscape of xanthones against AD, emphasizing the multifaceted potential of GD for further translational endeavors in neurodegenerative disorder research.


Subject(s)
Amyloid beta-Peptides , Animals, Genetically Modified , Caenorhabditis elegans , Molecular Docking Simulation , Peptide Fragments , Xanthones , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Xanthones/pharmacology , Xanthones/chemistry , Peptide Fragments/toxicity , Peptide Fragments/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Protein Aggregates/drug effects , Molecular Dynamics Simulation , Disease Models, Animal , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...