Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (149)

Search Parameters:
Keywords = protein fibrillation inhibition

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
27 pages, 3139 KiB  
Article
Distinctive Effects of Fullerene C60 and Fullerenol C60(OH)24 Nanoparticles on Histological, Molecular and Behavioral Hallmarks of Alzheimer’s Disease in APPswe/PS1E9 Mice
by Sholpan Askarova, Kseniia Sitdikova, Aliya Kassenova, Kirill Chaprov, Evgeniy Svirin, Andrey Tsoy, Johannes de Munter, Anna Gorlova, Aleksandr Litavrin, Aleksei Deikin, Andrey Nedorubov, Nurbol Appazov, Allan Kalueff, Anton Chernopiatko and Tatyana Strekalova
Antioxidants 2025, 14(7), 834; https://doi.org/10.3390/antiox14070834 - 8 Jul 2025
Viewed by 655
Abstract
Fullerenes and fullerenols exhibit antioxidant and anti-inflammatory properties, making them promising candidates for Alzheimer’s disease (AD) therapy. Unlike conventional anti-inflammatory drugs, these compounds have multitargeted effects, including their ability to inhibit amyloid fibril formation. However, few studies have explored their efficacy in high-validity [...] Read more.
Fullerenes and fullerenols exhibit antioxidant and anti-inflammatory properties, making them promising candidates for Alzheimer’s disease (AD) therapy. Unlike conventional anti-inflammatory drugs, these compounds have multitargeted effects, including their ability to inhibit amyloid fibril formation. However, few studies have explored their efficacy in high-validity AD models. Female APPswe/PS1E9 (APP/PS1) mice and their wild-type (WT) littermates were orally administered with fullerene C60 (0.1 mg/kg/day) or fullerenol C60(OH)24 (0.15 mg/kg/day) for 10 months starting at 2 months of age. Behavioral assessments were performed at 12 months of age. Amyloid plaque density and size were analyzed in the brain regions using Congo red staining. The expression of genes related to inflammation and plasticity was examined, and an in vitro assay was used to test the toxicity of fullerenol and its effect on amyloid β peptide 42 (Aβ42)-induced reactive oxygen species (ROS) production. Fullerenol reduced the maximum plaque size in the cortex and hippocampus, decreased the small plaque density in the hippocampus and thalamus, and prevented an increase in glial fibrillary acidic protein (GFAP) positive cell density in the mutants. Both treatments improved cognitive and emotional behaviors and reduced Il1β and increased Sirt1 expression. In vitro, fullerenol was non-toxic across a range of concentrations and reduced Aβ42-induced ROS production in brain endothelial cells and astrocytes. Long-term administration of fullerene or fullerenol improved behavioral and molecular markers of AD in APP/PS1 mice, with fullerenol showing additional benefits in reducing amyloid burden. Full article
(This article belongs to the Section Natural and Synthetic Antioxidants)
Show Figures

Figure 1

11 pages, 2361 KiB  
Communication
Inhibitory Effects of 3-(4-Hydroxy-3-methoxyphenyl) Propionic Acid on Amyloid β-Peptide Aggregation In Vitro
by Makoto Mori, Hiroto Nakano, Sadao Hikishima, Jota Minamikawa, Daiki Muramatsu, Yasuhiro Sakashita, Tokuhei Ikeda, Moeko Noguchi-Shinohara and Kenjiro Ono
Biomedicines 2025, 13(7), 1649; https://doi.org/10.3390/biomedicines13071649 - 6 Jul 2025
Viewed by 425
Abstract
Objectives: The compound 3-(4-Hydroxy-3-methoxyphenyl) propionic acid (HMPA) is a terminal metabolite derived from polyphenol compounds. It has been studied for its potential to support brain health indirectly through its anti-oxidant effects and ability to enhance the gut environment; however, its role in [...] Read more.
Objectives: The compound 3-(4-Hydroxy-3-methoxyphenyl) propionic acid (HMPA) is a terminal metabolite derived from polyphenol compounds. It has been studied for its potential to support brain health indirectly through its anti-oxidant effects and ability to enhance the gut environment; however, its role in dementia pathogenesis is unclear. Therefore, the aim of this study was to evaluate how HMPA inhibits Aβ42 aggregation in vitro. Methods: We examined the inhibitory effects of HMPA on amyloid-β protein (Aβ) aggregation using a thioflavin T (ThT) assay and electron microscopy (EM). Results: ThT assays demonstrated that HMPA inhibited both the nucleation and elongation phases of Aβ aggregation. Additionally, EM of low-molecular-weight (LMW) Aβ42 in the presence of HMPA demonstrated shorter fibrils compared to those formed without HMPA. The EC50 of HMPA in LMW Aβ42 was 5–6 mM. Conclusions: These findings indicate that, similar to several polyphenol compounds such as myricetin and rosmarinic acid, HMPA may inhibit Aβ pathogenesis, although it requires a fairly high concentration in vitro. These findings suggest the potential of HMPA as a lead compound for modulating Aβ-related neurodegeneration. Full article
(This article belongs to the Section Neurobiology and Clinical Neuroscience)
Show Figures

Figure 1

20 pages, 1609 KiB  
Review
Natural Products Acting as Senolytics and Senomorphics Alleviate Cardiovascular Diseases by Targeting Senescent Cells
by Hejing Tang, Xu Zhang, Senyang Hu, Yuhan Song, Wenhua Jin, Jianmin Zou, Yan Zhang, Jiayue Guo, Peng An, Junjie Luo, Pengjie Wang, Yongting Luo and Yinhua Zhu
Targets 2025, 3(3), 23; https://doi.org/10.3390/targets3030023 - 25 Jun 2025
Viewed by 902
Abstract
Taken together, cardiovascular diseases (CVDs) have become one of the prime causes of the global disease burden. Aging is closely related to CVDs and is considered to be one of the crucial factors in the incidence of CVDs. In the process of aging, [...] Read more.
Taken together, cardiovascular diseases (CVDs) have become one of the prime causes of the global disease burden. Aging is closely related to CVDs and is considered to be one of the crucial factors in the incidence of CVDs. In the process of aging, cellular senescence is an important cause of CVDs such as atherosclerosis and atrial fibrillation. The treatment for CVDs by targeting senescent cells has been carried out in cellular models, animal experiments, and anti-aging clinical trials. Chemical approaches to regulate the fate of senescent cells by senolytics and senomorphics, which could selectively eliminate senescent cells or inhibit their senescence-associated secretory phenotype (SASP) secretion, have been increasingly explored. Importantly, many natural products with promising biological activity extracted from food or medicine–food homology have the above-mentioned effects. Furthermore, the identification of the target cells or target proteins of these natural products is of great significance for the indication of their mechanism of action, and it also lays a scientific foundation for the realization of precision nutrition intervention in the future. This review details how senescent cells affect CVDs, how natural products target senescent cells through nutritional intervention, and research methods for natural products in cardiovascular aging. Full article
Show Figures

Figure 1

14 pages, 1413 KiB  
Review
Advances in the Exploration of Coordination Complexes of Vanadium in the Realm of Alzheimer’s Disease: A Mini Review
by Jesús Antonio Cruz-Navarro, Luis Humberto Delgado-Rangel, Ricardo Malpica-Calderón, Arturo T. Sánchez-Mora, Hugo Ponce-Bolaños, Andrés Felipe González-Oñate, Jorge Alí-Torres, Raúl Colorado-Peralta, Daniel Canseco-Gonzalez, Viviana Reyes-Márquez and David Morales-Morales
Molecules 2025, 30(12), 2547; https://doi.org/10.3390/molecules30122547 - 11 Jun 2025
Viewed by 594
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory loss and limited therapeutic options. Metal-based drugs have emerged as promising alternatives in the search for effective treatments, and vanadium coordination complexes have shown significant potential due to their neuroprotective [...] Read more.
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory loss and limited therapeutic options. Metal-based drugs have emerged as promising alternatives in the search for effective treatments, and vanadium coordination complexes have shown significant potential due to their neuroprotective and anti-aggregant properties. This review explores the advances in the development of vanadium-based metallodrugs for AD, focusing on their ability to modulate amyloid-beta (Aβ) aggregation, oxidative stress, and neuroinflammation. Recent in vitro and in vivo studies highlight the efficacy of oxovanadium (IV) and peroxovanadium (V) complexes in inhibiting Aβ fibril formation and reducing neuronal toxicity. Additionally, the interaction of vanadium complexes with key biological targets, such as peroxisome proliferator-activated receptor gamma (PPARγ) and protein-tyrosine phosphatase 1B (PTP1B), suggests a multifaceted therapeutic approach. While these findings underscore the potential of vanadium compounds as innovative treatments for AD, further research is needed to optimize their bioavailability, selectivity, and safety for clinical applications. Full article
Show Figures

Graphical abstract

26 pages, 1352 KiB  
Article
On the Potential Role of Phytate Against Neurodegeneration: It Protects Against Fe3+-Catalyzed Degradation of Dopamine and Ascorbate and Against Fe3+-Induced Protein Aggregation
by Samantha Rebeca Godoy, Pilar Sanchis, Juan Frau, Bartolomé Vilanova and Miquel Adrover
Int. J. Mol. Sci. 2025, 26(10), 4799; https://doi.org/10.3390/ijms26104799 - 16 May 2025
Cited by 1 | Viewed by 897
Abstract
Myo-inositol-1,2,3,4,5,6-hexakisphosphate (IP6) is commonly found in plant-derived foods and has important pharmacological properties against many pathologies. One of them appears to be neurodegeneration, which is notably stimulated by dysregulated metal metabolism. Consequently, we explore the role of IP6 in mitigating neurodegenerative events catalyzed [...] Read more.
Myo-inositol-1,2,3,4,5,6-hexakisphosphate (IP6) is commonly found in plant-derived foods and has important pharmacological properties against many pathologies. One of them appears to be neurodegeneration, which is notably stimulated by dysregulated metal metabolism. Consequently, we explore the role of IP6 in mitigating neurodegenerative events catalyzed by dysregulated free iron. More precisely, we performed spectrophotometric measurements in aqueous solutions to investigate the ability of IP6 to chelate Fe3+ and inhibit its role in catalyzing the oxidative degradation of dopamine and ascorbic acid, two key molecules in neuronal redox systems. Our results demonstrate that IP6 effectively prevents the formation of harmful intermediates, such as neuromelanin and reactive oxygen species, which are linked to neuronal damage. Additionally, we assessed the effect of IP6 on Fe3+-induced protein aggregation, focusing on α-synuclein, which is closely associated with Parkinson’s disease. Our data reveal that IP6 accelerates the conversion of toxic α-synuclein oligomers into less harmful amyloid fibrils, thereby reducing their neurotoxic potential. Our findings highlight the dual function of IP6 as a potent Fe3+ chelator and modulator of protein aggregation pathways, reinforcing its potential as a neuroprotective agent. Consequently, IP6 offers promising therapeutic potential for mitigating the progression of neurodegenerative disorders such as Parkinson’s and Alzheimer’s diseases. Full article
(This article belongs to the Special Issue Development of Dopaminergic Neurons 3.0)
Show Figures

Figure 1

24 pages, 2232 KiB  
Review
Nanoplatforms Targeting Intrinsically Disordered Protein Aggregation for Translational Neuroscience Applications
by Chih Hung Lo, Lenny Yi Tong Cheong and Jialiu Zeng
Nanomaterials 2025, 15(10), 704; https://doi.org/10.3390/nano15100704 - 8 May 2025
Viewed by 994
Abstract
Intrinsically disordered proteins (IDPs), such as tau, beta-amyloid (Aβ), and alpha-synuclein (αSyn), are prone to misfolding, resulting in pathological aggregation and propagation that drive neurodegenerative diseases, including Alzheimer’s disease (AD), frontotemporal dementia (FTD), and Parkinson’s disease (PD). Misfolded IDPs are prone to aggregate [...] Read more.
Intrinsically disordered proteins (IDPs), such as tau, beta-amyloid (Aβ), and alpha-synuclein (αSyn), are prone to misfolding, resulting in pathological aggregation and propagation that drive neurodegenerative diseases, including Alzheimer’s disease (AD), frontotemporal dementia (FTD), and Parkinson’s disease (PD). Misfolded IDPs are prone to aggregate into oligomers and fibrils, exacerbating disease progression by disrupting cellular functions in the central nervous system, triggering neuroinflammation and neurodegeneration. Furthermore, aggregated IDPs exhibit prion-like behavior, acting as seeds that are released into the extracellular space, taken up by neighboring cells, and have a propagating pathology across different regions of the brain. Conventional inhibitors, such as small molecules, peptides, and antibodies, face challenges in stability and blood–brain barrier penetration, limiting their efficacy. In recent years, nanotechnology-based strategies, such as multifunctional nanoplatforms or nanoparticles, have emerged as promising tools to address these challenges. These nanoplatforms leverage tailored designs to prevent or remodel the aggregation of IDPs and reduce associated neurotoxicity. This review discusses recent advances in nanoplatforms designed to target tau, Aβ, and αSyn aggregation, with a focus on their roles in reducing neuroinflammation and neurodegeneration. We examine critical aspects of nanoplatform design, including the choice of material backbone and targeting moieties, which influence interactions with IDPs. We also highlight key mechanisms including the interaction between nanoplatforms and IDPs to inhibit their aggregation, redirect aggregation cascade towards nontoxic, off-pathway species, and disrupt fibrillar structures into soluble forms. We further outline future directions for enhancing IDP clearance, achieving spatiotemporal control, and improving cell-specific targeting. These nanomedicine strategies offer compelling paths forward for developing more effective and targeted therapies for neurodegenerative diseases. Full article
(This article belongs to the Section Biology and Medicines)
Show Figures

Graphical abstract

17 pages, 2598 KiB  
Article
Molecular Dynamics Simulation Study of Stabilizer Association with the Val122Ile Transthyretin Variant
by Kevin Morris, John DeSalvo, Iman Deanparvar, Lucus Schneider, Kaleigh Leach, Matthew George and Yayin Fang
Biophysica 2025, 5(2), 16; https://doi.org/10.3390/biophysica5020016 - 23 Apr 2025
Cited by 1 | Viewed by 1073
Abstract
The tetrameric protein transthyretin (TTR) transports the hormone thyroxine in plasma and cerebrospinal fluid. Certain point mutations of TTR, including the Val122Ile mutation investigated here, destabilize the tetramer leading to its dissociation, misfolding, aggregation, and the eventual buildup of amyloid fibrils in the [...] Read more.
The tetrameric protein transthyretin (TTR) transports the hormone thyroxine in plasma and cerebrospinal fluid. Certain point mutations of TTR, including the Val122Ile mutation investigated here, destabilize the tetramer leading to its dissociation, misfolding, aggregation, and the eventual buildup of amyloid fibrils in the myocardium. Cioffi et al. reported the design and synthesis of a novel TTR kinetic stabilizing ligand, referred to here as TKS14, that inhibited TTR dissociation and amyloid fibril formation. In this study, molecular dynamics simulations were used to investigate the binding of TKS14 and eight TSK14 derivatives to the Val122Ile TTR mutant. For each complex, the ligand’s solvent accessible surface area (SASA), ligand–receptor hydrogen-bonding interactions, and the free energy of ligand-binding to TTR were investigated. The goal of this study was to identify the TSK14 functional groups that contributed to TTR stabilization. TKS14 was found to form a stable, two-point interaction with TTR by hydrogen bonding to Ser-117 residues in the inner receptor binding pocket and interacting through hydrogen bonds and electrostatically with Lys-15 residues near the receptor’s surface. The free energy of TKS14-TTR binding was −18.0 kcal mol−1 and the ligand’s average SASA value decreased by over 80% upon binding to the receptor. The thermodynamic favorability of TTR binding decreased when TKS14 derivatives contained either methyl ester, amide, tetrazole, or N-methyl functional groups that disrupted the above two-point interaction. One derivative in which a tetrazole ring was added to TKS14 was found to form hydrogen bonds with Thr-106, Thr-119, Ser-117, and Lys-15 residues. This derivative had a free energy of TTR binding of −21.4 kcal mol−1. Overall, the molecular dynamics simulations showed that the functional groups within the TKS14 structural template can be tuned to optimize the thermodynamic favorability of ligand binding. Full article
(This article belongs to the Special Issue Molecular Structure and Simulation in Biological System 3.0)
Show Figures

Figure 1

14 pages, 3899 KiB  
Article
The Inhibition of Bromodomain and Extraterminal Domain (BET) Proteins Protects Against Microglia-Mediated Neuronal Loss In Vitro
by Marta Matuszewska, Anna Wilkaniec, Magdalena Cieślik, Marcin Strawski and Grzegorz A. Czapski
Biomolecules 2025, 15(4), 528; https://doi.org/10.3390/biom15040528 - 4 Apr 2025
Viewed by 709
Abstract
Neuroinflammation is a key feature of all neurodegenerative disorders, including Alzheimer’s disease, and is tightly regulated by epigenetic mechanisms. Among them, bromodomain and extraterminal domain (BET) proteins play a crucial role by recognizing acetylated histones and acting as transcriptional co-regulators to modulate gene [...] Read more.
Neuroinflammation is a key feature of all neurodegenerative disorders, including Alzheimer’s disease, and is tightly regulated by epigenetic mechanisms. Among them, bromodomain and extraterminal domain (BET) proteins play a crucial role by recognizing acetylated histones and acting as transcriptional co-regulators to modulate gene expression. This study investigates the potential of inhibiting BET proteins in preventing microglia-mediated neuronal damage in vitro. Murine BV2 microglial cells were exposed to lipopolysaccharide (LPS) or amyloid-β (Aβ) to induce an inflammatory response, and the subsequent effects on murine HT22 neuronal cells were examined. Among the BET proteins tested, only Brd4 was significantly upregulated in BV2 cells upon pro-inflammatory stimulation. JQ1, a potent pan-inhibitor of BET proteins, suppressed LPS-induced upregulation of pro-inflammatory cytokine mRNA levels, including Il1b, Il6, and Tnf, in BV2 microglia. Pre-treatment with JQ1 attenuated the cytotoxicity of LPS-activated BV2 cells toward neurons. Additionally, conditioned media from Aβ fibril-stimulated BV2 cells induced neuronal cell death, which was partially prevented by pre-treatment with JQ1. Co-culture assays further demonstrated the beneficial effect of BET inhibition. Our findings suggest that targeting BET proteins may offer a neuroprotective strategy by modulating microglial activation, potentially providing therapeutic benefits in neurodegenerative diseases. Full article
(This article belongs to the Special Issue Molecular and Genetic Basis of Neurodegenerative Diseases)
Show Figures

Figure 1

45 pages, 9857 KiB  
Review
Plant-Based Inhibitors of Protein Aggregation
by Olha Zhytniakivska, Tanmay Chaturvedi and Mette Hedegaard Thomsen
Biomolecules 2025, 15(4), 481; https://doi.org/10.3390/biom15040481 - 25 Mar 2025
Cited by 2 | Viewed by 1932
Abstract
The assembly of amyloidogenic proteins and peptides into toxic oligomeric and fibrillar aggregates is closely connected to the onset and progression of more than 50 protein diseases, such as Alzheimer’s disease, Parkinson’s disease, prion disease, and type 2 diabetes, to name only a [...] Read more.
The assembly of amyloidogenic proteins and peptides into toxic oligomeric and fibrillar aggregates is closely connected to the onset and progression of more than 50 protein diseases, such as Alzheimer’s disease, Parkinson’s disease, prion disease, and type 2 diabetes, to name only a few. Considerable research efforts at identifying the therapeutic strategies against these maladies are currently focused on preventing and inhibiting pathogenic protein aggregation by various agents. Plant-based extracts and compounds have emerged as promising sources of potential inhibitors due to their dual role as nutraceuticals as part of healthy diets and as specific pharmaceuticals when administered at higher concentrations. In recent decades, several plant extracts and plant-extracted compounds have shown potential to modulate protein aggregation. An ever-growing body of research on plant-based amyloid inhibitors requires a detail analysis of existing data to identify potential knowledge gaps. This review summarizes the recent progress in amyloid inhibition using 17 flavonoids, 11 polyphenolic non-flavonoid compounds, 23 non-phenolic inhibitors, and 59 plant extracts, with the main emphasis on directly modulating the fibrillation of four amyloid proteins, namely amyloid-β peptide, microtubule-associated protein tau, α-synuclein, and human islet amyloid polypeptide. Full article
Show Figures

Figure 1

19 pages, 8357 KiB  
Article
Binding of Pro-Inflammatory Proteins S100A8 or S100A9 to Amyloid-β Peptide Suppresses Its Fibrillation
by Ekaterina A. Litus, Marina P. Shevelyova, Alisa A. Vologzhannikova, Evgenia I. Deryusheva, Andrey V. Machulin, Ekaterina L. Nemashkalova, Maria E. Permyakova, Andrey S. Sokolov, Valeria D. Alikova, Vladimir N. Uversky and Sergei E. Permyakov
Biomolecules 2025, 15(3), 431; https://doi.org/10.3390/biom15030431 - 17 Mar 2025
Cited by 1 | Viewed by 1030
Abstract
Human serum albumin (HSA) is a natural depot of amyloid-β peptide (Aβ), a key player in Alzheimer’s disease (AD). HSA and pro-inflammatory Ca2+-binding proteins S100A8 and S100A9 are involved in Aβ metabolism and its deposition in the brain, serving as probable [...] Read more.
Human serum albumin (HSA) is a natural depot of amyloid-β peptide (Aβ), a key player in Alzheimer’s disease (AD). HSA and pro-inflammatory Ca2+-binding proteins S100A8 and S100A9 are involved in Aβ metabolism and its deposition in the brain, serving as probable triggers and therapeutic targets in AD, but their interplay with regard to Aβ binding/fibrillation is unclear. To this end, here we explore the in vitro binding of Ca2+-bound S100A8 or S100A9 to monomeric Aβ and the influence of the S100 proteins on Aβ fibrillation. The equilibrium dissociation constants of the complexes of dimeric S100A8/S100A9 with Aβ40/42 estimated by biolayer interferometry are 1–5 µM. S100A8 and S100A9 interfere with HSA binding to Aβ. Thioflavin T assay and electron microscopy data show that micromolar S100A8/S100A9 inhibit Aβ40 fibrillation, and the inhibitory effect of S100A8 exceeds that for HSA. The competition for Aβ between HSA and S100A8/S100A9 may contribute to the Aβ-HSA imbalance in the pro-inflammatory conditions in AD. Full article
Show Figures

Figure 1

24 pages, 8837 KiB  
Article
Italian Biodiversity: A Source of Edible Plant Extracts with Protective Effects Against Advanced Glycation End Product-Related Diseases
by Giulia Moretto, Raffaella Colombo, Stefano Negri, Hellas Cena, Lorena Vailati and Adele Papetti
Nutrients 2025, 17(6), 935; https://doi.org/10.3390/nu17060935 - 7 Mar 2025
Viewed by 1144
Abstract
Background: Italy’s plant biodiversity, characterized by many plant species, is an important source of bioactive secondary metabolites that help reduce the risk of the development of advanced glycation end product (AGE)-related diseases. AGEs are involved in various diseases, such as diabetes, cardiovascular, [...] Read more.
Background: Italy’s plant biodiversity, characterized by many plant species, is an important source of bioactive secondary metabolites that help reduce the risk of the development of advanced glycation end product (AGE)-related diseases. AGEs are involved in various diseases, such as diabetes, cardiovascular, and neurodegenerative disorders. Therefore, the aim of the study was to investigate the antiglycative, hypoglycemic, and neuroprotective properties of nine edible plant extracts using different in vitro assays. Methods: The ability of the extracts to counteract AGE formation was evaluated at different stages of the glycation reaction using in vitro systems based on the determination of Amadori products and the co-incubation of a model protein with a dicarbonyl compound under different experimental conditions. In addition, the extracts’ methylglyoxal (MGO) and glyoxal (GO) trapping ability was investigated. Hypoglycemic activity was assessed by measuring α-amylase inhibition, while the neuroprotective effects were explored by testing amyloid β peptide 1-42 (Aβ1-42) fibrillogenesis inhibition. Results: All extracts generally had a dose-related capacity for the inhibition of AGE formation, mainly at the intermediate stage of the glycation reaction; high trapping capacity against MGO and GO; and promising hypoglycemic properties. In addition, they affected the fibrillogenesis process by reducing mature amyloid fibril formation and altering fibril morphology. Conclusions: All tested extracts had promising anti-fibrillogenic properties. Rosa canina extract was the most active among the tested plant species given its antiglycative activity (about 80% inhibition of AGE formation), trapping capacity against MGO and GO (almost 100%), hypoglycemic effects (66.20 ± 0.88%), and anti-fibrillogenic effects (69.00 ± 4.49% inhibition), indicating its suitability in the management of AGE-related diseases and for the potential development of a novel food ingredient. Full article
(This article belongs to the Special Issue Bioactive Ingredients in Plants Related to Human Health)
Show Figures

Figure 1

19 pages, 1622 KiB  
Article
Saponins Effect on Human Insulin Amyloid Aggregation
by Eleonora Mari, Silvia Vilasi, Paolo Moretti, Maria Rosalia Mangione, Giorgia Giorgini, Roberta Galeazzi and Maria Grazia Ortore
Biomolecules 2025, 15(1), 40; https://doi.org/10.3390/biom15010040 - 31 Dec 2024
Viewed by 1499
Abstract
The misfolding and amyloid aggregation of proteins have been attracting scientific interest for a few decades, due to their link with several diseases, particularly neurodegenerative diseases. Proteins can assemble and result in insoluble aggregates that, together with intermediate oligomeric species, modify the extracellular [...] Read more.
The misfolding and amyloid aggregation of proteins have been attracting scientific interest for a few decades, due to their link with several diseases, particularly neurodegenerative diseases. Proteins can assemble and result in insoluble aggregates that, together with intermediate oligomeric species, modify the extracellular environment. Many efforts have been and are devoted to the search for cosolvents and cosolutes able to interfere with amyloid aggregation. In this work, we intensively study the effect of saponins, bioactive compounds, on human insulin aggregation. To monitor the kinetic of amyloid aggregation following secondary structure changes, we perform fluorescence and UV-Visible absorption spectroscopies, using Thioflavin T and Congo Red as amyloid specific probes, and Circular Dichroism. To study the overall structural features and size of aggregates, we perform Synchrotron Small-Angle X-ray Scattering and Dynamic Light Scattering experiments. The morphology of the aggregates was assessed by Atomic Force Microscopy. To deepen the understanding of the saponins interaction with insulin, a Molecular Dynamics investigation is performed, too. The reported data demonstrate that saponins interfere with the amyloid aggregation by inducing a strong inhibition on the formation of insulin fibrils, likely through specific interactions with insulin monomers. A dose-dependent effect is evident, and amyloid inhibition is already clear when saponins are just 0.01% w/w in solution. We suggest that saponins, which are natural metabolites present in a wide range of foods ranging from grains, pulses, and green leaves to sea stars and cucumbers, can be promising metabolites to inhibit human insulin aggregation. This basic research work can pave the way to further investigations concerning insulin amyloidosis, suggesting the use of saponins as amyloid inhibitors and/or stabilizing agents in solution. Full article
(This article belongs to the Collection Feature Papers in Molecular Structure and Dynamics)
Show Figures

Graphical abstract

16 pages, 8969 KiB  
Article
The Contrasting Effect of Sodium Alginate on Lysozyme and Albumin Denaturation and Fibril Formation
by Diliara R. Khaibrakhmanova, Polina R. Kuzivanova, Bulat R. Gainutdinov, Timur I. Magsumov, Alena A. Nikiforova and Igor A. Sedov
Biophysica 2024, 4(4), 651-666; https://doi.org/10.3390/biophysica4040043 - 12 Dec 2024
Cited by 2 | Viewed by 1289
Abstract
The effect of sodium alginate on the denaturation and aggregation behavior of bovine serum albumin and hen egg-white lysozyme was studied. Large amounts of polysaccharide increase the thermal stability of albumin due to the weak binding interactions. At the same time, sodium alginate [...] Read more.
The effect of sodium alginate on the denaturation and aggregation behavior of bovine serum albumin and hen egg-white lysozyme was studied. Large amounts of polysaccharide increase the thermal stability of albumin due to the weak binding interactions. At the same time, sodium alginate can reduce the quantity of amyloid fibrils formed by albumin under denaturing conditions, which is a consequence of the stabilization of the native protein form by glycan binding. In the case of lysozyme, the polysaccharide has no influence on the thermal stability of the protein in 2 M guanidinium hydrochloride. However, the inhibition of fibril formation with an increase in the lag time was observed, which is explained by the binding of sodium alginate to lysozyme fibrils, but not to the protein monomer. The molecular nature of the binding interactions between alginate and the studied proteins was elucidated using molecular docking and known experimental structures of glycan–protein complexes. Full article
(This article belongs to the Collection Feature Papers in Biophysics)
Show Figures

Figure 1

13 pages, 2257 KiB  
Article
Exosomal MALAT1 from Rapid Electrical Stimulation-Treated Atrial Fibroblasts Enhances Sox-6 Expression by Downregulating miR-499a-5p
by Cheng-Yen Chuang, Bao-Wei Wang, Ying-Ju Yu, Wei-Jen Fang, Chiu-Mei Lin, Kou-Gi Shyu and Su-Kiat Chua
Cells 2024, 13(23), 1942; https://doi.org/10.3390/cells13231942 - 22 Nov 2024
Cited by 2 | Viewed by 1167
Abstract
Background: Atrial fibrillation (AF) is a common cardiac arrhythmia associated with significant morbidity and mortality. Rapid electrical stimulation (RES) of atrial fibroblasts plays a crucial role in AF pathogenesis, but the underlying molecular mechanisms remain unclear. This study investigates the regulatory axis involving [...] Read more.
Background: Atrial fibrillation (AF) is a common cardiac arrhythmia associated with significant morbidity and mortality. Rapid electrical stimulation (RES) of atrial fibroblasts plays a crucial role in AF pathogenesis, but the underlying molecular mechanisms remain unclear. This study investigates the regulatory axis involving MALAT1, miR-499a-5p, and SOX6 in human cardiac fibroblasts from adult atria (HCF-aa) under RES conditions. Methods: HCF-aa were subjected to RES at 0.5 V/cm and 10 Hz. The expression levels of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), miR-499a-5p, and SRY-Box Transcription Factor 6 (SOX6) were measured using qPCR and Western blot analyses. Luciferase reporter assays were performed to confirm target relationships. The effects of MALAT1 siRNA, miR-499a-5p mimics/inhibitors, and SOX6 overexpression on gene expression and apoptosis were assessed. Results: RES increased exosomal MALAT1 expression, peaking at 2 h. MiR-499a-5p levels initially increased, then decreased at 2 h, coinciding with peak MALAT1 expression. SOX6 mRNA and protein levels increased, peaking at 4 and 6 h, respectively. Luciferase assays confirmed MALAT1 and SOX6 as miR-499a-5p targets. MALAT1 knockdown increased miR-499a-5p levels and reduced SOX6 expression. MiR-499a-5p overexpression decreased SOX6 levels and inhibited RES-induced apoptosis. Conclusion: In HCF-aa under RES, increased exosomal MALAT1 expression counteracts miR-499-5p’s suppression of SOX6, suggesting that MALAT1-containing exsosomes derived from HCF-aa may offer a novel cell-free therapeutic approach for AF. Full article
Show Figures

Figure 1

28 pages, 7444 KiB  
Article
Exploring the Potential of Biomimetic Peptides in Targeting Fibrillar and Filamentous Alpha-Synuclein—An In Silico and Experimental Approach to Parkinson’s Disease
by Sophia A. Frantzeskos, Mary A. Biggs and Ipsita A. Banerjee
Biomimetics 2024, 9(11), 705; https://doi.org/10.3390/biomimetics9110705 - 18 Nov 2024
Viewed by 1703
Abstract
Alpha-synuclein (ASyn) is a protein that is known to play a critical role in Parkinson’s disease (PD) due to its propensity for misfolding and aggregation. Furthermore, this process leads to oxidative stress and the formation of free radicals that cause neuronal damage. In [...] Read more.
Alpha-synuclein (ASyn) is a protein that is known to play a critical role in Parkinson’s disease (PD) due to its propensity for misfolding and aggregation. Furthermore, this process leads to oxidative stress and the formation of free radicals that cause neuronal damage. In this study, we have utilized a biomimetic approach to design new peptides derived from marine natural resources. The peptides were designed using a peptide scrambling approach where antioxidant moieties were combined with fibrillary inhibition motifs in order to design peptides that would have a dual targeting effect on ASyn misfolding. Of the 20 designed peptides, 12 were selected for examining binding interactions through molecular docking and molecular dynamics approaches, which revealed that the peptides were binding to the pre-NAC and NAC (non-amyloid component) domain residues such as Tyr39, Asn65, Gly86, and Ala85, among others. Because ASyn filaments derived from Lewy body dementia (LBD) have a different secondary structure compared to pathogenic ASyn fibrils, both forms were tested computationally. Five of those peptides were utilized for laboratory validation based on those results. The binding interactions with fibrils were confirmed using surface plasmon resonance studies, where EQALMPWIWYWKDPNGS, PYYYWKDPNGS, and PYYYWKELAQM showed higher binding. Secondary structural analyses revealed their ability to induce conformational changes in ASyn fibrils. Additionally, PYYYWKDPNGS and PYYYWKELAQM also demonstrated antioxidant properties. This study provides insight into the binding interactions of varying forms of ASyn implicated in PD. The peptides may be further investigated for mitigating fibrillation at the cellular level and may have the potential to target ASyn. Full article
Show Figures

Figure 1

Back to TopTop