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

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (486)

Search Parameters:
Keywords = toxic protein aggregation

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
25 pages, 635 KB  
Review
Beyond Antioxidants: The Emerging Role of Nrf2 Activation in Amyotrophic Lateral Sclerosis (ALS)
by Minoo Sharbafshaaer, Roberta Pepe, Rosaria Notariale, Fabrizio Canale, Gioacchino Tedeschi, Alessandro Tessitore, Paolo Bergamo and Francesca Trojsi
Int. J. Mol. Sci. 2025, 26(20), 9872; https://doi.org/10.3390/ijms26209872 - 10 Oct 2025
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder involving the progressive degeneration of upper and lower motor neurons. While oxidative stress, RNA-binding protein (RBP) pathology, mitochondrial dysfunction, and glial–neuronal dysregulation is involved in ALS pathogenesis, current therapies provide limited benefit, underscoring the need [...] Read more.
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder involving the progressive degeneration of upper and lower motor neurons. While oxidative stress, RNA-binding protein (RBP) pathology, mitochondrial dysfunction, and glial–neuronal dysregulation is involved in ALS pathogenesis, current therapies provide limited benefit, underscoring the need for multi-target disease-modifying strategies. Nuclear factor erythroid 2-related factor 2 (Nrf2), classically regarded as a master regulator of redox homeostasis, has recently emerged as a central integrator of cellular stress responses relevant to ALS. Beyond its canonical antioxidant function, Nrf2 regulates critical pathways involved in mitochondrial quality control, proteostasis, nucleocytoplasmic transport, RNA surveillance, and glial reactivity. Experimental models demonstrate that astrocyte-specific Nrf2 activation enhances glutathione metabolism, suppresses neuroinflammation, promotes stress granule disassembly, and reduces RBP aggregation. In C9orf72-linked ALS, Nrf2 activation mitigates dipeptide repeat protein toxicity and restores RNA processing fidelity via modulation of nonsense-mediated decay and R-loop resolution. Recent advances in Nrf2-targeted interventions including Keap1–Nrf2 protein–protein interaction inhibitors, dual Nrf2/HSF1 activators, and cell-type-selective Adeno-associated virus 9 (AAV9) vectors show promise in preclinical ALS models. These multimodal approaches highlight Nrf2’s therapeutic versatility and potential to address the upstream convergence points of ALS pathogenesis. Taken together, positioning Nrf2 as a systems-level regulator offers a novel framework for developing precision-based therapies in ALS. Integrating Nrf2 activation with RNA- and glia-directed strategies may enable comprehensive modulation of disease progression at its molecular roots. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

22 pages, 1526 KB  
Article
Temporal Interactome Mapping of Human Tau in Drosophila Reveals Progressive Mitochondrial Engagement and Porin/VDAC1-Dependent Modulation of Toxicity
by Eleni Tsakiri, Martina Samiotaki, Efthimios M. C. Skoulakis and Katerina Papanikolopoulou
Int. J. Mol. Sci. 2025, 26(19), 9741; https://doi.org/10.3390/ijms26199741 - 7 Oct 2025
Viewed by 188
Abstract
Tau protein misfolding and aggregation are central to Tauopathies, yet the temporal dynamics of Tau interactions in vivo remain poorly understood. Here, we applied quantitative proteomics to demonstrate that the interactome of human Tau in adult Drosophila brains changes dynamically over a 12-day [...] Read more.
Tau protein misfolding and aggregation are central to Tauopathies, yet the temporal dynamics of Tau interactions in vivo remain poorly understood. Here, we applied quantitative proteomics to demonstrate that the interactome of human Tau in adult Drosophila brains changes dynamically over a 12-day time course, revealing a progressive shift from early cytosolic and ribosomal associations to late enrichment of mitochondrial and synaptic partners. Notably, the mitochondrial pore protein Porin/VDAC1 was identified as a late-stage interactor and functional analyses demonstrated that Tau overexpression impairs mitochondrial respiration, elevates oxidative damage, and disrupts carbohydrate homeostasis. To validate this temporally specific interaction, Porin was downregulated, resulting in reduced Tau mitochondrial association, phosphorylation and aggregation. Paradoxically, however, Porin attenuation exacerbated Tau-induced toxicity, including shortened lifespan, locomotor deficits, and impaired learning. These findings indicate that while Porin facilitates pathological Tau modifications, it is also essential for neuronal resilience, highlighting a complex role in modulating Tau toxicity. Our study provides a temporal map of Tau-associated proteome changes in vivo and identifies mitochondria as critical mediators of Tau-driven neurodegeneration. Full article
(This article belongs to the Special Issue Genetic Advances in Neurobiology of Health and Disease)
Show Figures

Figure 1

42 pages, 1304 KB  
Review
Exploring Protein Misfolding and Aggregate Pathology in Neurodegenerative Diseases: From Molecular Mechanisms to Clinical Interventions
by Joel Theophilus Johnson, Fila Winifred Awosiminiala and Christian Kosisochukwu Anumudu
Appl. Sci. 2025, 15(18), 10285; https://doi.org/10.3390/app151810285 - 22 Sep 2025
Viewed by 821
Abstract
Neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease are characterized by progressive neuronal loss, driven mainly by the misfolding, aggregation, and accumulation of each disease’s specific proteins. These pathogenic aggregates, including tau, α-synuclein, TDP-43, and huntingtin, disrupt cellular proteostasis and [...] Read more.
Neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease are characterized by progressive neuronal loss, driven mainly by the misfolding, aggregation, and accumulation of each disease’s specific proteins. These pathogenic aggregates, including tau, α-synuclein, TDP-43, and huntingtin, disrupt cellular proteostasis and initiate cascades of neuroinflammation, oxidative stress, mitochondrial dysfunction, and synaptic failure. While protein aggregation has been a long-recognized hallmark of these disorders, growing evidence points towards a more complex interplay of initial molecular pathways with defects in RNA processing, stress granule pathology, and cell-type-specific vulnerability. Notably, such events may manifest differentially with respect to sex and are further modulated by age-related loss of the protein quality control processes like the ubiquitin–proteasome pathway, autophagy–lysosome pathway, and molecular chaperones. This review synthesizes current insights into the structural and functional dynamics of protein aggregation and its significance for neuronal well-being. It highlights the role of post-translational modifications, prion-like transmission, and aggregation kinetics in the regulation of toxicity. The review further discusses promising therapeutic strategies centered on restoring proteostasis, including small molecules that inhibit aggregation, protein clearance pathway enhancers, immunotherapy, antioxidant therapy, and diagnostic prospects such as the identification of reliable molecular signatures in bodily fluids that can reflect pathological changes even before clinical symptoms emerge. Advancements in single-cell transcriptomics and multi-omics platforms, which are changing our understanding of disease onset and progression and opening avenues for precision medicine and personalized treatments, were also discussed. Ultimately, deciphering the molecular logic that distinguishes physiological from pathological protein assemblies and understanding how cellular systems fail to adapt under stress will be key to the development of effective, disease-modifying therapies for these debilitating disorders. Full article
Show Figures

Figure 1

15 pages, 13169 KB  
Article
Assessment of the Accumulation of Certain Metals in Human Globus pallidus Using Particle-Induced X-Ray Emission (PIXE), Scanning Electron Microscopy (SEM) and Energy-Dispersive Microanalysis (EDX)
by Martin Kopáni, Daniel Kosnáč, Ján Pánik, Miroslav Ješkovský, Jakub Zeman, Pavel P. Povinec and Štefan Polák
Appl. Sci. 2025, 15(18), 9897; https://doi.org/10.3390/app15189897 - 10 Sep 2025
Viewed by 346
Abstract
Metals are essential for the physiological and biochemical processes in the human brain. However, their accumulation can cause neurotoxic effects, including the generation of reactive oxygen species and structural changes in biomolecules. This study aimed to assess the presence and distribution of metals [...] Read more.
Metals are essential for the physiological and biochemical processes in the human brain. However, their accumulation can cause neurotoxic effects, including the generation of reactive oxygen species and structural changes in biomolecules. This study aimed to assess the presence and distribution of metals in the human globus pallidus internus using Particle-Induced X-ray Emission (PIXE) and Scanning Electron Microscopy with Energy-Dispersive X-ray (SEM-EDX). Post-mortem brain tissue samples from six individuals without clinical neuropathological findings were analysed. PIXE analysis revealed the presence of Fe, Cr, Al, Zn, Pb, and Ca. SEM-EDX analysis provided the qualitative elemental composition of an observed aggregate, revealing C, N, O, Na, Ca, Al, Si, S, K, Mg, Cl, Fe, Ni, and Cr. Our findings suggest that metal accumulation in the brain can result from environmental pollution and protein aggregation, as well as biomineralisation processes that sequester metal ions to mitigate their harmful effects. A deeper understanding of these accumulation pathways could contribute to improved therapeutic strategies for neurological diseases associated with metal toxicity. Full article
Show Figures

Figure 1

22 pages, 2847 KB  
Review
Catalase Functions and Glycation: Their Central Roles in Oxidative Stress, Metabolic Disorders, and Neurodegeneration
by Fahad A. Alhumaydhi, Hina Younus and Masood Alam Khan
Catalysts 2025, 15(9), 817; https://doi.org/10.3390/catal15090817 - 27 Aug 2025
Viewed by 1517
Abstract
Catalase, a pivotal antioxidant enzyme, plays a central role in converting hydrogen peroxide (H2O2) into oxygen and water, thereby safeguarding cells from oxidative damage. In patients with diabetes, obesity, Alzheimer’s disease (AD), and Parkinson’s disease (PD), catalase becomes increasingly [...] Read more.
Catalase, a pivotal antioxidant enzyme, plays a central role in converting hydrogen peroxide (H2O2) into oxygen and water, thereby safeguarding cells from oxidative damage. In patients with diabetes, obesity, Alzheimer’s disease (AD), and Parkinson’s disease (PD), catalase becomes increasingly susceptible to non-enzymatic glycation, resulting in enzyme inactivation, oxidative stress, and defective mitochondrial function. This review uniquely emphasizes catalase glycation as a converging pathological mechanism that bridges metabolic and neurodegenerative disorders, underscoring its translational significance beyond prior general reviews on catalase function. In patients with metabolic diseases, glycation impairs β-cell function and insulin signaling, while in patients with neurodegeneration, it accelerates protein aggregation, mitochondrial dysfunction, and neuroinflammation. Notably, the colocalization of glycated catalase with amyloid-β and α-synuclein highlights its potential role in protein aggregation and neuronal toxicity, a mechanism not previously addressed. Therapeutically, targeting catalase glycation opens up new avenues for intervention. Natural and synthetic agents can be used to protect catalase activity by modulating glyoxalase activity, heme integrity, or carbonyl stress. Vitamins C and E, along with agents like sulforaphane and resveratrol, exert protection through complementary mechanisms, beyond ROS scavenging. Moreover, novel strategies, including Nrf2 activation and receptor for advanced glycation end products (RAGE) inhibition, are showing promise in restoring catalase activity and halting disease progression. By focusing on glycation-specific mechanisms and proposing targeted therapeutic approaches, this review positions catalase glycation as a novel and clinically relevant molecular target in patients with chronic diseases and a viable candidate for translational research aimed at improving clinical outcomes. Full article
(This article belongs to the Section Biocatalysis)
Show Figures

Graphical abstract

30 pages, 5591 KB  
Article
Expanding the Knowledge of the Molecular Effects and Therapeutic Potential of Incomptine A for the Treatment of Non-Hodgkin Lymphoma: In Vivo and Bioinformatics Studies, Part III
by Normand García-Hernández, Fernando Calzada, Elihú Bautista, José Manuel Sánchez-López, Miguel Valdes, Claudia Velázquez and Elizabeth Barbosa
Pharmaceuticals 2025, 18(9), 1263; https://doi.org/10.3390/ph18091263 - 25 Aug 2025
Viewed by 723
Abstract
Background/Objectives: Non-Hodgkin lymphoma (NHL) is a group of blood cancers that arise in the lymphatic nodes and other tissues after an injury to the DNA of B/T lineage and NK lymphocytes. Recently, we reported that incomptine A (IA) has in [...] Read more.
Background/Objectives: Non-Hodgkin lymphoma (NHL) is a group of blood cancers that arise in the lymphatic nodes and other tissues after an injury to the DNA of B/T lineage and NK lymphocytes. Recently, we reported that incomptine A (IA) has in vivo antilymphoma properties. This research aimed to evaluate the effects of IA in the treatment of NHL using antilymphoma activity, Tandem Mass Tag (TMT), and bioinformatics approaches. Methods: The antilymphoma activity of IA was tested on male Balb/c mice inoculated with U-937 cells. Also, TMT, gene ontology enrichment, Reactome pathway, Kyoto Encyclopedia of Gene and Genomes pathway, molecular docking, toxicoinformatic, and pharmaceutical analyses were performed. Results: By TMT analysis of the altered levels of proteins present in the lymph nodes of Balb/c mice with NHL and treated with IA, we identified 106 significantly differentially expressed proteins (DEPs), including Il1rap, Ifi44, Timd4, Apoa4, and Fabp3 as well as Myh3, Eno 2, and H4c11. Among these, the Fhl1 result was the most important cluster altered and a potential core target of IA for the treatment of NHL. Network pharmacology studies have revealed that DEPs are associated with processes such as muscle contraction, glycolysis, hemostasis, epigenetic regulation of gene expression, transport of small molecules, neutrophil extracellular trap formation, adrenergic signaling in cardiomyocytes, systemic lupus erythematosus, alcoholism, and platelet activation, signaling, and aggregation. Computational studies revealed strong binding affinities with six proteins associated with cancer, positive pharmacokinetic properties, and no toxicity. Conclusions: Our contribution suggests that IA may be a compound with potential therapeutic effects against NHL. Full article
Show Figures

Figure 1

21 pages, 1692 KB  
Review
Unraveling the Mystery of Hemoglobin in Hypoxia-Accelerated Neurodegenerative Diseases
by Zhengming Tian, Feiyang Jin, Zhuowen Geng, Zirui Xu, Qianqian Shao, Guiyou Liu, Xunming Ji and Jia Liu
Biomolecules 2025, 15(9), 1221; https://doi.org/10.3390/biom15091221 - 25 Aug 2025
Viewed by 1051
Abstract
Hypoxic stress is increasingly recognized as a convergent pathological factor in various age-related neurodegenerative diseases (NDDs), encompassing both acute events such as stroke and traumatic brain injury (TBI), and chronic disorders including Parkinson’s disease (PD), Alzheimer’s disease (AD), and amyotrophic lateral sclerosis (ALS). [...] Read more.
Hypoxic stress is increasingly recognized as a convergent pathological factor in various age-related neurodegenerative diseases (NDDs), encompassing both acute events such as stroke and traumatic brain injury (TBI), and chronic disorders including Parkinson’s disease (PD), Alzheimer’s disease (AD), and amyotrophic lateral sclerosis (ALS). Recent studies have revealed that hemoglobin (Hb), beyond its classical oxygen-transport function, exhibits unexpected expression and functional relevance within the central nervous system. Notably, both cerebral and circulating Hb appear to be dysregulated under hypoxic and aging conditions, potentially influencing disease onset and progression of these diseases. However, Hb’s impact on neurodegeneration appears to be context-dependent: in acute NDDs, it may exert neuroprotective effects by stabilizing mitochondrial and iron homeostasis, whereas in chronic NDDs, aberrant Hb accumulation may contribute to toxic protein aggregation and neuronal dysfunction. This review provides an integrative overview of the emerging roles of Hb in hypoxia-related NDDs, highlighting both shared and distinct mechanisms across acute and chronic conditions. We further discuss potential therapeutic implications of targeting Hb-related pathways in NDDs and identify key gaps for future investigation. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

18 pages, 2471 KB  
Article
Coiled-Coil Structures Mediate the Intercellular Propagation of Huntingtin
by Marco Bosica, Chiara Grasselli, Andrea Panfili, Franca Orsini and Luana Fioriti
Int. J. Mol. Sci. 2025, 26(17), 8162; https://doi.org/10.3390/ijms26178162 - 22 Aug 2025
Viewed by 567
Abstract
Huntington’s Disease (HD) originates from the expansion of a polyglutamine (PolyQ) tract in the huntingtin protein (Htt), which can assume a coiled-coil fold (Cc). We previously found that Cc structures mediate the aggregation and toxicity of polyQ Htt. Since polyQ Htt aggregates were [...] Read more.
Huntington’s Disease (HD) originates from the expansion of a polyglutamine (PolyQ) tract in the huntingtin protein (Htt), which can assume a coiled-coil fold (Cc). We previously found that Cc structures mediate the aggregation and toxicity of polyQ Htt. Since polyQ Htt aggregates were previously found to be internalized by cells, here we hypothesize that Cc structures might be implicated in the intercellular propagation of Htt aggregates. To test this hypothesis, we performed experiments using human cell lines expressing Htt proteins with different probabilities to acquire a Cc fold. We found that Htt with reduced Cc structures were released significantly less compared to Htt with intact Cc structures. We also found that Cc structures mediate the internalization of Htt proteins in recipient cells. Together, these results underline the importance of the Cc structure in the process of intercellular propagation of Htt polyQ aggregates and suggest that interfering with Cc formation might be a therapeutic strategy for HD. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

22 pages, 4751 KB  
Article
Biophysical Insights into the Binding Interactions of Inhibitors (ICA-1S/1T) Targeting Protein Kinase C-ι
by Radwan Ebna Noor, Shahedul Islam, Tracess Smalley, Katarzyna Mizgalska, Mark Eschenfelder, Dimitra Keramisanou, Aaron Joshua Astalos, James William Leahy, Wayne Charles Guida, Aleksandra Karolak, Ioannis Gelis and Mildred Acevedo-Duncan
Biophysica 2025, 5(3), 36; https://doi.org/10.3390/biophysica5030036 - 11 Aug 2025
Viewed by 591
Abstract
The overexpression of atypical protein kinase C-iota (PKC-ι) is a biomarker for carcinogenesis in various cell types, such as glioma, ovarian, renal, etc., manifesting as a potential drug target. In previous in vitro studies, ICA-1S and ICA-1T, experimental candidates for inhibiting PKC-ι, have [...] Read more.
The overexpression of atypical protein kinase C-iota (PKC-ι) is a biomarker for carcinogenesis in various cell types, such as glioma, ovarian, renal, etc., manifesting as a potential drug target. In previous in vitro studies, ICA-1S and ICA-1T, experimental candidates for inhibiting PKC-ι, have demonstrated their specificity and promising efficacy against various cancers. Moreover, the in vivo studies have demonstrated low toxicity levels in acute and chronic murine models. Despite these prior developments, the binding affinities of the inhibitors were never thoroughly explored from a biophysical perspective. Here, we present the biophysical characterizations of PKC-ι in combination with ICA-1S/1T. Various methods based on molecular docking, light scattering, intrinsic fluorescence, thermal denaturation, and heat exchange were applied. The biophysical characteristics including particle sizing, thermal unfolding, aggregation profiles, enthalpy, entropy, free energy changes, and binding affinity (Kd) of the PKC-ι in the presence of ICA-1S were observed. The studies indicate the presence of domain-specific stabilities in the protein–ligand complex. Moreover, the results indicate a spontaneous reaction with an entropic gain, resulting in a possible entropy-driven hydrophobic interaction and hydrogen bonds in the binding pocket. Altogether, these biophysical studies reveal important insights into the binding interactions of PKC-ι and its inhibitors ICA-1S/1T. Full article
(This article belongs to the Collection Feature Papers in Biophysics)
Show Figures

Figure 1

9 pages, 1972 KB  
Communication
Proteomic Analysis of Heavy Metal-Induced Toxicity Using the Cellular Slime Mould Dictyostelium discoideum: Effects of Copper Exposure on Aggregation and Protein Expression
by Atsuko Itoh, Koji Kurihara and Ryo Shoji
Toxics 2025, 13(8), 665; https://doi.org/10.3390/toxics13080665 - 8 Aug 2025
Viewed by 481
Abstract
The cellular slime mould Dictyostelium discoideum is a soil-dwelling eukaryotic organism that undergoes distinctive morphological changes during starvation, making it a promising candidate for bioassay development. In this study, we evaluated the effects of copper (Cu) exposure on the morphological transformation of D. [...] Read more.
The cellular slime mould Dictyostelium discoideum is a soil-dwelling eukaryotic organism that undergoes distinctive morphological changes during starvation, making it a promising candidate for bioassay development. In this study, we evaluated the effects of copper (Cu) exposure on the morphological transformation of D. discoideum and performed a comparative proteomic analysis. Copper exposure on agar media delayed aggregate formation by 3.5 h compared to the controls. Approximately 280 protein spots were detected using immobilised pH gradient two-dimensional gel electrophoresis followed by silver staining. Three spots disappeared upon exposure to Cu. Based on isoelectric point and molecular weight analyses, the proteins were predicted to be formin-1, a cytoplasmic regulator of adenylyl cyclase (CRAC), and a tetratricopeptide repeat (TPR)-containing protein. Formin-1 and CRAC are involved in aggregation processes. These findings suggest that Cu disrupts aggregation-related protein expression in D. discoideum and highlight the potential of D. discoideum-based bioassays using proteomic biomarkers for environmental monitoring. Full article
(This article belongs to the Section Ecotoxicology)
Show Figures

Graphical abstract

14 pages, 4166 KB  
Article
Development and Characterization of a Novel α-Synuclein-PEST H4 Cell Line for Enhanced Drug Screening in α-Synucleinopathies
by Nancy Carullo, Viktor Haellman, Simon Gutbier, Sonja Schlicht, Thien Thuong Nguyen, Rita Blum Marti, Philippe Hartz, Lothar Lindemann and Lina Schukur
Int. J. Mol. Sci. 2025, 26(15), 7205; https://doi.org/10.3390/ijms26157205 - 25 Jul 2025
Viewed by 671
Abstract
Alpha-Synuclein (α-Syn) is a presynaptic neuronal protein implicated in the pathogenesis of Parkinson’s disease (PD) and other synucleinopathies, primarily through its aggregation into insoluble fibrils. The extended α-Syn half-life necessitates treatment durations that are incompatible with efficient high-throughput drug screening, can risk compound [...] Read more.
Alpha-Synuclein (α-Syn) is a presynaptic neuronal protein implicated in the pathogenesis of Parkinson’s disease (PD) and other synucleinopathies, primarily through its aggregation into insoluble fibrils. The extended α-Syn half-life necessitates treatment durations that are incompatible with efficient high-throughput drug screening, can risk compound stability or cause cellular toxicity. To address this, we inserted a PEST sequence, a motif known to promote rapid protein degradation, at the C-terminus of the SNCA gene using CRISPR/Cas9 to create a novel cell line with reduced α-Syn half-life. This modification accelerates α-Syn turnover, providing a robust model for studying α-Syn dynamics and offering a platform that is applicable to other long-lived proteins. Our results demonstrate a six-fold reduction in α-Syn half-life, enabling the rapid detection of changes in protein levels and facilitating the identification of molecules that modulate α-Syn production and degradation pathways. Using inhibitors of the proteasome, transcription, and translation further validated the model’s utility in examining various mechanisms that impact protein levels. This novel cell line represents a significant advancement for studying α-Syn dynamics and offers promising avenues to develop therapeutics for α-synucleinopathies. Future research should focus on validating this model in diverse experimental settings and exploring its potential in high-throughput screening applications. Full article
(This article belongs to the Special Issue Whole-Cell System and Synthetic Biology, 2nd Edition)
Show Figures

Figure 1

19 pages, 9689 KB  
Article
Anionic Lipid Catalyzes the Generation of Cytotoxic Insulin Oligomers
by Jhinuk Saha, Audrey Wolszczak, Navneet Kaur, Malitha C. Dickwella Widanage, Samuel D. McCalpin, Riqiang Fu, Jamel Ali and Ayyalusamy Ramamoorthy
Biomolecules 2025, 15(7), 994; https://doi.org/10.3390/biom15070994 - 11 Jul 2025
Viewed by 468
Abstract
The misfolding and aggregation of proteins into amyloidogenic assemblies are key features of several metabolic and neurodegenerative diseases. Human insulin has long been known to form amyloid fibrils under various conditions, which affects its bioavailability and function. Clinically, insulin aggregation at recurrent injection [...] Read more.
The misfolding and aggregation of proteins into amyloidogenic assemblies are key features of several metabolic and neurodegenerative diseases. Human insulin has long been known to form amyloid fibrils under various conditions, which affects its bioavailability and function. Clinically, insulin aggregation at recurrent injection sites poses a challenge for diabetic patients who rely on insulin therapy. Furthermore, decreased responsiveness to insulin in type 2 diabetic (T2D) patients may lead to its overproduction and accumulation as aggregates. Earlier reports have reported that various factors such as pH, temperature, agitation, and the presence of lipids or other proteins influence insulin aggregation. Our present study aims to elucidate the effects of non–micellar anionic DMPG (1,2–dimyristoyl–sn–glycero–3–phosphoglycerol) lipids on insulin aggregation. Distinct pathways of insulin aggregation and intermediate formation were observed in the presence of DMPG using a ThT fluorescence assay. The formation of soluble intermediates alongside large insulin fibrils was observed in insulin incubated with DMPG via TEM, DLS, and NMR as opposed to insulin aggregates generated without lipids. 13C magic angle spinning solid–state NMR and FTIR experiments indicated that lipids do not alter the conformation of insulin fibrils but do alter the time scale of motion of aromatic and aliphatic side chains. Furthermore, the soluble intermediates were found to be more cytotoxic than fibrils generated with or without lipids. Overall, our study elucidates the importance of anionic lipids in dictating the pathways and intermediates associated with insulin aggregation. These findings could be useful in determining various approaches to avoid toxicity and enhance the effectiveness of insulin in therapeutic applications. Full article
(This article belongs to the Collection Feature Papers in 'Biomacromolecules: Proteins')
Show Figures

Figure 1

17 pages, 807 KB  
Review
The Potential Regulators of Amyloidogenic Pathway of APP Processing in Alzheimer’s Disease
by Daria Krawczuk, Agnieszka Kulczyńska-Przybik and Barbara Mroczko
Biomedicines 2025, 13(7), 1513; https://doi.org/10.3390/biomedicines13071513 - 20 Jun 2025
Viewed by 1429
Abstract
The amyloidogenic processing of amyloid precursor protein (APP) plays a pivotal role in the pathogenesis of Alzheimer’s disease (AD), primarily through the generation of amyloid-beta (Aβ) peptides, which aggregate to form toxic plaques in the brain. The regulation of amyloidogenic APP processing is [...] Read more.
The amyloidogenic processing of amyloid precursor protein (APP) plays a pivotal role in the pathogenesis of Alzheimer’s disease (AD), primarily through the generation of amyloid-beta (Aβ) peptides, which aggregate to form toxic plaques in the brain. The regulation of amyloidogenic APP processing is a complex interplay of enzymes, proteins, and signaling pathways, all of which contribute to the development and progression of Alzheimer’s disease. Understanding the intricate mechanisms and molecular players involved in APP processing substantially enhances our knowledge of Alzheimer’s disease pathology and holds promise for the development of biomarkers of ongoing pathology at the earliest stages of Alzheimer’s disease. In this review, we aimed to investigate selected factors that regulate the amyloidogenic pathway of APP processing. Full article
(This article belongs to the Section Neurobiology and Clinical Neuroscience)
Show Figures

Figure 1

20 pages, 3980 KB  
Article
A Single-Chain Variable Fragment Antibody Alleviates Inflammation and Apoptosis of Neurons by Inhibiting Tau Aggregation
by Zongbao Wang, Jingye Lin, Peipei Chang, Mingzhu Sun and Sen Li
Biomolecules 2025, 15(6), 872; https://doi.org/10.3390/biom15060872 - 15 Jun 2025
Viewed by 835
Abstract
Tau pathology is one of the main pathological features of Alzheimer’s disease (AD). Intracellular Tau may be released to the extracellular space upon neuron degeneration, where it has the potential to be toxic to other neurons. The propagation of Tau pathology, mediated by [...] Read more.
Tau pathology is one of the main pathological features of Alzheimer’s disease (AD). Intracellular Tau may be released to the extracellular space upon neuron degeneration, where it has the potential to be toxic to other neurons. The propagation of Tau pathology, mediated by extracellular Tau aggregates, may underlie the pathogenesis of AD. Antibody therapies targeting Tau proteins are, therefore, considered highly promising. In this study, the cytotoxicity of extracellular Tau aggregates on SH-SY5Y cells was examined. The effect of extracellular Tau aggregates on intracellular Tau aggregation was also studied using a FRET-based assay. The extracellular Tau aggregates were found to cause intracellular Tau aggregation after entering the cells; meanwhile, ROS (reactive oxygen species) induced by Tau aggregates facilitated this process. A single-chain variable fragment antibody (scFv T1) inhibits Tau aggregation both extracellularly and intracellularly. ScFv T1 also inhibited the accumulation of ROS and alleviated the inflammation and apoptosis induced by Tau aggregates. These findings could provide experimental support for the study of neurotoxicity and related mechanisms of extracellular Tau aggregates, in addition to providing insights into the development of novel therapeutic agents to treat AD. Full article
(This article belongs to the Section Chemical Biology)
Show Figures

Figure 1

14 pages, 1413 KB  
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 929
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

Back to TopTop