bims-proned Biomed News
on Proteostasis in neurodegeneration
Issue of 2025–09–21
thirteen papers selected by
Verena Kohler, Umeå University



  1. Chin Med J (Engl). 2025 Sep 16.
       ABSTRACT: Neurodegenerative diseases constitute a group of chronic disorders characterized by the progressive loss of neurons. Major neurodegenerative conditions include Alzheimer's disease, Parkinson's disease, Huntington's disease, frontotemporal lobar degeneration, and amyotrophic lateral sclerosis. Pathologically, these diseases are marked by the accumulation of aggregates formed by pathological proteins such as amyloid-β, tau, α-synuclein, and TAR DNA-binding protein 43. These proteins assemble into amyloid fibrils that undergo prion-like propagation and dissemination, ultimately inducing neurodegeneration. Understanding the biology of these protein aggregates is fundamental to elucidating the pathophysiology of neurodegenerative disorders. In this review, we summarize the molecular mechanisms underlying the aggregation and transmission of pathological proteins, the processes through which these protein aggregates trigger neurodegeneration, and the interactions between different pathological proteins. We also provide an overview of the current diagnostic approaches and therapeutic strategies targeting pathological protein aggregates.
    Keywords:  Alzheimer’s disease; Amyloid-β; Amyotrophic lateral sclerosis; Biomarker; Huntington’s disease; Parkinson’s disease; Tau; α-synuclein
    DOI:  https://doi.org/10.1097/CM9.0000000000003802
  2. ACS Chem Neurosci. 2025 Sep 20.
      α-Synuclein aggregation in synucleinopathies involves abnormal accumulation of α-synuclein protein in neurons. This aggregation process generates oligomers, protofibrils, and fibrils, disrupting cellular function and contributing to the progression of neurodegeneration observed in Parkinson's disease, multiple system atrophy, and dementia with Lewy bodies. Typically, these aggregates range in size from tens to hundreds of nanometers to a few micrometers. The intermediate-sized aggregation species, so-called oligomers, have been implicated in the neurotoxicity observed in Parkinson's disease. Small-sized, soluble oligomers exhibit pore formation in lipid bilayers, impair synaptic transmission, and induce oxidative stress, ultimately leading to neuronal dysfunction. This review explores the size-dependent toxicity of α-synuclein aggregates, focusing on how variations in aggregate size influence their pathological effects in neurodegenerative diseases. It discusses the diverse structural forms of α-synuclein, including monomers, oligomers, protofibrils, and large-sized fibrils, and their differential impact on cellular function and viability. By elucidating the size-dependent mechanisms underlying α-synuclein toxicity, this review aims to inform therapeutic strategies targeting specific aggregate sizes to mitigate neuronal damage and halt Parkinson's disease progression.
    Keywords:  fibrils; oligomers; protofibrils; size-dependent toxicity; synucleinopathies; α-synuclein
    DOI:  https://doi.org/10.1021/acschemneuro.5c00356
  3. Front Neurol. 2025 ;16 1604076
      A key hallmark of neurodegenerative diseases (NDDs) is the formation of neurotoxic protein aggregates, which are considered to reflect inadequate protein quality control (PQC). In agreement with this fundamental pathophysiologic characteristic, the two main cellular systems responsible for cellular protein removal - the ubiquitin-proteasome system (UPS) and autophagy - have been extensively studied in the context of NDD. The involvement of these proteolytic machineries was interpreted in different ways - some pointed them as dysfunctional systems that may underlie pathogenesis, while others suggested they fulfill protective roles which delay the clinical presentation of these diseases. Perhaps not surprisingly, the growing body of knowledge concerning the different types of NDD portrays a more complex picture, and no distinct generalization can be made regarding the contribution of either the neurotoxic protein substrate(s) or proteolytic system(s) to the development of NDD. For instance, in Parkinson's disease, the toxic aggregation of α-synuclein, Parkinson's canonical culprit protein, can stem from seemingly unrelated events. Among them, alterations in α-synuclein itself, a mutation in Parkin - an E3 ubiquitin ligase targeting proteins and organelles to proteasomal and lysosomal degradation, respectively, as well as a mutation in LRRK2 - a kinase postulated to be linked with α-synuclein through their common removal by chaperone-mediated autophagy. Also, in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD), the toxic aggregation of one protein - TDP-43 - can result from defects in other proteins, some of which are related to proteostasis, such as the shuttle protein Optineurin and the E3 ubiquitin ligase VCP. In contrast, ALS and FTLD demonstrate how common abnormalities leading to neurotoxic aggregate formation, may present clinically in profoundly different ways, from motor dysfunction to behavioral changes. In Alzheimer's Disease, the leading cause for dementia, rare cases were linked directly with PQC as they are caused by a mutation in one of the genes encoding ubiquitin itself, while the majority of cases were not directly linked to components of the two main proteolytic systems. All-in-all, the UPS and autophagy are heavily intertwined with NDD, either as part of the problem or as mitigating factors, and hopefully - as platforms for future therapeutics. In this review, we shall dissect NDDs from the perspective of protein turnover pathways, aiming to track both common and unique patterns of PQC failure in this group of diseases, which differ significantly from one another both in their clinical manifestations and affected anatomic regions, yet share the common trait of abnormal protein accumulation. We shall review some of the mechanistic understandings concerning protein aggregation in NDDs, describing the interactions of aggregated proteins with the UPS and autophagy, discuss recent controversies around the protein aggregates' hypothesis, and point to implications for developing therapeutic strategies.
    Keywords:  autophagy; neurodegenarative disease; protein aggregates; protein quality control (PQC); ubiquitin-proteasome system
    DOI:  https://doi.org/10.3389/fneur.2025.1604076
  4. Biochim Biophys Acta Proteins Proteom. 2025 Sep 15. pii: S1570-9639(25)00037-8. [Epub ahead of print] 141099
      Protein aggregation plays a crucial role in various neurodegenerative diseases, including Huntington's disease. Understanding the factors influencing aggregation kinetics is essential for deciphering disease mechanisms. This research paper investigates the aggregation of a mutant Huntington protein (HD39Q) under various conditions, focusing on the impact of macromolecular crowding agents. The study employs multiple techniques, including fluorescence spectroscopy, circular dichroism, and nanoparticle tracking analysis, to characterize the aggregation kinetics and morphology. The results demonstrate that crowding agents significantly accelerate aggregation, with different agents exhibiting varying effects depending on their physicochemical properties. Fluorescence correlation spectroscopy provides insights into early-stage oligomerization. Confocal and scanning electron microscopy help visualize the resulting aggregates and fibrils. These findings contribute to a better understanding of how intracellular-like environments influence protein aggregation and provide valuable insights into the biophysical properties of aggregation-prone proteins.
    Keywords:  Dextran; Ficoll; Huntington protein aggregation; Imaging; Macromolecular crowding; Oligomer and fibril formation; PEG; Thioflavin T assay
    DOI:  https://doi.org/10.1016/j.bbapap.2025.141099
  5. bioRxiv. 2025 Sep 05. pii: 2025.09.05.674500. [Epub ahead of print]
      Cytoplasmic aggregation of nuclear proteins such as TDP-43 (TAR DNA-binding protein 43) and FUS (fused in sarcoma) is associated with several neurodegenerative diseases. Studies in higher cells suggest that these aggregates of TDP-43 and FUS sequester polysomes by binding RACK1 (receptor for activated C kinase 1), a ribosomal protein, thereby inhibiting global translation and contributing to toxicity. But RACK1 is also a scaffold protein with many other roles including a role in autophagy. Using yeast we find that deletion of the RACK1 ortholog, ASC1 , reduces TDP-43 toxicity, but not FUS toxicity. TDP-43 foci remain liquid like in the presence asc1Δ but they become smaller. This is consistent with the findings in cell culture. However, using double label tags we establish that ASC1 does not co-localize with TDP-43 foci, arguing against the sequestration hypothesis. Instead, ASC1 appears to influence toxicity through autophagy. We previously showed that expression of TDP-43 inhibits autophagy and TOROID (TORC1 Organized in Inhibited Domains) formation and that modifiers that rescue yeast from TDP-43 toxicity reverse these inhibitions. Here we show that FUS does not inhibit autophagy. This autophagy enhanced by asc1Δ is non-canonical, marked by reduced TOROID formation, and effectively counteracts the autophagy inhibition caused by TDP-43. Our findings suggest that ASC1 influences TDP-43 toxicity through autophagy regulation rather than polysome sequestration, highlighting autophagy as a key therapeutic target.
    Summary: TDP-43 and FUS aggregates are linked to neurodegenerative diseases. RACK1, a ribosomal protein, was previously thought to contribute to toxicity by co-localizing with these aggregates and sequestering polysomes. In yeast, deletion of ASC1 -the RACK1 homolog-reduces TDP-43 toxicity but not FUS toxicity. TDP-43 foci remain liquid-like, but ASC1 does not co-localize with them, challenging the sequestration hypothesis. Instead, asc1Δ enhances autophagy, rescuing cells from the autophagy inhibition caused by TDP-43. Unlike TDP-43, FUS does not inhibit autophagy. These findings highlight autophagy, rather than polysome sequestration, as the key mechanism of TDP-43 toxicity and its mitigation via ASC1/RACK1 reduction.
    DOI:  https://doi.org/10.1101/2025.09.05.674500
  6. Adv Protein Chem Struct Biol. 2025 ;pii: S1876-1623(25)00010-0. [Epub ahead of print]147 355-374
      Tau is a microtubule-binding, hydrophilic protein and appears randomly coiled in circular dichroism spectra. Tau can have many post-translational modifications such as phosphorylation, acetylation, SUMOylation, glycation, ubiquitinylation, etc. The abnormal phosphorylation of Tau lowers its affinity to bind the microtubules, causing to neuronal instability. Hyperphosphorylated Tau can get detach from the microtubules and get aggregate in neuronal cell body to form a neurofibrillary tangle, which leads to weaken axonal transport and cause synaptic dysfunction. Tau itself is a SUMO-1 target protein and the modified lysine has been identified as the K340 located within 4R-Tau. The interaction between Tau and SUMO-1 was confirmed by an independent study, by showing that the SUMO-1 immunoreactivity is co-localized with phosphorylated Tau. In addition to this, Tau can also be ubiquitinated and degraded by the proteasome through both ubiquitin-dependent and ubiquitin-independent pathways. Our study shows that SUMOylation at lysine K340 stimulates Tau phosphorylation and inhibits ubiquitination-mediated Tau degradation, thus favouring its aggregation.
    Keywords:  Alzheimer’s disease; Neuronal Tau; PTMs of Tau; Protein quality control; Tau SUMOylation; Tau aggregation; Tau phosphorylation
    DOI:  https://doi.org/10.1016/bs.apcsb.2025.02.001
  7. J Am Chem Soc. 2025 Sep 18.
      The aggregation of tau into amyloid fibrils is associated with Alzheimer's disease (AD) and related tauopathies. Since different tauopathies are characterized by the formation of distinct tau fibril morphologies, it is important to combine the search of tau aggregation inhibitors with the development of in vitro tau aggregation assays that recapitulate aggregation as it may occur in the brain. Here we address this problem by reporting an in vitro tau aggregation assay in which AD brain homogenates are used to seed the generation of first-generation tau fibrils in a polymorph-specific manner under quiescent conditions. These fibrils are then used to create amyloid seed libraries from which second-generation kinetic assays can be readily performed. Using this strategy, we illustrate an iterative machine learning method for the identification of small molecules for the polymorph-specific inhibition of the in vitro formation of tau fibrils. We further show that the small molecules selected by this procedure are potent inhibitors in a Drosophila tauopathy model.
    DOI:  https://doi.org/10.1021/jacs.5c12812
  8. Clin Chim Acta. 2025 Sep 15. pii: S0009-8981(25)00491-7. [Epub ahead of print] 120612
      Parkinson's disease (PD), a rapidly progressing neurodegenerative disorder, underscores the critical need for early diagnostic tools to mitigate its debilitating clinical trajectory. Central to PD pathology is the aggregation of α-synuclein (α-syn), a protein whose misfolded forms drive neuronal dysfunction and serve as a hallmark biomarker. This review explores the transformative role of fluorescent probes in detecting α-syn aggregates and other PD-related biomarkers, offering unprecedented sensitivity and specificity for early diagnosis. We systematically categorize probes into three classes, e.g., small molecules, nanomaterials, and macromolecules, detailing their design principles, mechanisms, and applications in real-time monitoring of α-syn fibrillation, dopamine metabolism, and oxidative stress pathways. Key advancements, such as the PP-BTA series for dual Aβ/α-syn detection and NIR-Lyso-FA for formaldehyde imaging, highlight the integration of multidisciplinary technologies to overcome current diagnostic limitations. Furthermore, we evaluate preclinical successes in animal models and emerging strategies for clinical translation, including high-throughput screening and multimodal imaging. By addressing challenges in probe standardization, biocompatibility, and scalability, this review provides a roadmap for advancing fluorescent probes from laboratory innovation to clinical utility, ultimately enhancing personalized therapeutic strategies and improving the quality of life for PD patients.
    Keywords:  Alpha-synuclein; Early diagnosis; Fluorescent probes; Parkinson's disease
    DOI:  https://doi.org/10.1016/j.cca.2025.120612
  9. Neurochem Int. 2025 Sep 16. pii: S0197-0186(25)00126-3. [Epub ahead of print] 106053
      The intricate interplay between brain iron homeostasis and pathological α-Synuclein (αSyn) in Parkinson's Disease (PD) is still unclear. αSyn is the primary protein involved in disease progression and promotes cytotoxicity by impairing dopamine synthesis, mitochondrial dysfunction and disrupts brain iron homeostasis. Iron has further revealed to induce αSyn aggregation however, the exact mechanism of interaction between iron and αSyn is uncertain. Many studies have demonstrated an imbalance of iron homeostasis in PD pathogenesis, including macroautophagy related to αSyn aggregate and accumulated ferritin degradation. Inducible ferritinophagy has been suggested as a possible therapeutic to combat raised ferritin levels and reinstate iron homeostasis however this may lead to increased ferroptosis. Despite limited knowledge of ferritinophagy induced ferroptosis in PD there is a possible link to neurodegeneration. This review aims to summarise the current understanding of the chemical interactions between iron and αSyn and possible binding affinities in biologically relevant models, while further highlighting αSyn's pathological role in neurodegeneration and aberrant iron homeostasis. Developing knowledge surrounding the complex interactions between αSyn and iron in PD will aid in possible therapeutic strategies to combat PD disease progression.
    Keywords:  Metal ions; Neurochemistry; Neurodegeneration; Parkinson’s Disease; α-Synuclein
    DOI:  https://doi.org/10.1016/j.neuint.2025.106053
  10. Adv Protein Chem Struct Biol. 2025 ;pii: S1876-1623(24)00089-0. [Epub ahead of print]147 333-353
      Tau protein accumulation is one of the characteristic features of Alzheimer's disease (AD). Their accumulation is driven by the formation of intermediate toxic oligomers of Tau to the highly ordered neurofibrillary tangles. Cellular machineries engage different types of proteins such as, chaperone-co-chaperones complex, ubiquitin, kinases, proteases etc., to clear the aberrantly accumulated Tau protein which otherwise would cause neuronal death. In the milieu of proteotoxicity, it would be significant for the cell to follow a specific path for Tau clearance. Under this circumstance, cells express key proteins and other accessory proteins specific to the pathway. This is known to be dependent on the post-translational modifications and mutations associated with Tau. The processes involved maintenance of proteins homeostasis in cells collectively called proteostasis. The proteostasis involve the synthesis of proteins by ribosomes, protein folding mostly by chaperons and the degradation of improperly folded or unwanted proteins. Autophagy is the mechanism to eradicate unwanted, non-functional and toxic proteins from the cell. Proteostasis plays a pivotal role in maintaining the normal cellular environment in the expense of considerable amount of energy. AD is the prevalent type of dementia associated with aging, which is characterized by aggregation of Tau.
    Keywords:  Alzheimer’s disease; Autophagy; Chaperone-mediated pathway; LAMP-2A; Proteostasis; Tauopathies
    DOI:  https://doi.org/10.1016/bs.apcsb.2024.09.003
  11. Sci Adv. 2025 Sep 19. 11(38): eadw1061
      Aggregation of α-synuclein (α-syn) represents a pathogenic hallmark of Parkinson's disease (PD). Using exogenous molecular chaperone systems has the potential to stabilize native conformations and inhibit the aberrant aggregation of α-syn, yet inefficient blood-brain barrier (BBB) penetration and insufficient accumulation at PD sites limit their application. Herein, we developed chemotactic chaperone nanomotors (CNMs) that exploit the unique pathological microenvironment of PD lesions, characterized by elevated inducible nitric oxide synthase (iNOS) levels. The CNMs consist of a chemotactic-targeting module capable of sensing an iNOS concentration gradient, an α-syn-specific recognition module with conformation-sensitive binding domains, and a protein regulatory module with a hydrophobic microdomain that can stabilize α-syn and prevent misfolding. Results demonstrate that CNMs can achieve active chemotactic navigation across BBB and dual modulation of α-syn proteostasis through aggregate dissolution and prevention of misfolding. CNMs not only supplement exogenous chaperone activity but also restore endogenous protein stabilization mechanisms, concurrently reducing neuroinflammatory markers, promoting the application of nanochaperones for PD treatment.
    DOI:  https://doi.org/10.1126/sciadv.adw1061
  12. Biomacromolecules. 2025 Sep 19.
      The aggregation of intrinsically disordered proteins (IDPs) is of significant interest due to its role in proteopathies and nutritional and pharmaceutical potential. This study investigates the mechanism of Ca(II)-induced aggregation of an IDP β-casein (β-CN) using dynamic light scattering, cryo-transmission electron microscopy, and molecular dynamics simulations. Upon Ca(II) addition, β-CN undergoes successive induction, growth, and saturation phases, forming amorphous aggregates. Aggregation kinetics are highly dependent on the Ca(II) concentration. At a molar ratio exceeding 5:1, the hydrodynamic diameter of β-CN increased from 20 nm (oligomer) to >100 nm (aggregate product) in a minute. The induction phase is driven by neutralizing the phosphorylated groups via Ca(II) binding, while subsequent growth and saturation phases are governed by agglomeration of intermediate aggregates with nuclei-exposed oligomers, eventually forming aggregate products after conformational relaxation. We demonstrate that the porosity and tunable assembly of β-CN aggregates enable efficient encapsulation of bioactive molecules, offering promising applications in nanonutrition and nanotheranostics.
    DOI:  https://doi.org/10.1021/acs.biomac.5c01076
  13. J Mol Biol. 2025 Sep 11. pii: S0022-2836(25)00504-2. [Epub ahead of print] 169438
      The co-deposition of amyloid fibrils from different precursor proteins is a topic of increasing relevance for protein misfolding diseases. Using cryo-electron microscopy (cryo-EM), we here determined the structures of two serum amyloid A (SAA) protein-derived amyloid fibril morphologies that were extracted from a mouse strain that is primarily known to be associated with apolipoprotein A-II-derived amyloid fibrils. The two fibril morphologies show the same protomer conformation as in previously reported ex vivo amyloid fibrils from SAA protein but a different relative arrangement of fibril protein stacks. These data establish that serum amyloid A-derived amyloid fibrils share the same fibril protein fold in different mouse strains and disease contexts.
    Keywords:  Amyloid; amyloidosis; apolipoproteins; protein aggregation
    DOI:  https://doi.org/10.1016/j.jmb.2025.169438