Fact-checked by Grok 2 weeks ago

Protein aggregation

Protein aggregation is the process by which multiple protein molecules assemble into higher-order structures, such as soluble oligomers, insoluble , or amorphous aggregates, typically driven by non-native intermolecular interactions that arise from partial unfolding, misfolding, or of the proteins. This phenomenon is a fundamental aspect of protein behavior in , influenced by factors including protein concentration, environmental conditions like and temperature, and intrinsic sequence properties that expose aggregation-prone regions. While aggregation can occur reversibly under physiological conditions, uncontrolled or pathological aggregation often leads to stable, dysfunctional complexes that disrupt cellular . In biological contexts, protein aggregation plays essential roles in cellular organization and adaptation. For instance, proteins can form membraneless biomolecular condensates through liquid-liquid phase separation (LLPS), creating like stress granules that sequester RNAs and signaling molecules during cellular stress, thereby protecting and facilitating recovery in organisms such as and mammals. These aggregates, ranging from liquid-like droplets to more solid fibers, also contribute to developmental processes, such as the formation of P granules in C. elegans embryos for specification, and functional amyloids like Pmel17 fibrils that support synthesis in melanosomes. Regulation of these processes involves molecular chaperones, such as and Hsp100 families, which prevent unwanted aggregation or promote disaggregation via ATP-dependent mechanisms. Conversely, aberrant protein aggregation is a hallmark of numerous neurodegenerative diseases, where misfolded proteins form toxic inclusions that impair neuronal function and promote . In , aggregation of amyloid-β peptides into plaques and hyperphosphorylated into neurofibrillary tangles disrupts synaptic transmission and induces . features α-synuclein aggregates in Lewy bodies, which interfere with mitochondrial function and signaling, while involves polyglutamine-expanded protein forming intranuclear inclusions that alter . is linked to aggregates of TAR DNA-binding protein-43 (TDP-43) and superoxide dismutase 1 (), contributing to degeneration through prion-like propagation and collapse. These pathological aggregates often resist degradation by cellular quality control systems, including the ubiquitin-proteasome pathway and autophagy-lysosome machinery, exacerbating disease progression. Beyond biology, protein aggregation poses significant challenges in biotechnology, particularly for therapeutic proteins, where even low levels of aggregates can trigger immunogenicity and reduce efficacy during storage or administration. Strategies to mitigate aggregation include engineering mutations to shield aggregation-prone regions and optimizing formulation conditions to enhance protein stability. Overall, understanding the dual nature of protein aggregation—beneficial in regulated forms and detrimental when dysregulated—remains crucial for advancing treatments for aggregation-related disorders and improving biopharmaceutical design.

Fundamentals

Definition and Overview

Protein aggregation refers to the abnormal association of misfolded or unfolded proteins into larger, often insoluble structures such as , plaques, or amorphous aggregates. This process typically arises from the exposure of hydrophobic regions in destabilized proteins, leading to self-association that bypasses normal folding pathways. The biophysical principles underlying protein aggregation are driven primarily by interactions, where non-polar residues cluster to minimize contact with the aqueous environment, supplemented by hydrogen bonding that stabilizes β-sheet-rich structures and van der Waals forces that contribute to close packing within aggregates. These non-covalent interactions facilitate the transition from soluble monomers to oligomeric intermediates and eventually to higher-order assemblies, with the often initiating the process by promoting partial unfolding. Early observations of amyloid deposits date to the mid-19th century, when Rudolph Virchow in 1854 coined the term "" to describe waxy tissue abnormalities exhibiting iodine staining similar to starch, based on light microscopy studies of pathological specimens. A key milestone came in 1959, when electron microscopy by Cohen and Calkins revealed the fibrillar ultrastructure of deposits, approximately 80–100 Å in width and of indeterminate length, shifting understanding toward protein-based fibril formation. Protein aggregation occurs across all organisms, from prokaryotes to eukaryotes, where it serves pathological roles in over 30 human diseases including neurodegenerative disorders, but also fulfills normal functions such as forming reversible in under stress to sequester misfolded proteins temporarily. In higher eukaryotes, however, persistent aggregates often disrupt cellular and contribute to toxicity. The of protein aggregation are commonly modeled using the nucleation-polymerization framework, which captures the lag phase of formation followed by rapid . A basic form of this model describes the rate of change in aggregate concentration [A] as: \frac{d[A]}{dt} = k_n [M]^n + k_e [A][M] where [M] is the concentration, k_n is the nucleation rate constant, n is the size of the critical , and k_e is the rate constant; this highlights the concentration-dependent step and linear growth onto existing aggregates.

Types of Protein Aggregates

Protein aggregates can be broadly classified into amorphous and ordered types based on their structural organization, with each exhibiting distinct morphological properties, formation characteristics, and detection methods. Amorphous aggregates are disordered, non-fibrillar clumps that lack long-range order, often forming rapidly under denaturing conditions such as heat or chemical agents like high concentrations of or . These aggregates typically arise from partially folded intermediates exposing hydrophobic regions, leading to nonspecific associations, and they are prevalent in both refolding experiments and cellular overexpression systems. Ordered aggregates, in contrast, display highly structured architectures, most notably amyloid fibrils characterized by a cross-β-sheet conformation where β-strands align perpendicular to the fibril axis. These fibrils exhibit a diffraction pattern, including a 4.7 meridional reflection indicative of hydrogen-bonded β-strands and a 10-11 equatorial reflection from inter-sheet spacing. fibrils are thermodynamically stable, insoluble without denaturants, and often 7-12 nm in diameter, composed of twisted protofilaments. Detection commonly relies on Thioflavin T (ThT) fluorescence, which increases upon binding to the cross-β structure, or birefringence under polarized light. Oligomers represent early, soluble intermediates in the aggregation pathway, typically comprising 10-100 protein molecules and enriched in β-sheet content, though heterogeneous in structure. These species, such as Aβ dimers or pentamers/hexamers, are often globular and more cytotoxic than mature due to their ability to interact with cellular membranes and disrupt function. Unlike , oligomers may form off-pathway and are detected using coupled with (SEC-MALS) or fluorescence probes like aniline blue or pentameric thiophenyl (pFTAA) that bind β-sheets. Inclusion bodies are dense, cytoplasmic aggregates primarily observed in prokaryotic cells, such as Escherichia coli, during overexpression of recombinant proteins. Composed mainly of misfolded proteins with 50-70% native-like secondary structure but increased β-sheet content, these amorphous structures appear as refractile inclusions under phase-contrast microscopy and are insoluble, often requiring denaturation for recovery. Detection involves techniques like transmission electron microscopy (TEM) or attenuated total reflectance Fourier-transform infrared (ATR-FTIR) spectroscopy, which reveals β-sheet bands around 1623-1637 cm⁻¹. Prion aggregates exemplify infectious conformers, where a misfolded , such as PrPSc, propagates by templating the conversion of native PrPC into the pathogenic state, forming amyloid-like fibrils with in-register parallel β-sheets. These self-propagating structures underlie transmissible spongiform encephalopathies and are detected via cryo-electron microscopy (cryo-EM) or protein misfolding cyclic amplification (PMCA) assays that amplify the infectious conformer. Hybrid aggregates, involving co-aggregation of multiple proteins like Aβ and , further complicate morphology by mixing distinct misfolded species into shared β-sheet-rich assemblies.47636-7/fulltext) Amorphous aggregates are readily identified by their rapid onset without a lag and detection via measurements, where light scattering increases due to particle formation larger than the incident wavelength, or (DLS) for size distribution. In contrast, amyloid fibrils show a sigmoidal kinetic profile with a lag , confirmed by the specific 4.7 X- reflection diagnostic of cross-β ordering.

Causes and Mechanisms

Genetic and Mutational Factors

Genetic alterations, including s and repeat expansions in protein-coding genes, can destabilize protein structures, promoting misfolding and subsequent aggregation. These mutations often alter key physicochemical properties such as hydrophobicity or charge, reducing the stability of the native fold and favoring aberrant intermolecular interactions. For instance, in sickle cell anemia, a in the β-globin gene substitutes with at position 6 (Glu6Val), introducing a hydrophobic residue that facilitates the of deoxygenated into rigid fibers under low-oxygen conditions. This change exemplifies how single substitutions can shift the thermodynamic balance toward aggregation-prone conformations. Repeat expansions, particularly of trinucleotide sequences, represent another critical mutational mechanism driving protein aggregation in neurodegenerative disorders. In , expansion of CAG repeats in the huntingtin (HTT) gene beyond 36 units encodes an elongated polyglutamine tract in the HTT protein, exceeding a pathological that promotes into β-sheet-rich . This length-dependent aggregation is thought to arise from increased propensity for polyglutamine chains to form intermolecular hydrogen bonds, leading to insoluble inclusions that impair cellular function. Mutations affecting chaperone proteins or the folding machinery further exacerbate aggregation by impairing mechanisms. For example, variants in BAG3, a co-chaperone that regulates HSP70-mediated refolding, disrupt the network, causing accumulation of misfolded clients and widespread protein aggregates in myofibrillar . Such defects reduce the capacity for recognition and ATP-dependent remodeling, allowing aggregation-prone intermediates to persist. Inheritance patterns of aggregation-related disorders often follow autosomal dominant transmission, where a single mutant suffices to confer risk. Familial amyloidosis, caused by mutations in the TTR , exemplifies this, with over 100 variants leading to destabilized tetrameric transthyretin that dissociates and aggregates into in tissues like the heart and nerves. Similarly, in familial , mutations in the precursor protein () , such as the London mutation (Val717Ile), enhance Aβ peptide production or alter its aggregation kinetics, accelerating plaque formation. From an evolutionary perspective, the accumulation of deleterious mutations contributes to age-related protein aggregation vulnerability. Mutation accumulation theory posits that late-acting harmful variants, including those predisposing to proteostasis collapse, evade purifying selection due to reduced reproductive fitness pressure post-reproduction, leading to their buildup in aging populations. This and accrual aligns with observed increases in protein misfolding during , underscoring aggregation as a hallmark of evolutionary trade-offs in .

Errors in Protein Synthesis and Folding

Errors in protein synthesis and folding represent a primary intrinsic cause of protein aggregation, distinct from sequence alterations due to mutations. During , the decodes mRNA into polypeptides, but inaccuracies such as tRNA misacylation—where aminoacyl-tRNA synthetases incorrectly charge tRNAs with non-cognate —can incorporate erroneous residues, leading to misfolded proteins prone to aggregation. Similarly, ribosomal stalling, often triggered by rare codons or damaged mRNA, exposes nascent chains to premature folding attempts, resulting in aberrant polypeptides that aggregate co-translationally. These translational defects increase the population of non-native conformers, which intermolecular interactions can stabilize into aggregates, as evidenced in studies of and bacterial systems where stalling induces inclusion body formation. Co- and post-translational modifications further contribute to aggregation when dysregulated. Improper disulfide bond formation, catalyzed by oxidoreductases in the (), can lock proteins into non-native conformations if incorrect pairings occur, promoting misfolding and subsequent aggregation in secretory pathways. For instance, in neurodegenerative contexts, aberrant crosslinking in proteins like TDP-43 exacerbates amyloidogenic aggregation by stabilizing toxic oligomers. Likewise, errors in , such as incomplete or hyper-glycosylation, disrupt the steric shielding that normally prevents hydrophobic exposure; hyper-glycosylation in recombinant systems has been shown to induce ER stress and aggregation by altering protein . These modifications, intended to guide folding, instead create kinetic barriers when erroneous, leading to accumulation of aggregation-prone intermediates. Chaperone overload exacerbates these issues during high-expression scenarios, such as viral infections or recombinant production, where the demand for / in eukaryotes or /GroES in prokaryotes outstrips availability. binds nascent chains to prevent premature collapse, but saturation allows unfolded domains to interact, forming aggregates; experiments in E. coli demonstrate that overexpressing aggregation-prone proteins depletes , resulting in up to 50% insoluble fractions. translation systems lacking chaperones further illustrate this: purified ribosomes synthesizing proteins like yield rapid aggregation without addition, highlighting the essential role of chaperones in averting kinetic traps during folding. Failures in signal peptide cleavage also contribute to aggregation, particularly for ER-targeted proteins. The signal peptidase complex cleaves the N-terminal signal sequence post-translocation, but mutations or overload can leave uncleaved peptides, causing membrane anchoring defects and exposure of hydrophobic regions that drive oligomerization. In mammalian cells, uncleaved signal sequences in HLA class I molecules lead to ER retention and aggregation, mimicking inclusion bodies in disease states. Such errors compound translational inaccuracies, increasing the aggregate burden without invoking external stresses. The concept, introduced by Wolynes and colleagues, provides a kinetic for these errors: proteins navigate a rugged energy landscape toward the native state, but mistranslation or modification defects deepen off-pathway traps, where partially folded states with exposed hydrophobics aggregate intermolecularly. In this model, the 's width reflects sequence-specific , with translational errors narrowing productive paths and favoring kinetic arrest in aggregated minima, as simulated for helical bundle proteins. This theoretical lens underscores how maintains smoothness, preventing the aggregation cascades observed in proteotoxic conditions.

Environmental and Cellular Stresses

Environmental and cellular stresses play a critical role in inducing protein misfolding and subsequent aggregation by disrupting the native conformational stability of proteins. , particularly heat shock, causes partial unfolding of proteins, exposing hydrophobic regions that promote intermolecular interactions and aggregation. For instance, acute heat stress in cells leads to the reversible aggregation of proteins, including those involved in processing, as a protective against denaturation. The rate of protein aggregation often exhibits a U-shaped dependence, with minimal aggregation near physiological temperatures (around 37°C) due to optimal folding , increasing at both lower temperatures through cold-induced structural rigidification and higher temperatures via enhanced unfolding and collision rates. This non-Arrhenius behavior has been observed in the aggregation of IgG1 Fc fragments, where rates accelerate below 40°C and above 60°C under neutral conditions.31366-8) Oxidative stress, arising from elevated (ROS), further exacerbates protein aggregation by chemically modifying vulnerable residues, such as , leading to the formation of disulfide-linked aggregates. ROS-induced oxidation of cysteine thiols can form aberrant bridges, stabilizing misfolded conformations and promoting oligomerization, as seen in various neurodegenerative contexts. In conditions like cerebral ischemia-reperfusion injury, triggers superoxide dismutase 1 () aggregation in through disulfide reduction and monomer formation, contributing to neuroinflammatory damage. These modifications not only impair protein function but also hinder clearance mechanisms, amplifying aggregate accumulation. Alterations in and ionic environments also drive protein aggregation, particularly in intracellular compartments like lysosomes where acidic conditions prevail. Low promotes amyloid formation by destabilizing soluble monomers, favoring β-sheet-rich structures; for example, β-amyloid (Aβ) rapidly assembles into toxic at 5.0–6.0, mimicking lysosomal acidity. Ionic imbalances, such as elevated concentrations, modulate electrostatic interactions, accelerating fibrillation of Aβ40 by screening repulsive charges between . In lysosomal dysfunction, impaired acidification leads to incomplete and aggregate buildup, underscoring the role of in preventing amyloidogenic pathways. Chemical denaturants like and hydrochloride induce aggregation by disrupting non-covalent interactions essential for protein stability, including hydrogen bonds. preferentially forms hydrogen bonds with the protein backbone, weakening intramolecular interactions and exposing aggregation-prone surfaces, while hydrochloride primarily acts through ionic disruption of salt bridges. These agents are commonly used to study unfolding pathways, revealing that intermediate denaturant concentrations often yield the highest aggregate levels due to partial unfolding without complete solubilization. In eukaryotic cells, (ER) stress activates the unfolded protein response (UPR), which, if unresolved, leads to the accumulation of aggregate-prone proteins. ER stress from unfolded protein overload triggers UPR sensors like IRE1, PERK, and ATF6, aiming to restore by upregulating chaperones; however, chronic activation promotes widespread aggregation beyond the ER, including cytosolic proteins forming prion-like structures. This response highlights how cellular stressors can propagate misfolding cascades, linking acute ER perturbations to broader proteotoxic events.

Contribution of Aging

Aging profoundly impacts , the cellular network responsible for maintaining , leading to a progressive collapse that favors protein aggregation. This decline manifests as reduced capacity to handle proteotoxic stress, with chaperone proteins playing a central role. Molecular chaperones, such as , assist in and prevent misfolding; however, their expression and activity diminish over time. In humans, basal levels of decrease significantly with age, contributing to impaired stress responses and increased vulnerability to protein damage. This proteostasis collapse is exacerbated in senescent cells, where chaperone induction under heat stress is markedly reduced compared to young cells, allowing unfolded proteins to accumulate and form aggregates. Mitochondrial dysfunction, a hallmark of aging, further drives protein aggregation through heightened oxidative modifications. Mitochondria produce (ROS) as byproducts of , and age-related impairments in mitochondrial function elevate ROS levels, leading to oxidative damage of proteins. This results in and other modifications that alter , promoting insolubility and aggregation; for instance, oxidized protein levels increase by approximately 50-70% in aging brains. Such damage overwhelms the network, as modified proteins resist degradation and sequester functional chaperones, creating a vicious cycle of aggregate formation. Additionally, activity, essential for clearing damaged proteins, declines substantially in aged , with 26S proteasome function dropping by about 50% in the brains of old mice compared to young ones. This reduction links to the accumulation of indigestible aggregates like , a fluorescent composed of 30-70% oxidized proteins and lipids that forms in lysosomes and inhibits further , accelerating . From an evolutionary perspective, protein aggregation serves as a marker of , reflecting trade-offs in efficiency across . Long-lived invest more in protective mechanisms like chaperones and rapid to minimize aggregation risk, but even they experience age-dependent declines. Proteins with high turnover rates exhibit greater intrinsic aggregation propensity yet are safeguarded by efficient ; however, aging impairs this , increasing aggregate burden as a senescence indicator. Longitudinal studies in model organisms underscore this correlation: in , protein aggregation burden rises with age, involving over 460 proteins becoming insoluble, and directly inversely correlates with lifespan—mutations extending longevity, such as reduced insulin/IGF-1 signaling, delay aggregation and enhance survival. These findings highlight how chronic erosion over time, distinct from acute stresses, culminates in widespread aggregation as a fundamental aging feature.

Cellular Localization

In Prokaryotic Cells

In prokaryotic cells, protein aggregates primarily form in the due to the absence of membrane-bound organelles, often resulting from environmental stresses or high-level expression of foreign proteins. These aggregates, known as , are dense, amorphous structures composed of misfolded proteins that can impair cellular function if not managed. Prokaryotic aggregates, such as , primarily form intracellularly in the , though some functional amyloids assemble extracellularly via dedicated secretion pathways. Cytoplasmic in exhibit polar localization, preferentially accumulating at the cell poles rather than dispersing evenly throughout the . This positioning arises from initial formation at discrete sites such as poles, mid-cell, or quarter-cell positions, with aggregates showing limited mobility (<10% relocation per division) and tending to segregate asymmetrically into daughter cells inheriting the old pole. In non-stressed conditions, this leads to higher aggregate density in aging cells at old poles, correlating with reduced reproductive fitness (>30% growth rate decline). During recombinant , are a common outcome when overexpressing proteins, as rapid synthesis overwhelms folding chaperones, leading to hydrophobic exposure and aggregation; however, they serve as a protective sink, enabling high yields (up to grams per liter) that can be solubilized post-purification for industrial applications like enzyme production. Polar foci of aggregates in bacteria like and are driven by exclusion, where the dense DNA acts as a barrier, confining misfolded proteins and chaperone-substrate complexes to nucleoid-free polar regions. Under stress conditions such as or limitation, this mechanism seeds aggregate growth in polar zones, as seen with type III substrates forming large dynamic complexes that reverse upon stress relief. Specific chaperones like IbpA and mark these sites in E. coli, with IbpA acting as a sequestrase to bind and hold aggregation-prone proteins, preventing further while facilitating later processing; complements by aiding substrate handover to disaggregases. Bacterial disaggregases, notably ClpB, enable aggregate reversibility by cooperating with the DnaK () system to solubilize and refold proteins during recovery phases. ClpB threads aggregate polypeptides through its central pore using , initiated by DnaK coating the aggregate surface, which activates ClpB's and promotes partial unfolding for chaperone-mediated refolding rather than degradation. This dynamic process restores post-stress, with ClpB essential for thermotolerance in bacteria like E. coli. In contrast to eukaryotic systems with compartmentalized clearance, prokaryotic aggregates rely on these cytoplasmic mechanisms for sequestration and dissolution.

In Eukaryotic Cells

In eukaryotic cells, protein aggregation exhibits organelle-specific patterns that reflect the compartmentalized nature of these organisms, differing from the more uniform cytoplasmic deposition observed in prokaryotes. Misfolded secretory proteins, which enter the secretory pathway, frequently aggregate within the (ER) and Golgi apparatus due to impaired folding or overload of the machinery. In the ER, accumulation of unfolded or misfolded proteins triggers the unfolded protein response (UPR), but persistent leads to aggregation of these proteins, often forming insoluble deposits that disrupt ER . Similarly, at the Golgi, post-ER quality control mechanisms retain unfolded proteins, preventing their trafficking; failure of this retention can result in aggregation within Golgi compartments, as seen in mammalian cells where misfolded glycoproteins accumulate and are targeted for degradation. These ER- and Golgi-associated aggregates highlight the secretory pathway's vulnerability to proteotoxic in fungi, , and animals. In the and , protein aggregates often manifest as ubiquitin-positive inclusions, serving as sites for sequestration of misfolded proteins. Cytosolic aggregates are frequently transported along to form aggresomes, perinuclear structures enriched in and molecular chaperones, which act as temporary storage for damaged proteins in mammalian cells and . Nuclear aggregates, similarly ubiquitin-marked, arise from intranuclear misfolding events and can fuse to form larger inclusions, as observed in response to proteasomal inhibition across eukaryotic models. In , specific cytosolic and nuclear sites include the insoluble protein deposit (), a peripheral aggregate lacking that recruits amyloid-like structures, and the juxtanuclear (JUNQ) compartment, which is dynamic, proteasome-associated, and facilitates refolding or of ubiquitinated proteins near the . Mitochondrial protein aggregation is prominently linked to oxidative damage, where (ROS) from dysfunction oxidize proteins, promoting their misfolding and intramitochondrial inclusion formation. In eukaryotes ranging from to mammals, these aggregates sequester chaperones like Hsp60 and impair mitochondrial function, exacerbating proteotoxic stress under conditions such as aging or . Distinct intramitochondrial aggregates, including spherical bodies and tubular structures, have been identified as protective mechanisms that isolate damaged proteins while preserving integrity. In , vacuolar aggregates represent a key under abiotic stresses like , where water deficit induces protein misfolding and oxidative damage, leading to aggregate formation that is subsequently sequestered to the via for degradation. adaptors such as NBR1 target ubiquitinated aggregates, including stress-responsive proteins like COST1, transporting them into autophagosomes that fuse with the , thereby enhancing by recycling nutrients and mitigating proteotoxicity. This vacuolar sequestration underscores the plant 's role as a major protein quality control hub during environmental stress.

Aggregate Dynamics and Sequestration

Protein aggregates within s exhibit dynamic behaviors that facilitate their transport, sequestration, and structural evolution, often serving as protective mechanisms against . In eukaryotic s, misfolded proteins forming small aggregates are actively transported along toward the microtubule-organizing center (MTOC) via motors, leading to the consolidation into larger structures known as aggresomes. This -mediated process is size-selective, with larger aggregates preferentially mobilized through episodic bursts of motility, enhancing their sequestration efficiency. In , an -dependent mechanism complements this, where aggregates associate with Hsp104 and are retrogradely transported along actin cables to the mother , promoting asymmetric and clearance from daughter s. Sequestration of aggregates often occurs in membrane-less compartments formed by liquid-liquid (LLPS), which temporarily isolate potentially harmful species. In Caenorhabditis elegans, P granules exemplify such sites, where RNA-binding proteins like LAF-1 drive LLPS to create dynamic droplets that sequester client proteins, including aggregates, while maintaining fluidity for selective partitioning during . These phase-separated structures prevent widespread proteotoxicity by concentrating aggregates away from functional cellular regions, though prolonged residence can lead to further maturation. The of aggregates involve transitions from liquid-like to solid states, influencing their and propagation. Initially fluid condensates can undergo liquid-to-solid phase transitions, forming rigid that resist and serve as for further aggregation. This maturation is evident in prion-like propagation, where fibrillar the misfolding of soluble monomers, enabling intercellular spread and amplification of aggregates in a self-perpetuating manner. (FRAP) studies reveal that mature aggregates, such as those in plaques, possess immobile cores with minimal exchange, indicating a stable, non-dynamic architecture that perpetuates seeding. Additionally, aggregate is pH-sensitive; acidic environments can promote of certain assemblies, reversing formation and highlighting environmental of dynamics.

Clearance Mechanisms

Molecular Chaperone Refolding

Molecular chaperones play a crucial role in counteracting protein aggregation by actively refolding misfolded or aggregated polypeptides, thereby restoring functional conformations without . These ATP-dependent machines recognize hydrophobic regions exposed in unfolded or aggregated states, preventing further assembly into insoluble aggregates. The process involves sequential , unfolding, and release cycles that solubilize proteins, particularly under cellular stress conditions where aggregation propensity increases. The chaperone system exemplifies this refolding mechanism, utilizing to thread substrates through its central cavity in an iterative manner. Hsp70 binds to exposed hydrophobic segments of aggregated proteins, facilitated by collaboration with Hsp40 co-chaperones, which deliver substrates and stimulate to lock Hsp70 in a high-affinity state. This threading action extracts polypeptides from aggregates, allowing subsequent refolding upon release by nucleotide exchange factors. Cryo-EM structures from the 2010s have illuminated this model, revealing how Hsp70's ATPase domain powers conformational changes that pull substrates unidirectionally through the pore. However, under conditions of severe aggregate overload, such as prolonged heat stress, the system's efficiency diminishes as chaperone capacity is saturated, leading to incomplete disaggregation. In prokaryotes and , specialized disaggregases like bacterial ClpB and Hsp104 enhance Hsp70's capabilities as hexameric + ATPases that actively pull polypeptides from aggregates. These ring-shaped enzymes form central pores through which substrates are threaded in an ATP-fueled process, uncoiling even tightly packed structures like fibrils. ClpB relies on DnaK (the bacterial Hsp70 homolog) for targeting, while Hsp104 integrates with Ssa1, ensuring coordinated disaggregation. Structural insights from cryo-EM confirm a conserved threading mechanism across these homologs, where ATP binding and hydrolysis drive subunit rotations that grip and translocate chains. Complementing these systems, small heat shock proteins (sHSPs) function as ATP-independent holdases that bind to aggregation-prone intermediates, stabilizing them in a soluble state to avert further clumping. By sequestering hydrophobic surfaces, sHSPs like alphaB-crystallin prevent irreversible aggregation during acute stress, later handing off clients to ATP-dependent chaperones for refolding. This holdase activity is vital in the initial of misfolded proteins, maintaining until disaggregases can intervene.

Proteasomal and Lysosomal Degradation

Protein aggregation poses a challenge to cellular , and cells employ the -proteasome system () to degrade misfolded or aggregated proteins that cannot be refolded by chaperones. The begins with the covalent attachment of to target proteins through a hierarchical enzymatic involving E1 ubiquitin-activating enzymes, E2 ubiquitin-conjugating enzymes, and E3 ubiquitin ligases. E1 enzymes activate by forming a bond using ATP, which is then transferred to E2 enzymes; subsequently, E3 ligases facilitate the transfer of to residues on the substrate protein, often recognizing specific degradation signals. This process typically requires the formation of polyubiquitin chains, where the type of linkage determines the fate of the modified protein: K48-linked chains signal for proteasomal degradation, while K63-linked chains primarily mediate non-degradative functions such as signaling or trafficking. The 26S proteasome, a large ATP-dependent complex approximately 2.5 MDa in size, executes the degradation of ubiquitinated proteins. It consists of a 20S core particle with proteolytic activity and two 19S regulatory particles that recognize and prepare substrates. The 20S core features three main catalytic activities, including chymotrypsin-like activity at the β5 subunit, which cleaves peptide bonds after large hydrophobic residues to generate peptides from unfolded proteins. The 19S regulatory particle's Rpt ATPase subunits unfold substrates and open the gate of the 20S core, ensuring selective entry and preventing unregulated proteolysis. In parallel, lysosomal degradation handles extracellular or membrane-associated aggregates through , where cells internalize protein aggregates via receptor-mediated or fluid-phase pathways, delivering them to lysosomes for . Within the acidic lysosomal (pH ~4.5-5.0), cathepsin proteases, such as and D, perform the bulk degradation by cleaving proteins into , with cathepsins exhibiting optimal activity in this low-pH environment. Unlike the , which targets soluble or small oligomeric aggregates, lysosomal pathways are more suited for larger extracellular complexes, though both systems can intersect with chaperone-assisted delivery for efficient clearance. Recent studies have identified a stage-dependent mechanism for clearing protein aggregates during in cells, involving the chaperone BiP, activity, and inactivation of (CDK), which facilitates the dissolution and of ER-associated aggregates. Age-related decline in proteasomal function exacerbates protein aggregation, as oxidative damage and reduced expression of catalytic subunits lead to inhibited activity and accumulation of ubiquitinated substrates. In , the insoluble protein deposit (), a perivacuolar structure, sequesters non-ubiquitinated aggregates, which under stress conditions can be delivered to the for .

Autophagy Pathways

Autophagy represents a critical cellular for the degradation of large protein aggregates that exceed the capacity of proteasomal pathways, particularly through , where double-membrane vesicles known as autophagosomes engulf cytoplasmic cargo for delivery to lysosomes. In the context of protein aggregation, selective forms of , such as aggrephagy, target ubiquitinated protein aggregates for specific recognition and clearance. Aggrephagy relies on adaptor proteins like p62/SQSTM1, which bind to chains on aggregates via its ubiquitin-associated domain and simultaneously interact with lipidated LC3 on autophagosomal membranes through its LC3-interaction region, thereby linking aggregates to the autophagic machinery. The formation of autophagosomes is orchestrated by autophagy-related (Atg) proteins, which coordinate membrane nucleation, elongation, and closure; core components include the Atg1/ULK complex for initiation, the phosphatidylinositol 3-kinase complex for phospholipid production, and the Atg12 and LC3 conjugation systems for membrane expansion. A hallmark of this process is the lipidation of LC3 to form LC3-II, where LC3 is conjugated to on the autophagosomal membrane, serving as a key marker for autophagosome maturation and cargo recruitment. Once formed, autophagosomes fuse with s to form autolysosomes, where degradation occurs; this fusion is mediated by SNARE proteins, including syntaxin 17 (STX17) on the autophagosome, SNAP29, and VAMP7 or VAMP8 on the , facilitated by tethering complexes like . Beyond aggrephagy, mitophagy selectively degrades mitochondria containing aggregated or damaged proteins, often triggered by PINK1-Parkin ubiquitination pathways that recruit autophagy receptors to damaged organelles, thereby preventing the release of toxic aggregates from mitochondria. (CMA), while primarily targeting soluble proteins bearing KFERQ-like motifs via HSC70 chaperone recognition and LAMP2A translocation to lysosomes, can indirectly influence aggregation by clearing monomeric precursors before they form insoluble aggregates. In models of neurodegeneration, such as those involving Huntington's or Parkinson's diseases, pathways, including aggrephagy and mitophagy, are often upregulated as a compensatory response to protein accumulation, with enhanced LC3-II levels and Atg protein expression observed in affected neurons to mitigate proteotoxicity.

Pathological Consequences

Mechanisms of Toxicity

Protein aggregates exert toxicity through gain-of-function mechanisms, where soluble oligomers interact directly with cellular components to disrupt normal function. These oligomers, often prefibrillar species, can insert into lipid bilayers to form pore-like structures, such as β-barrel channels, leading to permeabilization and uncontrolled fluxes. This disruption compromises and membranes, particularly in mitochondria, causing leakage of cellular contents and impairment of bioenergetic processes. A key consequence is the dysregulation of calcium , as oligomers facilitate calcium influx, which triggers downstream signaling cascades including and activation of proteases like calpains. Beyond direct membrane interactions, protein aggregates induce toxicity by sequestering essential components of the network, overwhelming cellular systems. Misfolded proteins and aggregates bind to molecular chaperones such as and , titrating them away from their normal substrates and impairing the refolding of nascent or stressed proteins. Similarly, aggregates can clog the by forming insoluble inclusions that inhibit ubiquitin-dependent , leading to backlog of ubiquitinated proteins. This overload culminates in a collapse of global , where the cell's capacity to maintain protein is eroded, exacerbating misfolding of unrelated proteins and amplifying aggregate formation. Aggregates also trigger inflammatory responses by activating innate immune pathways, notably through the . Fibrillar aggregates, such as those from amyloidogenic proteins, serve as danger signals that prime and assemble the -ASC-caspase-1 complex in immune cells like and macrophages. This activation processes pro-IL-1β and pro-IL-18 into mature cytokines, promoting a proinflammatory milieu that amplifies via recruitment of additional immune cells and release of reactive mediators. At the synaptic level, certain oligomers exemplify neurotoxic gain-of-function by interfering with mechanisms; for instance, amyloid-β (Aβ) oligomers bind to receptors like NMDA and , suppressing (LTP) through oxidative modulation of synaptic proteins and disruption of glutamate signaling. This selective impairment of LTP, without affecting basal transmission, underscores how oligomers target vulnerable neuronal compartments to erode memory-related circuits. Toxicity is further propagated through seeding mechanisms, where aggregates act as templates to recruit and convert soluble monomers into misfolded conformers, facilitating intercellular . In transmissible aggregates akin to prions, this templated misfolding enables cell-to-cell propagation via exosomes, tunneling nanotubes, or direct uptake, perpetuating a cycle of aggregation across tissues. Finally, aggregates contribute to by catalyzing (ROS) generation, often through Fenton-like chemistry involving bound transition metals like iron or . Oligomers and coordinate these metals at their hydrophobic cores, enabling the reduction of H₂O₂ to hydroxyl radicals (•OH), which damage , nucleic acids, and proteins in a vicious cycle that promotes further aggregation.

Associated Diseases and Disorders

Protein aggregation is a central pathological feature in numerous human diseases, particularly those involving the accumulation of misfolded proteins into or , leading to cellular dysfunction and tissue damage. In neurodegenerative disorders, intracellular and extracellular aggregates disrupt neuronal function, while in systemic conditions, they affect multiple organs. These pathologies often share mechanisms such as prion-like propagation of aggregates, though disease-specific proteins and triggers vary. Alzheimer's disease (AD) is characterized by the formation of extracellular amyloid-beta (Aβ) plaques and intracellular neurofibrillary tangles composed of hyperphosphorylated , both resulting from aberrant protein aggregation. Less than 5% of AD cases are early-onset familial forms linked to mutations in genes like , PSEN1, and PSEN2 that accelerate Aβ production and aggregation. (PET) imaging with amyloid-specific tracers, such as florbetapir, enables noninvasive detection of Aβ plaques, aiding early diagnosis and differentiation from other dementias. In Parkinson's disease (PD), aggregation of α-synuclein into Lewy bodies within neurons of the substantia nigra contributes to dopaminergic neuron loss and motor symptoms. These intraneuronal inclusions are a hallmark of both sporadic and familial PD, with mutations in SNCA or LRRK2 promoting α-synuclein misfolding and fibrillization. Amyotrophic lateral sclerosis (ALS) involves the aggregation of superoxide dismutase 1 (SOD1) in motor neurons, forming cytoplasmic inclusions that impair axonal transport and lead to progressive paralysis. Familial ALS cases, accounting for about 10% of all ALS, often feature SOD1 mutations that destabilize the protein and enhance its propensity to aggregate. Systemic diseases also feature protein aggregation, as seen in type 2 diabetes mellitus (T2DM), where islet amyloid polypeptide (IAPP) aggregates into deposits in pancreatic β-cells, contributing to β-cell and insulin deficiency. These extracellular IAPP are present in up to 90% of T2DM pancreata at . Similarly, arises from (TTR) protein misfolding and aggregation into fibrils that infiltrate the myocardium, causing ; wild-type TTR aggregates in senile systemic , while mutant forms drive hereditary variants. Prion diseases, such as Creutzfeldt-Jakob disease (CJD), are caused by the conformational conversion of cellular protein (PrPC) into the pathogenic scrapie isoform (PrPSc), which self-propagates through templated misfolding and forms extracellular in the brain, leading to spongiform . Sporadic CJD, the most common form, accounts for about 85% of human diseases and involves PrPSc generation without identifiable external exposure. Extracellular aggregates also occur in , a secondary form triggered by chronic inflammation from conditions like or infections, where protein (SAA) deposits as primarily in kidneys and other organs, leading to organ failure. This reactive amyloidosis highlights how sustained inflammatory responses can drive systemic protein aggregation independently of genetic factors.

Biotechnological and Therapeutic Aspects

Challenges in Biomanufacturing

In recombinant protein production using as a host, protein aggregation into (IBs) represents a major obstacle, often leading to reduced yields and complicating . IBs form when overexpressed proteins misfold and aggregate, resulting in insoluble particles that can constitute up to 30-50% of total cellular protein, thereby lowering the recoverable soluble fraction and necessitating additional solubilization and refolding steps. These refolding protocols, which typically involve denaturants like or guanidine hydrochloride followed by controlled renaturation, are labor-intensive and can achieve recovery rates as low as 20-50% for some proteins, further impacting overall process efficiency. During purification, protein stability is frequently compromised by mechanical and chemical stresses, such as forces from pumping or mixing and shifts in steps, which expose hydrophobic regions and promote aggregation. High rates, often exceeding 10^4 s^-1 in industrial-scale operations, can induce partial unfolding, particularly in viscous solutions, leading to dimer and higher-order aggregate formation at rates that correlate with exposure duration. Similarly, excursions below 4 or above 8 during ion-exchange or inactivation can alter electrostatic interactions, destabilizing proteins and increasing aggregate content by 5-20% in susceptible cases like monoclonal antibodies (mAbs). In mAb , aggregation contributes to substantial production losses, with levels during reaching up to 30% in some processes, necessitating removal steps that can reduce overall yields by 20-50%. Regulatory guidelines emphasize minimizing aggregates due to risks, with acceptable levels of soluble aggregates typically limited to below 5-10% in injectable to ensure product and . To mitigate interface-induced aggregation during and storage, non-ionic such as or 80 are commonly added at concentrations of 0.01-0.1% (w/v), where they compete for adsorption at air-liquid or solid-liquid interfaces, reducing protein exposure and formation by up to 90% in stressed conditions.

Strategies for Prevention and Treatment

Strategies for preventing and treating protein aggregation primarily involve pharmacological interventions that stabilize proteins, inhibit and elongation steps in the aggregation pathway, or enhance clearance mechanisms, alongside genetic approaches targeting underlying mutations. Small-molecule stabilizers, such as , bind to (TTR) to prevent its dissociation into monomers that initiate formation, thereby reducing all-cause mortality and cardiovascular hospitalizations in patients with TTR cardiomyopathy (ATTR-CM). In the phase 3 ATTR-ACT , treatment resulted in a 30% reduction in mortality risk over 30 months compared to placebo. Kinetic inhibitors target the early phase of aggregation, where monomers form critical oligomers, by altering the thermodynamic and kinetic barriers to formation; for instance, compounds designed to bind transiently to aggregation-prone intermediates can extend the lag phase of assembly by orders of magnitude . Gene therapies, particularly antisense oligonucleotides (ASOs), offer targeted silencing of mutant alleles prone to aggregation. Tofersen, an ASO approved for SOD1-mutated amyotrophic lateral sclerosis (ALS), binds SOD1 mRNA to reduce SOD1 protein levels by 33% (95% CI -47 to -16%) in cerebrospinal fluid, slowing disease progression as evidenced by decreased neurofilament light chain levels in phase 1-2 trials. Chaperone inducers like arimoclomol amplify the heat shock protein (HSP) response under stress, enhancing protein refolding and inhibiting aggregation in neurodegenerative contexts; in the phase 2/3 trial for rapidly progressive SOD1-ALS, arimoclomol treatment was associated with a slower decline of 0.5 points per month on the ALSFRS-R scale compared to placebo (95% CI -0.63 to 1.63) over up to 12 months. Antibody-based therapies promote clearance of extracellular aggregates, such as amyloid-beta (Aβ) plaques in . Lecanemab, a targeting protofibrils, reduces Aβ deposition and slows cognitive decline by 27% on the CDR-SB scale in early-stage patients over 18 months, as shown in the phase 3 Clarity AD trial. In , where islet amyloid polypeptide (IAPP) aggregation contributes to beta-cell dysfunction, agonists like have demonstrated preclinical efficacy in reducing IAPP accumulation by improving remodeling and turnover in obese mouse models. For biotechnological applications, techniques engineer aggregation-resistant protein variants by iteratively mutating and selecting for improved and during . An selection platform using assays has successfully evolved single-chain variable fragments (scFvs) with enhanced resistance to aggregation, yielding variants that maintain functionality when reformatted as full immunoglobulins for therapeutic . These approaches collectively address aggregation in both therapeutic and industrial settings by prioritizing interventions that modulate or without relying solely on downstream clearance.

References

  1. [1]
    Therapeutic Protein Aggregation: Mechanisms, Design, and Control
    Protein aggregation (sometimes referred to as non-native aggregation) denotes the process(es) by which protein molecules assemble into stable complexes composed ...
  2. [2]
    A guide to studying protein aggregation - Housmans - FEBS Press
    Dec 4, 2021 · In this review we summarize the most commonly investigated aspects of protein aggregation with some popular corresponding methods.Protein thermodynamics · Aggregation prediction tools · Methods for investigating...
  3. [3]
    Protein Aggregation and Disaggregation in Cells and Development
    This review presents examples of aggregates found in biological systems, how they are used, and cellular strategies that control aggregation and disaggregation.
  4. [4]
    Protein misfolding in neurodegenerative diseases: implications and ...
    Mar 13, 2017 · A hallmark of neurodegenerative proteinopathies is the formation of misfolded protein aggregates that cause cellular toxicity and contribute to cellular ...Roles Of Misfolded Proteins... · Therapeutic Targets · Targeting Chaperones
  5. [5]
    Distinct stress conditions result in aggregation of proteins ... - Nature
    Apr 18, 2016 · Protein aggregation is the abnormal association of misfolded proteins into larger, often insoluble structures. Aggregation can be classified ...
  6. [6]
    Intermolecular interactions underlie protein/peptide phase ... - Nature
    Oct 4, 2023 · The driving forces and kinetics for assembly vary from protein to protein ... H-bonding and van der Waals forces) stock solutions were prepared.<|separator|>
  7. [7]
    Theoretical Aspects of Protein Aggregation and Neurodegenerative ...
    The hydrophobic effect is an important driving force behind the folding processes that allow the hydrophobic residue to find in the internal core of the 3D ...3.1 Protein Quality Control · 4. Free Energy Landscape · 6. Neurodegenerative...
  8. [8]
    Review: history of the amyloid fibril - PubMed
    Rudolph Virchow, in 1854, introduced and popularized the term amyloid to denote a macroscopic tissue abnormality that exhibited a positive iodine staining ...
  9. [9]
    Increased Aggregation Is More Frequently Associated to Human ...
    Sep 4, 2015 · Protein aggregation has been recognized to contribute to the development of more than 30 human diseases such as Alzheimer and Parkinson disease.
  10. [10]
    Protein aggregation in bacteria - PMC - PubMed Central
    Protein aggregation generally results in a loss of protein function and can thus impair critical cellular functions that are required for growth and survival.
  11. [11]
  12. [12]
  13. [13]
    Amyloid-type Protein Aggregation and Prion-like Properties of ...
    Jun 17, 2021 · This review will focus on the process of amyloid-type protein aggregation. Amyloid fibrils are an important hallmark of protein misfolding diseases.
  14. [14]
    ThT 101: a primer on the use of thioflavin T to investigate amyloid ...
    This review aims to provide a conceptual instruction manual, covering appropriate considerations and pitfalls related to the use of ThT.
  15. [15]
    Amyloid oligomers: formation and toxicity of Aβ ... - FEBS Press
    Feb 24, 2010 · The high toxicity of low-MW Aβ oligomers is also supported by in vitro studies showing that Aβ dimers are threefold more toxic than monomers, ...
  16. [16]
    Accumulation of protein aggregates induces autolytic programmed ...
    Jul 15, 2019 · These findings demonstrate that accumulation of protein aggregates, including proteasome subunits, eventually cause autolytic PCD in hybrid cells.
  17. [17]
    Measurement of amyloid formation by turbidity assay—seeing ...
    Nov 23, 2016 · Detection of amyloid growth is commonly carried out by measurement of solution turbidity, a low-cost assay procedure based on the intrinsic ...
  18. [18]
    Analysis of Protein Aggregation in Neurodegenerative Disease
    Mar 8, 2013 · Spectrometry methods: (a) Turbidity, oligomers and aggregates with a hydrodynamic radius larger than the wavelength of the incident lights ...<|control11|><|separator|>
  19. [19]
    Sickle Cell Disease—Genetics, Pathophysiology, Clinical ...
    May 7, 2019 · Sickle cell disease (SCD) is a monogenetic disorder due to a single base-pair point mutation in the β-globin gene resulting in the substitution ...
  20. [20]
    Protein aggregation in disease: a role for folding intermediates ...
    For example, in sickle cell disease, the polymerization of sickle hemoglobin into fibrils in the deoxygenating environment of the microvasculature causes ...
  21. [21]
    The threshold for polyglutamine-expansion protein aggregation and ...
    This finding has led to a disease hypothesis that protein aggregation and cellular dysfunction can occur at a threshold of approximately 40 glutamine residues.
  22. [22]
    A CAG repeat threshold for therapeutics targeting somatic instability ...
    May 3, 2024 · The Huntington's disease mutation is a CAG repeat expansion in the huntingtin gene that results in an expanded polyglutamine tract in the huntingtin protein.
  23. [23]
    Myopathy BAG3 Mutations: Protein Aggregation, Stalling Hsp70
    Dec 17, 2018 · Mutations in either the IPV or BAG domain of BAG3 cause a dominant form of myopathy, characterized by protein aggregation in both skeletal and cardiac muscle ...
  24. [24]
    [PDF] Myopathy associated BAG3 mutations lead to protein aggregation ...
    As a con- sequence, the mutant BAG3 proteins become the node for a dominant gain of function causing aggregation of itself, Hsp70, Hsp70 clients and tiered ...
  25. [25]
    Hereditary Transthyretin Amyloidosis - GeneReviews - NCBI - NIH
    Nov 5, 2001 · ATTRv amyloidosis is inherited in an autosomal dominant manner. Each child of an individual who is heterozygous for a TTR pathogenic variant has ...
  26. [26]
    Genetics of β-Amyloid Precursor Protein in Alzheimer's Disease - NIH
    The amyloid hypothesis proposes that AD is caused by altered APP expression or APP-mutation-induced Aβ aggregation, following an imbalance between Aβ production ...
  27. [27]
    Aging: progressive decline in fitness due to the rising deleteriome ...
    Apr 6, 2016 · The theory posits that certain alleles could be selected for and mutations could accumulate in the genomes over evolutionary timescales, if ...
  28. [28]
    Protein aggregation as a paradigm of aging - ScienceDirect
    In aging the accumulation of misfolded and aggregated proteins is increased because of the higher rate of PTMs, more efficient action of toxic agents on the one ...Missing: aspects | Show results with:aspects
  29. [29]
    The central role of transfer RNAs in mistranslation - PMC
    The principal function of tRNAs is to bring amino acid building blocks to the ribosomes for protein synthesis. In the ribosome, tRNAs interact with messenger ...
  30. [30]
    Co-translational protein aggregation and ribosome stalling as a ...
    Feb 12, 2025 · RNA sequencing of inclusion bodies points towards ribosome stalling and nascent chain-mediated aggregation. The enrichment of ribosomal ...
  31. [31]
    The Rqc2/Tae2 subunit of the ribosome-associated quality control ...
    Mar 4, 2016 · These findings uncover a translational stalling-dependent protein aggregation mechanism, and provide evidence that proteins can become ...
  32. [32]
    Disulfide Bonding in Neurodegenerative Misfolding Diseases - PMC
    The role of disulfide bonding in preventing and managing protein misfolding and aggregation is currently under investigation.1. Introduction · 2. Disulfide Bonds In... · 4. Conclusions
  33. [33]
    Pathological Disulfide Bond Crosslinking: Molecular Insights into ...
    Jun 10, 2025 · This review explores pathological disulfide-crosslinking as a key driver of amyloidogenic protein misfolding and aggregation.2.1 Oxidative Stress · 2.2 Er Stress · 3.2 Tdp-43
  34. [34]
    Effect of glycosylation on protein folding: A close look at ... - PNAS
    A thermodynamic analysis showed that the origin of the enhanced protein stabilization by glycosylation is destabilization of the unfolded state.Missing: over- | Show results with:over-
  35. [35]
    Protein Misfolding and Aggregation in Proteinopathies: Causes ...
    Feb 9, 2023 · Factors affecting protein folding include errors in post-translational modifications, protein environment alterations [56], increased ...
  36. [36]
    Global analysis of chaperone effects using a reconstituted cell-free ...
    May 21, 2012 · After the emergence of the chaperone concept, many in vitro studies have shown that chaperones prevent protein aggregation (4, 9).
  37. [37]
    Elimination of a signal sequence-uncleaved form of defective HLA ...
    Nov 6, 2017 · A portion of newly synthesized transmembrane domain proteins tend to fail to assemble correctly in the lumen of the endoplasmic reticulum, ...
  38. [38]
    Weak protein interactions and pH- and temperature-dependent ...
    Temperature dependence of aggregation rates and mechanism(s)​​ Isothermal aggregation was monitored for Fc1 over a broad temperature range, from 30°C to 77°C, as ...
  39. [39]
    Non-Arrhenius Protein Aggregation - PMC - NIH
    This review discusses the non-Arrhenius nature of the temperature dependence of protein aggregation, explores possible causes, and considers inherent hurdles.
  40. [40]
    Causative Links between Protein Aggregation and Oxidative Stress
    Aug 9, 2019 · Compelling evidence supports a tight link between oxidative stress and protein aggregation processes, which are noticeably involved in the development of ...Missing: ischemia | Show results with:ischemia
  41. [41]
    SOD1 aggregation in astrocytes following ischemia/reperfusion injury
    Mutations in SOD1 protein promote the formation of disulfide-reduced monomers, which are prone to forming aggregates. Hence, modulation of disulfide bond ...
  42. [42]
    Multivariate effects of pH, salt, and Zn 2+ ions on Aβ 40 fibrillation
    Dec 13, 2022 · The Aβ aggregation is highly influenced by the pH, which is facilitated during sample preparation where a high pH (app. pH 11) is used to ...Results · Buffer Matrix Design · Tht Fluorescence Assays
  43. [43]
    Urea, but not guanidinium, destabilizes proteins by forming ... - PNAS
    The mechanism by which urea and guanidinium destabilize protein structure is controversial. We tested the possibility that these denaturants form hydrogen bonds ...
  44. [44]
    ER stress causes widespread protein aggregation and prion formation
    ER stress results in widespread aggregation of proteins that are not localized to the ER or are part of the secretory system.
  45. [45]
    Endoplasmic reticulum stress: molecular mechanism and ... - Nature
    Sep 15, 2023 · ER stress is activated when proteostasis is broken with an accumulation of misfolded and unfolded proteins in the ER.
  46. [46]
    Stress Proteins in Aging and Life Span - PMC - NIH
    HSP level decreases in cells with aging and it has been shown that the serum concentration of Hsp70 in a healthy individual decreases with age as well [43].
  47. [47]
    Circulating Heat Shock Protein 70 in Health, Aging and Disease
    Mar 28, 2011 · The present data provide new evidence that serum concentration of Hsp70 decreases with age in a normal population.
  48. [48]
    The aging proteostasis decline: From nematode to human - PMC
    Aging proteostasis decline is a failure to respond to proteotoxic challenges, involving a broad network and a decline in protein clearance functions.
  49. [49]
    Oxidative modifications, mitochondrial dysfunction, and impaired ...
    Jun 5, 2009 · Due to impaired degradation, proteins with oxidative modifications accumulate in the cell, increasing their propensity for aggregation [47,216].
  50. [50]
    Proteasome Augmentation Mitigates Age‐Related Cognitive Decline ...
    Our study reveals a marked reduction in 20S and 26S proteasome activities in aged mice brains, including in the hippocampus, this is driven by reduced ...Missing: rodents | Show results with:rodents
  51. [51]
    Lipofuscin: formation, effects and role of macroautophagy - PMC - NIH
    Here we provide an overview about effect of these aggregates on the proteasomal system, followed by transcription factor activation and loss of cell viability.
  52. [52]
    An Evolutionary Trade-Off between Protein Turnover Rate and ...
    We reasoned that proteins with high turnover rate and thus short lifetime will have, on average, lower risk of misfolding than long-living proteins. Their ...Missing: senescence species
  53. [53]
    Widespread Protein Aggregation as an Inherent Part of Aging in C ...
    Our results suggest that inherent protein aggregation has the potential to influence lifespan and protein aggregation disease. Results. Proteomic Identification ...
  54. [54]
    Asymmetric segregation of protein aggregates is associated ... - PNAS
    Inclusion Bodies Are Formed in Discrete Cellular Positions and Accumulate in Old Poles Under Nonstressing Conditions. To determine the presence and localization ...
  55. [55]
    Challenges Associated With the Formation of Recombinant Protein ...
    In this review, the mechanism and the factors that influence the formation of recombinant protein IBs will be discussed together with their unique features.
  56. [56]
    Protein polarization driven by nucleoid exclusion of DnaK(HSP70)
    May 23, 2018 · Here, we show that the DnaK (HSP70) chaperone controls unipolar localization of the Shigella IpaC type III secretion substrate.
  57. [57]
    Escherichia coli small heat shock protein IbpA plays a role in ... - PNAS
    ... IbpA acts as a “sequestrase” chaperone to recruit aggregation-prone proteins. This titrates the free IbpA away to alleviate the translation suppression.
  58. [58]
  59. [59]
    Protein folding state-dependent sorting at the Golgi apparatus - PMC
    Here we describe how the Golgi apparatus mitigates unfolded proteins and present a strategy for studying the post-ER secretory pathway QC machinery in mammalian ...
  60. [60]
    Aggresomes: A Cellular Response to Misfolded Proteins - PMC
    Intracellular deposition of misfolded protein aggregates into ubiquitin-rich cytoplasmic inclusions is linked to the pathogenesis of many diseases.
  61. [61]
    Nuclear Aggresomes Form by Fusion of PML-associated Aggregates
    Jul 29, 2005 · Cytoplasmic and nuclear aggregates have been shown to be positive for ubiquitin (Johnston et al., 1998; Waelter et al., 2001). In agreement ...
  62. [62]
    The Insoluble Protein Deposit (IPOD) in Yeast - PMC - NIH
    Here, we will review what is known about IPOD composition and the mechanisms of recognition and recruitment of amyloid aggregates to this site in yeast. Finally ...
  63. [63]
    Protein Quality Control: On IPODs and Other JUNQ - ScienceDirect
    Nov 11, 2008 · Therefore, this compartment is called 'juxtanuclear quality control' or JUNQ. ... Quality control compartments in yeast or mammalian cells.
  64. [64]
  65. [65]
    COST1 regulates autophagy to control plant drought tolerance - PNAS
    Mar 13, 2020 · We propose a model in which COST1 negatively regulates autophagy, and aggregation/degradation of COST1 during drought therefore activates ...
  66. [66]
    Can autophagy enhance crop resilience to environmental stress?
    May 29, 2025 · Autophagy facilitates the removal of oxidized and aggregated proteins under various stress conditions, including drought stress [11,90]. Under ...
  67. [67]
    Characterization and Dynamics of Aggresome Formation by a ...
    Overexpression of p50/dynamitin, which causes the dissociation of the dynactin complex, significantly inhibited the formation of aggresomes, suggesting that the ...
  68. [68]
    The disordered P granule protein LAF-1 drives phase separation ...
    May 26, 2015 · We show that the Caenorhabditis elegans protein LAF-1, a DDX3 RNA helicase found in P granules, phase separates into P granule-like droplets in vitro.
  69. [69]
    Polar positioning of phase-separated liquid compartments in cells ...
    P granules are non-membrane-bound RNA-protein compartments that are involved in germline development in C. elegans. They are liquids that condense at one end of ...
  70. [70]
    Challenges in studying the liquid-to-solid phase transitions of ... - NIH
    In this review, we highlight recent biophysical studies which provide new insights into the molecular mechanisms underlying liquid-to-solid (fibril) phase ...
  71. [71]
    Self-propagation of pathogenic protein aggregates in ... - NIH
    The essence of prion disease is a crystallization-like chain reaction by which malformed PrP seeds force naive PrP molecules into a similar pathogenic ...
  72. [72]
    Reversibility of β-Amyloid Self-Assembly: Effects of pH and Added ...
    Aug 6, 2025 · Edwin et al. [70] have used FRAP to study the reversibility of β-amyloid (Aβ) peptide aggregate formation. The Aβ peptide is believed to be the ...
  73. [73]
    Structural mechanisms of chaperone mediated protein disaggregation
    The ClpB/Hsp104 and Hsp70 classes of molecular chaperones use ATP hydrolysis to dissociate protein aggregates and complexes, and to move proteins through ...
  74. [74]
    Metazoan Hsp70 machines use Hsp110 to power protein ...
    Sep 18, 2012 · Hsp40 co‐chaperones stimulate ATP hydrolysis by Hsp70, resulting in an Hsp70 conformation with high affinity for substrate. Nucleotide ...
  75. [75]
    Hsp70 targets Hsp100 chaperones to substrates for protein ...
    Aug 6, 2012 · Disaggregation relies on Hsp70 function and on ATP-dependent threading of aggregated polypeptides through the pore of the Hsp100 AAA+ hexamer.
  76. [76]
    Cryo-EM Structures of the Hsp104 Protein Disaggregase Captured ...
    Jan 2, 2019 · Hsp104 is a ring-forming ATPase that facilitates the disaggregation of amorphous and amyloid-forming protein aggregates. Lee et al. present ...
  77. [77]
    Bacterial and Yeast AAA + Disaggregases ClpB and Hsp104 ...
    We show that protein disaggregation initiated by Hsp70 binding to protein aggregates is not bypassed by artificial ClpB or Hsp104 activation. ClpB and Hsp104 ...
  78. [78]
    Structural basis for substrate gripping and translocation by the ClpB ...
    Jun 3, 2019 · Bacterial ClpB and yeast Hsp104 are homologous Hsp100 protein disaggregases that serve critical functions in proteostasis by solubilizing ...
  79. [79]
    Spiral architecture of the Hsp104 disaggregase reveals the ...
    Hsp104 and its bacterial homolog, ClpB, form large hexameric-ring structures that cooperate with the Hsp70 system to unfold and rescue aggregated protein states ...
  80. [80]
    Small Heat Shock Proteins, Big Impact on Protein Aggregation in ...
    Sep 18, 2019 · Chaperones can be classified as holdases (that bind and hold partially folded protein intermediates to prevent their aggregation), foldases ( ...
  81. [81]
    Small heat shock proteins: Simplicity meets complexity - PMC
    Cellular function. Mechanistically, sHsps bind misfolded or unfolding proteins and prevent them from irreversible, uncontrolled aggregation in an ATP- ...
  82. [82]
    Ubiquitin-like protein conjugation and the ubiquitin–proteasome ...
    Dec 10, 2010 · The ubiquitin–proteasome system (UPS) and ubiquitin-like protein (UBL) conjugation pathways are integral to cellular protein homeostasis.Missing: paper | Show results with:paper
  83. [83]
    Targeted protein degradation: mechanisms, strategies and application
    Apr 4, 2022 · Proteins marked with K48-linked ubiquitin chains are often targeted to proteasome for degradation; in contrast, K63-linked chains do not ...
  84. [84]
    The life cycle of the 26S proteasome: from birth, through regulation ...
    Jul 22, 2016 · The 26S proteasome is a large, ∼2.5 MDa, multi-catalytic ATP-dependent protease complex that serves as the degrading arm of the ubiquitin system.Missing: paper | Show results with:paper
  85. [85]
    Lysosomes as a therapeutic target | Nature Reviews Drug Discovery
    Sep 2, 2019 · Lysosomes have long been known to have a key role in the degradation and recycling of extracellular material via endocytosis and phagocytosis, ...Missing: paper | Show results with:paper
  86. [86]
    Targeting lysosomes in human disease: from basic research to ...
    Nov 8, 2021 · The expression of cathepsins, the important executors of lysosomal degradation, was deregulated in cells of neurodegenerative diseases, and ...
  87. [87]
    The role of protein clearance mechanisms in organismal ageing and ...
    Dec 8, 2014 · Chaperones assure the proper folding of proteins throughout their life cycle and under stress conditions but their activity declines with age ( ...<|separator|>
  88. [88]
    The autophagy receptor p62/SQST-1 promotes proteostasis ... - Nature
    Dec 11, 2019 · Autophagy can degrade cargos with the help of selective autophagy receptors such as p62/SQSTM1, which facilitates the degradation of
  89. [89]
    Ubiquitination and selective autophagy | Cell Death & Differentiation
    Jun 22, 2012 · In this review, we focus on the special role of ubiquitin signals and selective autophagy receptors in sorting a variety of autophagic cargos.
  90. [90]
    Emerging roles of ATG proteins and membrane lipids in ... - Nature
    May 26, 2020 · Autophagosome biogenesis is a dynamic membrane event, which is executed by the sequential function of autophagy-related (ATG) proteins.
  91. [91]
    Molecules and their functions in autophagy - Nature
    Jan 19, 2012 · The conversion of LC3 to LC3-II is thus well-known as a marker of autophagy-induction. However, the increase of LC3-II alone is not enough to ...
  92. [92]
    How lysosomes SNARE autophagosomes - Nature
    Dec 19, 2012 · The SNARE protein STX17 is recruited specifically to completed autophagosomes to mediate fusion with lysosomes.
  93. [93]
    Lysosome biology in autophagy | Cell Discovery - Nature
    Feb 11, 2020 · Autophagosome-lysosome fusion is executed by either of two SNARE complexes: STX17-SNAP29-VAMP7/VAMP8 or STX7-SNAP29-YKT6. SNARE ...
  94. [94]
    The mitophagy pathway and its implications in human diseases
    Aug 16, 2023 · In this review, we summarize the molecular mechanisms of mitophagy ... Atg32 is a mitochondrial protein that confers selectivity during mitophagy.
  95. [95]
    Neuronal autophagy and neurodegenerative diseases - Nature
    Jan 19, 2012 · In this review, we consider both the normal and pathophysiological roles of neuronal autophagy and its potential therapeutic implications for common ...
  96. [96]
    Toxic oligomers of the amyloidogenic HypF-N protein form pores in ...
    Oct 20, 2020 · These results suggest an inherent toxicity of membrane-active aggregates of amyloid-forming proteins to mitochondria, and that targeting of ...
  97. [97]
    Emerging biophysical origins and pathogenic implications ... - Nature
    Mar 26, 2025 · In membrane environments, β-barrel oligomers form pore-like structures that disrupt membrane integrity, causing ion leakage, calcium influx, and ...Oligomer Formation Under The... · Effects Of Llps On β-Barrel... · Amyloid Oligomers And...Missing: gain- | Show results with:gain-
  98. [98]
    Individual aggregates of amyloid beta induce temporary calcium ...
    Aug 24, 2016 · These data are consistent with individual oligomers larger than trimers inducing calcium entry as they cross the cell membrane.Results · Astrocyte Dosing Experiments · Material And Methods
  99. [99]
    Molecular chaperones: from proteostasis to pathogenesis - PMC
    At the cellular level, protein aggregation is counteracted by powerful mechanisms comprising of a cascade of enzymes and chaperones that operate in a ...Missing: overload collapse seminal review<|separator|>
  100. [100]
    Proteostasis collapse, a hallmark of aging, hinders the chaperone ...
    Sep 13, 2019 · Loss of proteostasis and cellular senescence are key hallmarks of aging, but direct cause-effect relationships are not well understood.
  101. [101]
    A Futile Battle? Protein Quality Control and the Stress of Aging
    Jan 22, 2018 · Protein misfolding and aggregation can be a serious threat to cellular homeostasis, and perturbations in cellular proteostasis are seen during ...
  102. [102]
    The NLRP3 Inflammasome: An Overview of Mechanisms of ...
    The NLRP3 inflammasome is a critical component of the innate immune system that mediates caspase-1 activation and the secretion of proinflammatory cytokines.
  103. [103]
    Soluble Oligomers of the Amyloid β-Protein Impair Synaptic ...
    We find that these naturally secreted soluble oligomers -- at picomolar concentrations -- can disrupt hippocampal LTP in slices and in vivo and can also impair ...
  104. [104]
    Soluble Aβ Oligomers Inhibit Long-Term Potentiation through a ...
    May 4, 2011 · Numerous studies report that hippocampal long-term potentiation (LTP) can be inhibited by soluble oligomers of amyloid β-protein (Aβ), but the ...Ltp Inhibition By Soluble... · Soluble Aβ Activates... · Longer Aβ Exposure...
  105. [105]
    Prion-like transmission of protein aggregates in neurodegenerative ...
    Recent studies suggest that these aggregates are capable of crossing cellular membranes and can directly contribute to the propagation of neurodegenerative ...
  106. [106]
    Cellular mechanisms of protein aggregate propagation - PMC
    Seeded aggregation is the process by which protein aggregates induce misfolding of the cognate monomer in vitro. Transcellular propagation is the process by ...
  107. [107]
    Protein aggregation, metals and oxidative stress in ... - Portland Press
    Oct 26, 2005 · Our recent results suggest that ROS are generated during the very early stages of protein aggregation, when protofibrils or soluble oligomers ...
  108. [108]
    Protein aggregation as a cellular response to oxidative ... - PNAS
    Nov 7, 2016 · The labile Fe can catalyze the generation of ROS via a Fenton reaction (7). Given our results showing that the formation of ALIS induced by ...
  109. [109]
    Protein aggregation and neurodegenerative disease - Nature
    Jul 1, 2004 · Covalent modifications of proteins may facilitate aggregation. Sporadic neurodegenerative diseases are generally associated with aging, which ...
  110. [110]
    New Insights into the Molecular Bases of Familial Alzheimer's Disease
    Apr 19, 2020 · In particular, it has been estimated that 15–25% of total AD accounts for familial Alzheimer's disease (FAD) (Figure 1) [5]. Figure 1. Figure 1.
  111. [111]
    The Role of Amyloid PET in Imaging Neurodegenerative Disorders
    Amyloid PET is a crucial tool for the diagnosis of Alzheimer disease, as it allows the noninvasive detection of amyloid plaques.
  112. [112]
    Type 2 Diabetes as a Protein Misfolding Disease - PubMed Central
    Recent evidence suggests that formation of toxic aggregates of the islet amyloid polypeptide (IAPP) might contribute to β-cell dysfunction and disease.
  113. [113]
    Cellular and Molecular Mechanisms of Prion Disease - PMC
    Prion diseases are rapidly progressive, incurable neurodegenerative disorders caused by misfolded, aggregated proteins known as prions, which are uniquely ...
  114. [114]
    AA Amyloidosis: A Contemporary View - PMC - PubMed Central
    Apr 3, 2024 · Amyloid A (AA) amyloidosis is an organ- or life-threatening complication of chronic inflammatory disorders. Here, we review the epidemiology, ...
  115. [115]
    Protein recovery from inclusion bodies of Escherichia coli using mild ...
    Mar 25, 2015 · Inclusion body aggregates are of highly dynamic nature. Besides the recombinant protein, inclusion bodies also contain components from bacterial ...
  116. [116]
    Advances in Escherichia coli-Based Therapeutic Protein Expression
    Proteins overexpressed in E. coli may form insoluble aggregates known as inclusion bodies, requiring specific solubilization and refolding steps, adding ...5. Bioinformatics... · 6. Gene Cloning And Design · 6.1. Ribosomes
  117. [117]
    Shear stress as a driver of degradation for protein-based therapeutics
    Jan 25, 2024 · A product may be sensitive to numerous stability stressors, such as temperature, pH or ionic strength changes, oxidation, light exposure, ...
  118. [118]
    Impact of Cavitation, High Shear Stress and Air/Liquid Interfaces on ...
    Mar 25, 2018 · The reported impact of shear stress on protein aggregation has been contradictory. At high shear rates, the occurrence of cavitation or ...
  119. [119]
    Stabilization challenges and aggregation in protein-based ... - NIH
    Dec 11, 2023 · Alterations in pH can disturb the charge balance within the protein resulting in modifications of its conformation and stability. Additionally, ...
  120. [120]
    Aggregates in Monoclonal Antibody Manufacturing Processes
    In this review it is analyzed how aggregates are formed during monoclonal antibody industrial production, why they have to be removed and the manufacturing ...
  121. [121]
    Protein Aggregation and Immunogenicity of Biotherapeutics - PMC
    A small amount of soluble aggregates, between 5 to 10%, for example, may be acceptable in biologic products, because the belief is that it is generally ...
  122. [122]
    Exploring the Protein Stabilizing Capability of Surfactants Against ...
    The application of surfactants in liquid protein formulation is a common practice to protect proteins from liquid-air interface-induced protein aggregation.
  123. [123]
    Tafamidis Treatment for Patients with Transthyretin Amyloid ...
    Sep 13, 2018 · Methods: In a multicenter, international, double-blind, placebo-controlled, phase 3 trial, we randomly assigned 441 patients with transthyretin ...
  124. [124]
    Thermodynamic and kinetic design principles for amyloid ... - PNAS
    We present here a comprehensive kinetic theory of amyloid-aggregation inhibition that reveals the fundamental thermodynamic and kinetic signatures ...
  125. [125]
    Phase 1–2 Trial of Antisense Oligonucleotide Tofersen for SOD1 ALS
    Jul 8, 2020 · Tofersen is an antisense oligonucleotide that mediates the degradation of superoxide dismutase 1 (SOD1) messenger RNA to reduce SOD1 protein synthesis.
  126. [126]
    Efficacy of Arimoclomol for Amyotrophic Lateral Sclerosis
    Apr 7, 2025 · The results show that arimoclomol was associated with a slightly slower decline in ALSFRS-R scores compared to placebo, with a mean difference of 2.64.
  127. [127]
    Lecanemab in Early Alzheimer's Disease
    Nov 29, 2022 · Lecanemab reduced markers of amyloid in early Alzheimer's disease and resulted in moderately less decline on measures of cognition and function than placebo at ...
  128. [128]
    Treatment with semaglutide, a GLP-1 receptor agonist, improves ...
    Apr 15, 2023 · Semaglutide improved the turnover of islet heparan sulfate proteoglycans, hyaluronan, chondroitin sulfate proteoglycans, and collagens in the islet ECM.Missing: clinical trial
  129. [129]
    An in vivo platform to select and evolve aggregation-resistant proteins
    Apr 14, 2020 · Using this assay as a directed evolution screen, we demonstrate the generation of aggregation resistant scFv sequences when reformatted as IgGs.Results · Protein Aggregation... · MethodsMissing: biomanufacturing | Show results with:biomanufacturing