Abstract
Amyloid fibrils cause organ and tissue dysfunction in numerous severe diseases. Despite the prevalence and severity of amyloidoses, there is still no effective and safe anti-amyloid therapy. This study investigates the impact of cysteine protease cathepsin B (CTSB) on amyloids associated with Alzheimer’s and Parkinson’s diseases, hemodialysis, and lysozyme amyloidosis. We analyzed the effect of CTSB on the size, structure, and proteotoxicity of amyloid fibrils formed from alpha-synuclein, abeta peptide (1-42), insulin, and lysozyme using a combination of spectroscopic, microscopic, electrophoretic, and colorimetric methods. Our comprehensive research revealed a dual effect of CTSB on amyloid fibrils. Firstly, CTSB induced amyloid fragmentation while preserving their ordered morphology, and, secondly, it “loosened” the tertiary structure of amyloids and reduced the regularity of the secondary structure. This dual mechanism of action was universal across fibrils associated with different pathologies, although the disruption efficacy and predominant type of degradation products depended on the amyloids’ structure, size, and clustering. Notably, CTSB-induced irreversible degradation significantly reduced the toxicity for immortalized and primary cell lines of low-clustered fibrils, such as alpha-synuclein amyloids associated with Parkinson’s disease. These findings enhance our understanding of how endogenous CTSB may regulate amyloid content at the molecular level in different neuropathologies. In addition, our results suggest the potential of CTSB as a component of anti-amyloid drugs in combination with agents that enhance the accessibility of proteolytic sites within amyloid clots and reduce these clusters stability.

Similar content being viewed by others
Introduction
Diseases accompanied by the accumulation of ordered protein aggregates, amyloid fibrils, are becoming increasingly common each year. Specifically, about 6.1 million patients suffer from Parkinson’s disease [1,2,3], a neurodegenerative condition marked by the formation of pathogenic amyloids from alpha-synuclein. Alzheimer’s disease, characterized by the accumulation of plaques from abeta peptide (1-42) [4,5,6] in the brain, is even referred to as the “epidemic of the 21st century” with the World Health Organization predicting it to become the most prevalent disease worldwide in the coming decades [7]. This condition has already been identified in 43.8 million individuals, and this number is expected to rise to 152 million by 2050, according to forecasts [8]. These figures highlight the global scale of the problem associated with the formation of pathological amyloids in the body in several dozen severe diseases [9,10,11,12,13,14].
Despite the prevalence and severity of amyloidoses, there is still no effective and safe anti-amyloid therapy available [15, 16]. Currently, one of the promising strategies for treating these diseases is the destruction and clearance of mature amyloid fibrils and plaques [17,18,19,20]. Several new drugs based on this approach are in various stages of development and clinical trials [21,22,23,24,25]. This strategy is rooted in the “amyloid hypothesis”, a longstanding paradigm that attributes the development of Alzheimer’s disease and other neurodegenerative disorders to the accumulation of amyloid plaques. These insoluble deposits are believed to cause cell death and cognitive impairment characteristic of these diseases. For example, studies have shown that amyloid deposits significantly increase the risk of developing amnestic mild cognitive impairment and its progression to Alzheimer’s disease [26]. Furthermore, cerebral amyloid angiopathy, characterized by plaque accumulation on cerebral vessel walls, interacts with pathological accumulation of neuritic amyloid plaques, exacerbating tau pathology and accelerating cognitive decline [27]. Similarly, in the case of primary or light-chain (AL) amyloidosis, the most common type of systemic amyloidosis, the accumulation of insoluble fibrils in tissues leads to organ dysfunction and, ultimately, to fatal outcomes [28, 29].
However, emerging evidence suggests that fibril formation may not be the sole mechanism driving tissue and cellular damage in amyloidoses. For instance, hyper-aggregation of abeta peptide induced by reduced IGF1 signaling was found to ameliorate Alzheimer’s-like symptoms in model mice, rather than exacerbating them [30]. Moreover, some clinical studies have revealed a weak correlation between amyloid plaque burden and the severity of dementia. For instance, some patients with significant plaque deposition, for some reason, do not exhibit dementia symptoms [31]. These findings highlight additional pathogenic factors beyond plaque accumulation. Among these, oligomeric forms of amyloidogenic proteins have gained attention as potentially more toxic than mature fibrils under certain conditions. For example, abeta peptide dimers were shown to exhibit greater toxicity than large aggregates [32]. Similarly, smaller PrP aggregates demonstrated significantly higher toxicity and infectivity compared to larger fibrils [33].
The accumulating data on the cytotoxicity of various protein species challenge the original “amyloid hypothesis” and expand our understanding of amyloidoses pathogenesis. While insoluble deposits remain the primary pathogenic factor in systemic [28, 29] and some localized amyloidoses [34,35,36,37], neurodegenerative diseases associated with amyloids present a more complex scenario. In addition to the direct toxicity of amyloid plaques, these aggregates can also serve as a source from which soluble amyloid forms, including low-molecular-weight oligomers, are released during degradation. These oligomers exhibit comparable, and in some cases even greater, toxicity to neurons [38]. Despite the well-documented toxicity of both mature plaques and these oligomeric intermediates, the development of anti-amyloid drugs often fails to adequately address the analysis of not only the efficiency of mature amyloid clearance but also the cytotoxicity of the low-molecular-weight degradation products.
In this study, we performed such an analysis after cysteine protease cathepsin B (CTSB) treatment, which is being considered as a potential therapeutic agent in several studies [39, 40]. Interest in this enzyme, which participates in the degradation of lysosomal proteins, has arisen because it is expressed in macrophages and multinucleated histiocytic giant cells (MGC) found in close proximity to amyloid deposits in the body [41]. Furthermore, CTSB itself has been detected in the amyloid deposits of patients [41,42,43,44]. Given this fact, the question of whether cells of the mononuclear phagocyte system can destroy amyloids either through phagocytosis or by releasing active proteases, including CTSB, is being actively investigated.
In analyzing the potential use of CTSB as a therapeutic agent with neuroprotective and anti-amyloidogenic properties, its ability to prevent the formation of neurotoxic plaques by various amyloidogenic proteins (abeta peptide, immunoglobulin light chain, serum amyloid A, and alpha-synuclein) as well as to cleave amyloid deposits was noted [40,41,42, 45,46,47,48,49,50]. It is important to emphasize that the current body of evidence regarding the impact of CTSB on mature amyloids is both limited and highly contradictory. The studies available in the literature, using genetic, immunohistochemical and pharmacological approaches, demonstrated the fundamental ability of the enzyme to degrade amyloids formed from alpha-synuclein and immunoglobulin light chain [40, 41, 50, 51]. Furthermore, the ability of CTSB to reduce the amount of abeta peptide fibrillar structures in vitro was demonstrated, and CTSB over-expression was shown to contribute to the reduction of amyloid deposits formation in aged hAPP mice [42], similar to neprilysin or insulin degrading enzyme (IDE) [52]. Some of these studies suggested that this unique ability of CTSB reveals its potential as a therapeutic agent for diseases where controlling amyloid quantity is a crucial aspect of treatment. However, some studies [48, 51] reported that incomplete degradation of amyloids leads to increased nucleation activity and induction of new aggregate formation, which could potentially exacerbate the disease. Consequently, these authors reached the opposite conclusion that inhibiting the effect of CTSB on mature amyloids might serve as a therapeutic strategy. The consequences of the CTSB gene knockout (or knockdown) in neurologic disorder models was also studied. For example, a number of studies in mouse models of Alzheimer’s disease found improved brain function following knockout of the CTSB gene [53]. Similarly, knockdown of the CTSB gene in the nematode Caenorhabditis elegans protected model worms from abeta peptide toxicity [54]. In contrast, the enhanced polyQ-associated proteotoxicity observed upon CTSB knockdown in model worms led to the suggestion that inhibition of CTSB activity should be approached with caution [54].
The aim of current study was to clarify the conflicting views on the effect of CTSB on mature amyloid fibrils by filling the gaps in the understanding of molecular basis of this process and identifying the factors determining the disruption efficiency. For this purpose, we for the first time analyzed in vitro the mechanism of CTSB-induced amyloid degradation, as well as the structure and properties of the resulting degradation products by various biochemical and physicochemical methods including special procedure of sample preparation. To ensure the universality of the observed effects and to identify patterns in the processes observed, we used amyloids formed from several proteins and peptides, significantly differing in structure and properties. Specifically, we examined the degradation of amyloids accumulating in the bodies of patients with Alzheimer’s disease (formed from abeta peptide (1-42)) [55, 56], Parkinson’s disease (formed from alpha-synuclein) [57,58,59], hemodialysis amyloidosis in acute renal failure (formed from beta-2-microglobulin) [60, 61], and systemic lysozyme amyloidosis (formed from lysozyme) [62, 63].
Results
CTSB-induced degradation of model amyloids
To refine research techniques and determine the duration of the experiment, we first analyzed the impact of CTSB on amyloid fibrils formed from lysozyme, which serves as a convenient and accessible model object for studying amyloids in vitro. To visualize the degradation process of mature amyloids and analyze the dynamics of this process, aliquots were collected at 2, 6, and 24 h of incubation (the time after which, according to literature, CTSB completely loses its activity [64]) in the presence of the protease, and then the enzyme was inactivated [65] (see the “Materials and Methods” section). The selected samples were visualized using transmission electron microscopy (TEM) (Fig. 1A). The presence of monomeric or oligomeric protein forms that could not be visualized using TEM was assessed using pseudo-native SDS-PAGE (Fig. 1B, C). The efficiency of amyloid degradation was determined by the proportion of the degraded low-molecular-weight protein fraction using absorption spectroscopy of the supernatants obtained after centrifuging the selected samples (Fig. 1D).
A Visualization of amyloids using transmission electron microscopy before and at different time intervals (2, 6, and 24 h, indicated above the panels) after enzyme addition. Scale bars are 5 μm. Assessment of monomeric/oligomeric protein content in the sample using pseudo-native (B) 17% and (C) 8% SDS-PAGE. Monomeric protein (M) at a concentration equal to that of amyloids was loaded in lane 5 as a control. Low-molecular-weight (LMW) and high-molecular-weight (HMW) marker proteins were loaded in the last lanes. Their molecular weights are indicated to the right of the gels. D Quantification of the degraded fraction in the supernatant using absorption spectroscopy after centrifugation of samples collected at different time intervals (2, 6, 24, and 120 h) after enzyme addition. Values are calculated relative to the concentration of intact amyloid fibrils. Data are mean ± SD (n = 5).
It was shown that under the action of CTSB, a gradual degradation of the studied amyloids occurred (Fig. 1D). We observed a significant reduction in their fiber length (Fig. S1A) and loss of fibrillar morphology of some amyloid fragments 24 h after the start of the experiment (Fig. 1A). We assessed which protein fractions were present in samples before and after CTSB exposure using SDS-PAGE under pseudo-native conditions (Fig. 1B, C) not causing dissociation of aggregates in the sample under study. The sample with intact lysozyme amyloid was found to be virtually free of protein monomer (Fig. 1B, 0 h) of molecular weight 14.3 kDa (monomeric protein at a concentration equal to the amyloid concentration is shown in lane «M» as a control). The intact sample also lacked protein oligomers and small aggregates with molecular weight less than 220 kDa. This confirms that all of the protein in the intact sample is incorporated into substantially larger amyloid fibrils. Given the CTSB-induced degradation of amyloid (Fig. 1А), the appearance of oligomeric fractions of lysozyme and monomeric protein in the samples over time could be expected. However, we did not find an increase in the level of lysozyme monomers or oligomers with a molecular weight of less than 220 kDa in the samples (Fig. 1B, C, lanes «2 h», «6 h», «24 h»), indicating the relatively large size of the degradation products. It was noted that even 24 h after the start of the treatment, amyloid fibrils were still detected in the sample (Fig. 1A).
To ensure that 24 h is sufficient time for the protease’s action on the amyloids and for the system of degradation products to reach equilibrium, we extended the incubation time of the samples to 5 days. It was found that the morphology and size of the fibril degradation products, as well as the proportion of the degraded fraction (which amounted to about 40%), were the same after 24 h and 5 days of protease treatment (Fig. 1A, D). The sufficiency of 24-h exposure for degradation of other tested amyloids was confirmed on the example of amyloid formed from alpha-synuclein (Fig. S2 and S1B). As a result, the duration of the experiments on the degradation of other amyloids by CTSB was limited to 24 h.
Analysis of the morphology and stability of amyloid degradation products formed from various proteins after CTSB treatment
The degradation products of amyloids formed from lysozyme, abeta peptide (1-42), alpha-synuclein, and beta-2-microglobulin, after 24 h of their incubation in the presence of CTSB, were visualized using transmission electron microscopy (TEM, Fig. 2A). To assess changes in the size of large fibrillary clusters, confocal laser scanning microscopy was used in the presence of the amyloid-specific probe thioflavin T (Fig. 2B). Noted that intact alpha-synuclein amyloids were poorly detectable by confocal laser scanning microscopy, suggesting very few large fibrillar clusters in this sample. It was found that CTSB treatment caused the destruction of all studied amyloids. We observed a noticeable decrease in the size of amyloid clusters (Fig. S1C) and fragmentation of fibrils (shown by blue arrows) and disruption of the ordered morphology of fibrils (shown by red arrows).
Data are presented for amyloids formed from lysozyme (Lys), abeta peptide (1-42) (Ab42), beta-2-microglobulin (b2m), and alpha-synuclein (a-syn) (indicated above the panels) before (control) and 24 h after (+CTSB) CTSB action. A Transmission electron microscopy images of the most characteristic morphology of objects in the samples. Scale bars are 0.5 μm. The Insets show images of minor protein fractions in the samples. Scale bars are 0.2 μm. The arrows indicate two different types of degradation products: amyloid fragments with intact fibrillar morphology (blue arrows) and aggregates with disrupted morphology (red arrows). B Confocal microscopy images of the samples in the presence of ThT. Scale bars are 15 μm.
We also measured the turbidity and Rayleigh light scattering (RLS) of the samples (Fig. 3A, B). A decrease in these parameters as a result of protease action was observed for all amyloid fibrils, confirming the decrease in the quantity/size of the studied aggregates after treatment. However, the efficiency of amyloid degradation was found to vary. The most pronounced reduction in turbidity and RLS was observed in the case of amyloid fibrils formed from alpha-synuclein (by 70-85%), and the least in the case of amyloid fibrils formed from lysozyme (7–15%).
Data are presented for amyloids formed from lysozyme (Lys), abeta peptide (1-42) (Ab42), beta-2-microglobulin (b2m), and alpha-synuclein (a-syn) before (blue bars) and 24 h after (red bars) CTSB action. (A) Turbidity, (B) Rayleigh light scattering (RLS), (C) integrated intensity of intrinsic UV fluorescence, λex = 280 nm for abeta peptide (1-42) and alpha-synuclein amyloids, λex = 295 nm for lysozyme and beta-2-microglobulin amyloids, (D) parameter A, λex = 295 nm, (E) fluorescence anisotropy (r), λex = 295 nm, λem = 365 nm. In (A–C), values for samples of amyloids with CTSB are normalized to the values for samples of the same intact amyloids. Data are mean ± SD (n = 5). (F, G) Far-UV CD spectra of amyloid fibrils before (Lys, Ab42, b2m, a-syn) and 24 h after (Lys + CTSB, Ab42 + CTSB, b2m + CTSB, a-syn + CTSB) enzyme action.
To confirm the assumption of structural transformations of amyloids in the presence of CTSB, inferred from TEM results, intrinsic UV fluorescence spectra were determined and the value of the integral fluorescence intensity of the samples was calculated (Fig. 3C). Additionally, for fibrils formed from tryptophan-containing proteins, values of fluorescence anisotropy and the parameter A (a characteristic defined as the ratio of fluorescence intensity at registration wavelengths of 320 and 365 nm, sensitive to the microenvironment properties of tryptophan residues) were also assessed (Fig. 3D, E). Notably, the intensity of intrinsic UV fluorescence increased in samples with amyloids formed from abeta peptide (1-42), alpha-synuclein, and beta-2-microglobulin after enzyme treatment, indicating a loosening of the protein environment (and the removal of its quenching effect on fluorescence) of aromatic residues. Concurrently, there was also detected a change in parameter A for amyloids formed from tryptophan-containing proteins: an increase in this characteristic for beta-2-microglobulin amyloids and a slight decrease for lysozyme amyloids, as well as a decrease in fluorescence anisotropy for beta-2-microglobulin amyloids. Collectively, these results indicate a change in the protein environment of residues contributing to the intrinsic fluorescence of amyloids, which is consistent with the hypothesis of fibril structure disruption upon protease action.
Measuring the circular dichroism (CD) spectra of samples in the far UV region allowed for the assessment of changes in the structural properties of aggregates under the action of CTSB (Fig. 3F, G). All fibrils analyzed in current study display a typical negative band at about 217–225 nm, depending on the fibril morphology, confirming their enrichment in beta-sheet content. For most amyloids, we observed a change in the shape and intensity of the CD spectra after CTSB treatment (Fig. 3F). In particular, after protease action on fibrils of abeta peptide (1-42), alpha-synuclein, and beta-2-microglobulin, the minimum around 220 nm, characteristic of the beta-sheet structure [66] became less pronounced. This confirms the assumption of a decrease in the orderliness of the structure of amyloid fragments, made based on TEM data. However, we did not observe a noticeable change in the CD spectra of lysozyme amyloids after enzyme treatment (Fig. 3G). It can be assumed that changes in the structure of these amyloids cannot be detected by the CD spectroscopy method, both because of their negligibility (which is consistent with changes in intrinsic fluorescence characteristics).
Studying the interaction of fibrils and their degradation products with the amyloid-specific probe thioflavin T
Along with fibrillary morphology and high content of beta-sheet structure [67], the presence of amyloids in the sample is confirmed by the tinctorial properties evaluated by binding of amyloid-specific probe thioflavin T (ThT) [68]. So, this dye can be effectively used for analyzing fibrils degradation processes [69,70,71,72]. The use of ThT for this task is rationalized because the dye specifically incorporates into the grooves formed by the side chains of amino acids of the beta-strands of the fibril, along the long axis of its fiber perpendicular to the beta-sheets (4-5 stacked beta-strands are required for binding 1 molecule of ThT) [73, 74]. The dye does not interact with monomeric proteins in globular, unfolded, or partially folded states, with oligomers, or with amorphous protein aggregates. That means the interaction of ThT with the beta-sheet structure of amyloids is highly specific, and therefore, information about the quantity of bound dye molecules can allow for the assessment of changes in the number of beta-strands in amyloid fibers during their degradation.
Accurate estimation of the amount (concentration) of ThT bound to fibrils requires the separation of the absorption spectra of the free and fibril-bound dye fractions present in the sample. To address this task, we used a special approach, based on preparing samples via equilibrium microdialysis [75, 76] (Fig. 4A). The essence of this approach lies in introducing amyloids (or their degradation products) suspension into one chamber of the equilibrium microdialysis device and ThT solution into another. Since the chambers are separated by a membrane permeable to the dye but impermeable to the fibrils, after equilibration the system the concentration of free ThT molecules in the chambers becomes equal. However, the total concentration of the dye in the chamber with fibrils (or their degradation products) exceeds the dye concentration in the second chamber by the concentration of ThT molecules bound to undegraded amyloid fibers or their fragments with intact structure (Fig. 4A). Thus, the differential absorption spectrum of the dye in these chambers (adjusted for light scattering of fibrils) represents the spectrum of the dye bound to fibrils, and its amplitude decreases upon amyloid degradation (shown for lysozyme amyloids in Fig. 4B, C).
A Equilibrium microdialysis device consisting of two chambers (chamber 1 and chamber 2) of equal volume, separated by a membrane permeable to the fluorescent probe thioflavin T (ThT) but impermeable to protein aggregates and oligomers. In the experiments, amyloids before treatment (control, left) or amyloid degradation products after CTSB action (+CTSB, right) were placed in chamber 1, and a dye solution in the same solvent was placed in the chamber 2. Measurements were taken after the system reached equilibrium and free dye concentration in the chambers became equal (shown at the A). Determination of the absorption spectrum of ThT bound to lysozyme amyloid fibrils (B) before and (C) after CTSB treatment, using samples prepared by equilibrium microdialysis. After reaching equilibrium, the absorption spectrum in chamber 2 (ch2) represented the absorption spectrum of free ThT, while the absorption spectrum of the solution in the chamber 1 (ch1) represented the total absorption spectrum of free ThT and dye bound to fibrils (against a background of “apparent absorption” caused by fibrils light scattering (scatt), which was accounted by a standard procedure (ch1-scatt)). This approach allowed us to obtain solutions of the sample and the reference, whose difference spectrum (ch1-scatt)-ch2) was determined by absorption spectroscopy. This spectrum represents the absorption spectrum of ThT incorporated into the lysozyme fibrils in samples before or after CTSB addition. Similar experiments were performed for all amyloids tested. D Absorbance (at the spectral maximum) of ThT (AThT) bound to fibrils formed from lysozyme (Lys), abeta peptide (1-42) (Ab42), beta-2-microglobulin (b2m), and alpha-synuclein (a-syn) before (blue bars) and after (red bars) CTSB action, determined using samples prepared by equilibrium microdialysis. Values for samples of amyloids with CTSB are normalized to the values for samples of the same intact amyloids. Data are mean ± SD (n = 5).
The reduction in absorption (and thus, concentration) of ThT molecules bound to the studied amyloids before and after their degradation, indicating a decrease in the content of ordered beta-strands in the amyloid degradation products, is presented in Fig. 4D. The most significant decrease in ThT absorption was observed in the case of amyloids formed from alpha-synuclein (over 70%), while the least significant was in the case of amyloids formed from lysozyme and abeta peptide (1-42) (30-40%), confirming the varying efficiency of their degradation.
Analysis of the impact of amyloids before and after their degradation on the viability of immortalized and primary cell lines
The assessment of the impact on cells of amyloid degradation products after CTSB treatment was conducted using a colorimetric MTT assay. To obtain a comprehensive picture of the cytotoxicity of the studied amyloids and their degradation products during the experiments, several human cell lines were used. Specifically, the study subjects were human embryonic kidney cell lines (Hek-293), epithelioid carcinoma of the cervix cell lines (HeLa TK-), glioblastoma (T98G), and dermal fibroblasts (DF1). The cells were incubated in the presence of amyloids and their degradation products for 24 h.
The results of our studies showed (Fig. 5) that amyloids formed from abeta peptide (1-42), alpha-synuclein, lysozyme, and beta-2-microglobulin reduced the metabolic activity of all studied cell lines, which can be considered as a quantitative assessment of amyloid-induced stress in cell culture. The patterns observed in tumor cells (changes in cytotoxicity of amyloids before and after their degradation) were shown to be well reproducible in the primary line of dermal fibroblasts (Fig. 5D).
Relative metabolic activity of (A) human embryonic kidney (Hek-293), (B) epithelioid cervix carcinoma (HeLa TK-), (C) glioblastoma (T98G), and (D) dermal fibroblast (DF1) cell lines after 24 h of exposure to mature amyloids formed from lysozyme (Lys), abeta peptide (1-42) (Ab42), beta-2-microglobulin (b2m), and alpha-synuclein (a-syn) (blue bars) and their degradation products (red bars) generated by the proteolytic enzyme CTSB (Lys + CTSB, Ab42 + CTSB, b2m + CTSB, a-syn + CTSB). In experiments with lysozyme and beta-2-microglobulin amyloids values are normalized to the metabolic activity of control cells to which the target proteins were added in the same buffer and at the same concentration as their corresponding fibrils. In experiments with abeta peptide (1-42) and alpha-synuclein, the cells treated with experimental buffer solution were used as controls. Data are illustrated as the mean (±SD). An unpaired t-test was performed to validate the statistical significance. ***p < 0.0001.
Our findings indicate a general level of cell line sensitivity to stress induced by extracellular amyloid aggregates. It turned out that the viability of cells in the presence of fibrils was determined not only by amyloid properties but also by the characteristics of the cell lines. For instance, the glioblastoma line (T98G) demonstrated the least sensitivity to all studied amyloids and their degradation products, with the metabolic activity of its cells decreasing by about 30% compared to control cells under the action of all studied amyloids (Fig. 5C). The highest sensitivity was found in human embryonic kidney cells (Hek-293), whose metabolic activity in the presence of some amyloids decreased by almost 70% compared to control samples (Fig. 5A). Interestingly, the sensitivity of the primary dermal fibroblast line (DF1) was comparable to the sensitivity of the Hek-293 line (Fig. 5D), both regarding amyloids associated with neurodegenerative diseases and amyloids leading to local and systemic forms of amyloidosis.
It was found that the impact of intact fibrils formed from lysozyme, beta-2-microglobulin, and abeta peptide (1-42) on the viability of cell lines was comparable to that of the degradation products of these amyloids (Fig. 5). Meanwhile, the cytotoxicity of fibrils formed from alpha-synuclein significantly decreased after their degradation by the action of CTSB. This effect was observed for all studied cell lines. In particular, when cells of the HeLa TK- line were exposed to the degradation products of amyloids formed from alpha-synuclein, their viability was almost completely restored to the level of intact cells (Fig. 5B).
Discussion
Factors determining the mechanism and efficiency of CTSB action on amyloid fibrils
The experimental data obtained allowed us to establish that CTSB-induced proteolysis of proteins in the composition of fibrils leads to the fragmentation of these aggregates (Figs. 1, 2). This means that the action of the enzyme on amyloid-forming proteins results in the disruption of hydrogen bonds between the beta-strands of the amyloid fiber. However, the mechanism of amyloid degradation under the action of CTSB appears to be dual. Along with fragmentation, the loss of the ordered structure of fibril clusters and fragments was observed (Figs. 1, 2), implying that CTSB can also causes destabilization of intramolecular contacts (hydrophobic, ionic, and/or covalent). None of the fibrils studied appeared to undergo complete dissociation into monomeric subunits. The degradation products after CTSB action predominantly consisted of fragments of the amyloid fiber varying in size with intact beta-strands, and protein aggregates of different sizes with a “loosened” tertiary structure and a less defined secondary structure. No reassembly of the fibrils was observed (as is the case with some other factors that disrupt amyloids) following termination of proteolytic enzyme action. Thus, CTSB-induced degradation of amyloids is irreversible.
It was found that despite a similar dual mechanism of enzyme influence on different amyloid fibrils, the effectiveness of this action noticeably varies. The most efficient degradation was observed in the case of aggregates formed from alpha-synuclein (Figs. 2–4). These amyloids predominantly consist of thin individual fibers which, in rare cases, are capable of interacting with each other, forming a small number of «loose» clusters. Meanwhile, amyloids formed from lysozyme and abeta peptide (1-42), which have a larger diameter and/or are assembled into large number of dense clusters, proved to be the most resistant to the effect and did not change their size and morphology as significantly. This means that the efficacy of the fibrils degrading likely depends on the accessibility of CTSB binding sites. In particular, the enzyme cannot act on protein sites located within a dense/thick amyloid fiber and on proteins located deep within dense fibrillar clusters, resulting only in partial degradation of such amyloids at the periphery or in the least shielded areas from the action.
Thus, considering the results obtained, it can be hypothesized that in the case of CTSB application in anti-amyloid therapy, the enzyme will exhibit a universal dual mechanism of action on fibrils regardless of the pathology and type of amyloids causing it. However, the properties of these amyloids (structure, size, ability to form dense clusters, etc.) will largely determine the effectiveness of the degrading action as well as predominant type of degradation products.
The capacity of CTSB to reduce the cytotoxicity of amyloid fibrils
A survey of the available literature indicates that exposure to a variety of exogenous factors, particularly proteolytic enzymes such as trypsin and alpha-B-crystallin, can result in elevated amyloid toxicity towards cells [70, 71, 77]. This heightened toxicity is hypothesized to arise from either an increase in the affinity of amyloids degradation products for the cell membrane, leading to its excessive destabilization, or conversely, addition stabilization. However, our evaluation of the cytotoxic effects of amyloid degradation products formed from various proteins (lysozyme, beta-2-microglobulin, and abeta peptide (1-42), associated with lysozyme and hemodialysis amyloidosis as well as Alzheimer’s disease, respectively) suggests that CTSB can degrade amyloid fibrils without forming low-molecular-weight products with higher cytotoxicity (Fig. 5). Moreover, this enzyme not only reduces the size of aggregates (thus facilitating their further degradation) but, in some cases, also alleviates cell stress induced by amyloids. Specifically, for all cell lines examined (T98G, Hek-293, DF1, and Hela), the toxicity of degradation products of alpha-synuclein amyloids, associated with Parkinson’s disease, was significantly lower compared to intact fibrils (Fig. 5). This is consistent with literature data showing the positive impact of this enzyme on the pathogenesis of amyloidoses [40, 42, 50]. We believe that the significant reduction in cytotoxicity specifically for alpha-synuclein fibrils is attributed to the most efficient degradation of these amyloids, leading both to effective fragmentation and destruction of the ordered fibrillar structure. As previously noted, this is likely related to the thin structure and low tendency of these fibrils to form dense clusters, resulting in higher accessibility of proteolytic cleavage sites. Therefore, CTSB and other factors with similar mechanisms of action can be considered potential components of anti-amyloid therapeutic agents, leading to effective degradation and reduced cytotoxicity of degradation products, provided that amyloid plaques are pre-declustered.
“Pitfalls” and advantages of using CTSB as an anti-amyloid agent
Despite our findings indicating no increase and, in some cases, a significant decrease, in amyloid fibril cytotoxicity following CTSB exposure, suggesting effective degradation, the observed mechanism of degradation suggests potential “side effects” of such treatment. We demonstrated that one of the degradation products of amyloids under CTSB action are fibril fragments with a structure identical to intact amyloids. These short fibrils and oligomers can serve as “seeds” for the formation of new amyloid fibrils (i.e., they can accelerate fibrillogenesis by increasing the number of “free sticky ends”) and can also rapidly spread between cells. For instance, it has been previously shown that the degradation of amyloids into fibrillar fragments can be a critical step in the propagation of prions and prion-like aggregates in yeast [78, 79]. Similar mechanisms are likely to occur in mammalian cells during amyloid degradation [80]. It can be speculated that such a CTSB-induced mechanism of alpha-synuclein amyloid degradation accounts for the increased nucleation activity and induction of intracellular alpha-synuclein aggregate formation observed in studies [48, 51]. However, it is important to note that other studies emphasize the positive effect of CTSB on the pathogenesis of amyloid-related diseases [40, 42, 50].
According to our results, we hypothesize that the conflicting data on the role of CTSB in amyloidoses may be attributed to the destruction mechanism duality. We consider by this enzyme’s ability to trigger two different processes: 1) accelerating fibrillogenesis through fibril fragmentation (a negative effect), and 2) reducing the cytotoxic and seeding activity of amyloids by “disordering” their structure (a positive effect). Thus, the observed change in amyloid pathogenicity will largely depend on which of these processes prevails. If amyloids are highly clustered (as observed in the case of lysozyme amyloids in our experiment) and resistant to CTSB degradation, their fragmentation will probably be the predominant outcome. This process would generate smaller clusters without substantially impacting cytotoxicity but would, however, facilitate a more rapid rate of amyloid formation and propagation between cells and tissues. In contrast, when amyloids present as thin, individual fibers, that do not form dense clusters, as observed with the alpha-synuclein amyloids in our study, or have undergone a preliminary declustering process, the action of CTSB is expected be more effective. This enhanced action would lead not only to a reduction in aggregate size but also to a disruption of the fibrillar structure itself. In this scenario, degradation products will not act as seeds for fibrillogenesis and will have low cytotoxicity for cells. Thus, our results suggest that CTSB could become a component of effective and safe anti-amyloid drugs, in combination with other factors that enhance the accessibility of proteolytic sites and reduce the stability of amyloid clusters.
Conclusions
This study focused on evaluating the degradation potential of CTSB on amyloid fibrils with varying clustering tendencies, associated with Alzheimer’s and Parkinson’s diseases, hemodialysis, and lysozyme amyloidosis. We demonstrated, for the first time, the ability of CTSB to induce both the disruption of amyloid structures and their fragmentation, with corresponding changes in amyloid cytotoxicity depending on which process predominated (Fig. 6). These findings significantly advance our understanding of how endogenous CTSB may regulate amyloid accumulation at the molecular level across diverse neuropathologies. Furthermore, our results indicate the potential of CTSB as a therapeutic component in anti-amyloid drug strategies, particularly when used in combination with agents that enhance the accessibility of proteolytic sites within amyloid aggregates and destabilize dense fibrillar clusters.
Our study shows that CTSB induces both disruption of the structure and fragmentation of amyloids. The change in amyloid cytotoxicity depends on which process prevails. For highly clustered amyloids resistant to CTSB degradation, fragmentation is likely the predominant outcome (top). This process generates smaller clusters and fragments with intact fibrillar structure without significantly impacting cytotoxicity. In contrast, for amyloids present as thin individual fibers or those that have undergone preliminary declustering, the action of CTSB is expected to be more effective (bottom). This enhanced action leads to a reduction in aggregate size and disruption of the fibrillar structure itself. In this scenario, degradation products have reduced toxicity for cells and do not act as seeds for fibrillogenesis like non-disrupted fragments. Thus, CTSB could be a key component in safe, effective anti-amyloid drugs, combined with factors enhancing proteolytic site accessibility and cluster stability reduction.
Materials and methods
Materials
Fluorescent dye thioflavin T (ThT) “UltraPure Grade” from AnaSpec (USA), 1,1,1,3,3,3-Hexafluoro-2-propanol (HFIP), 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), and buffer components from Sigma (USA) were used without additional purification. Reagents for cell cultivation, including DMEM medium (glucose 4.5 g/l), fetal bovine serum (FBS), and 0.25% Trypsin-EDTA were acquired from Gibco (Thermo Fisher Scientific, USA). Culture flasks and 96-well plates (flat bottom) were purchased from Corning (USA).
The recombinant human beta-2-microglobulin was expressed, purified using the protocol [81], human abeta peptide (1-42) was acquired from GL Biochem (China), lysozyme from chicken egg white was acquired from Sigma-Aldrich (USA), cathepsin B was acquired from Merck (Germany).
The recombinant human alpha-synuclein was expressed in E. coli BL21(DE3) star cells transformed with the pT7-7 plasmid encoding for the target protein. Cells were cultivated in Terrific Broth (TB) medium supplied with 100 μg/ml of ampicillin. Expression of the target protein was induced by 0.5 mM of isopropyl beta-D-1-thiogalactopyranoside (IPTG) for 20 h at 18 °C. The cells harvested by centrifugation (5000 rpm, 30 min, 4 °C) were frozen/thawed, resuspended in 10 mM TrisHCl buffer, pH 8.0, 1 mM EDTA, 1 mM protease inhibitor phenylmethylsulfonyl fluoride (PMSF) and undergone to hyperosmotic shock [82]. The fraction of target protein was collected as supernatant by centrifugation for 30 min at 5000 rpm. Further purification of alpha-synuclein was achieved by thermo-precipitation of impurity proteins (boiling at 100 °C for 15 min, cooling at 4 °C for 5 min, followed by centrifugation at 12000 rpm for 30 min at 4 °C). Next, the target protein was precipitated by 40% ammonium sulfate. Final purification of alpha-synuclein was carried out with Mono-Q 5/50 GL (GE Healthcare, Danderyd, Sweden). The purified alpha-synuclein were dialyzed against 10 mM TrisHCl buffer, pH7.6, 25мМ NaCl and stored at −80 °C.
Amyloid fibrils preparation and their treatment with CTSB
Abeta peptide (1-42) and alpha-synuclein in final concentration 1 mg/ml were solubilized in 50% organic solvent 1,1,1,3,3,3-Hexafluoro-2-propanol (HFIP) and incubated for 7 days [83,84,85]. Following this period, the HFIP was slowly removed through evaporation with a stream of nitrogen. Then volume of the sample was adjusted with distilled water to the initial one, and the samples were incubated for an additional week. Lysozyme in concentration 2 mg/ml was incubated in 100 mM KH2PO4-NaOH buffer (pH 7) in the presence of 3 М guanidine hydrochloride (GdnHCl) at 57 °C for 2 days [86]. For beta-2-microglobulin amyloid fibrils preparation, isolated and purified protein in concentration 1 mg/ml was incubated in 100 mM Gly-HCl buffer (pH 2) at 37 °C for 7 days [81]. All amyloid fibrils were prepared at constant agitation (500 rpm).
The presence of amyloid fibrils in the samples was confirmed by transmission electron microscopy. Following the formation of mature amyloid fibrils for performing experiments protein aggregates were dialyzed against Milli-Q water. The fibril stability and the retention of their characteristics for at least the time during which the experiment with cathepsin B was carried out were controlled.
Pre-activated in 50 mM sodium acetate buffer (pH 5)/1 mM ethylenediaminetetraacetic acid (EDTA) with 1 mM dithiothreitol (DTT) according to the manufacturing protocol, cathepsin B was added to the amyloid fibrils in a 1/50 ratio. To inactivate cathepsin B samples were incubated at 57 °C for 10 min [65]. Fibrils obtained using highly reproducible protocols were subjected to protease treatment multiple times with repeatable effects. All samples were included in the analysis.
Transmission electron microscopy
A transmission electron microscope Libra 120 (Carl Zeiss, Germany) was used to produce the micrographs of amyloid fibrils and products of their degradation. Samples were put on the copper grids coated with formvar/carbon films (Electron Microscopy Sciences, USA) and stained by a 1% aqueous solution of uranyl acetate. The length of amyloid fibrils and their fragments was estimated by the ImageJ program.
Confocal microscopy
Confocal laser scanning microscope Olympus FV 3000 (Olympus, Japan) and oil immersion objective with a 60× magnification, the numerical aperture (NA) of 1.42, and laser with excitation line of 405 nm were applied to visualize the ThT-stained amyloid samples. The size of amyloid fibril clusters was estimated as the area in the ImageJ program.
Spectral measurements
A U-3900H spectrophotometer (Hitachi, Japan) was applied to collect the absorption spectra of the samples. The absorption spectra of amyloid fibrils and mixtures of fibrils with ThT were corrected by the light scattering according to the standard procedure [87]. The size of aggregates was characterized by turbidity recorded at 530 nm.
Fluorescence spectra of the samples were obtained through the Cary Eclipse spectrofluorimeter (Varian, Australia). Intrinsic fluorescence of tryptophan and tyrosine residues of amyloid-forming proteins was recorded using 295 and 280 nm excitation wavelengths. Using parameter A, that is ratio of the fluorescence intensities at the emission wavelengths of 320 and 365 nm, the change of the position and shape of the fibrils fluorescence spectra was analyzed. Rayleigh light scattering (RLS) was determined using the same wavelength of excitation and registration (530 nm). The anisotropy of tryptophan fluorescence was calculated using the vertical and horizontal components of the fluorescence intensity excited by vertically polarized light and recorded at the wavelength of 365 nm. ThT fluorescence was excited at 440 nm. All recorded values of fluorescence intensity were adjusted for the primary inner filter effect [88].
For the measurement of circular dichroism (CD) spectra, a J-810 spectropolarimeter (Jasco, Japan) was used. The far-UV CD spectra were recorded in the range of 190–260 nm with a step of 0.2 nm and standard instrument sensitivity using a 1 mm path length cell. To prevent the formation of optically active ozone, the measurements were carried out under continuous nitrogen supply, which ensured efficient removal of oxygen from the optical path of the instrument. Aqueous solutions (Milli-Q water) of amyloid fibrils were used to record optical activity. Milli-Q water is optically transparent up to 180 nm at 1 mm optical path. To achieve an optimal signal-to-noise ratio in circular dichroism measurements and to reduce the light scattering of the samples, we used a relatively low sample concentration (0.15–0.2 mg/mL). Spectra were recorded at a scanning speed of 10 nm/min and a response time of 8 s to increase the signal-to-noise ratio as well. Three scans of CD spectra were averaged and corrected for baseline drift using the signal of Milli-Q water.
Equilibrium microdialysis
The tested solutions of ThT with amyloid fibrils were prepared by equilibrium microdialysis using a Harvard Apparatus/Amika (USA) device [75, 76]. Equilibrium microdialysis was performed at fibrils (or their degradation products) and ThT concentrations of 0.5 mg/ml and 30 μM, respectively. The concentration of free ThT was quantified using molar extinction coefficient at 412 nm of 31600 M−1 cm−1 [89].
MTT assay
Human embryonic kidney (Hek-293), cervical epithelioid carcinoma (HeLa TK-), glioblastoma (T98G) and dermal fibroblast (DF1) cells were used. Cells were obtained from the shared research facility “Vertebrate cell culture collection”. All cell lines were authenticated by STR profiling and were checked for mycoplasma infections. The metabolic activity was evaluated in 24 h after the addition of the analyzed amyloids and their degradation products at 0.003 mg/ml concentration, using an MTT inhibition assay according to a standard protocol as described earlier [90, 91].
Pseudo-native sodium dodecyl sulfate (SDS) gel electrophoresis
The size of amyloid fibril degradation products after CTSB action was analyzed by sodium dodecyl-sulfate polyacrylamide gel electrophoresis (SDS-PAGE) analysis on 17% and 8% polyacrylamide gel (0.375 M Tris HCl, pH 8.8, 0.1% SDS). Samples were loaded on the gel in a buffer containing 0.0625 M Tris HCl, pH 6.8, 1% SDS, 10% glycerol, and 0.002% bromophenol blue without boiling.
Statistical analysis
All data were obtained in 5 independent biological replicates. GraphPad Prism 10.3.0 (GraphPad, CA, USA) was used for statistical analysis, with the data presented as mean ± standard deviation. All data followed a normal distribution as determined by the Shapiro–Wilk test. The equality of variances between groups was validated by Levene’s test. Statistical analysis of the differences between groups were performed using an unpaired two-tailed t-test with a 99% confidence interval. Results were considered statistically significant when p-value of less than 0.01.
Data availability
All analyzed and generated data during current study are included in the article and Supplementary information. Further information is available from the corresponding author on reasonable request.
References
Rocca WA. The burden of Parkinson’s disease: a worldwide perspective. Lancet Neurol. 2018;17:928–9.
Iadanza MG, Jackson MP, Hewitt EW, Ranson NA, Radford SE. A new era for understanding amyloid structures and disease. Nat Rev Mol Cell Biol. 2018;19:755–73.
Tofaris GK. Initiation and progression of alpha-synuclein pathology in Parkinson’s disease. Cell Mol Life Sci. 2022;79:210.
Koike H, Katsuno M. The ultrastructure of tissue damage by amyloid fibrils. Molecules. 2021;26:4611.
Ashrafian H, Zadeh EH, Khan RH. Review on Alzheimer’s disease: inhibition of amyloid beta and tau tangle formation. Int J Biol Macromol. 2021;167:382–94.
Hampel H, Hardy J, Blennow K, Chen C, Perry G, Kim SH, et al. The amyloid-beta pathway in Alzheimer’s disease. Mol Psychiatry. 2021;26:5481–503.
Hickman RA, Faustin A, Wisniewski T. Alzheimer disease and its growing epidemic: risk factors, biomarkers, and the urgent need for therapeutics. Neurol Clin. 2016;34:941–53.
Li X, Feng X, Sun X, Hou N, Han F, Liu Y. Global, regional, and national burden of Alzheimer’s disease and other dementias, 1990-2019. Front Aging Neurosci. 2022;14:937486.
Picken MM. The pathology of amyloidosis in classification: a review. Acta Haematol. 2020;143:322–34.
Gilstrap LG, Dominici F, Wang Y, El-Sady MS, Singh A, Di Carli MF, et al. Epidemiology of cardiac amyloidosis-associated heart failure hospitalizations among fee-for-service medicare beneficiaries in the United States. Circ Heart Fail. 2019;12:e005407.
Quock TP, Yan T, Chang E, Guthrie S, Broder MS. Epidemiology of AL amyloidosis: a real-world study using US claims data. Blood Adv. 2018;2:1046–53.
Nastou KC, Nasi GI, Tsiolaki PL, Litou ZI, Iconomidou VA. AmyCo: the amyloidoses collection. Amyloid. 2019;26:112–7.
Gottwald J, Rocken C. The amyloid proteome: a systematic review and proposal of a protein classification system. Crit Rev Biochem Mol Biol. 2021;56:526–42.
De Michieli L, Stoppa G, Sinigiani G, Previato L, Lorenzoni G, Salvalaggio A, et al. Hospitalization-based epidemiology of systemic and cardiac amyloidosis in the Veneto Region, Italy. Int J Cardiol. 2024;400:131804.
Luo JJ, Wallace W, Kusiak JW. A tough trek in the development of an anti-amyloid therapy for Alzheimer’s disease: Do we see hope in the distance? J Neurol Sci. 2022;438:120294.
Kalia LV, Lang AE. Parkinson’s disease. Lancet. 2015;386:896–912.
Chuang E, Hori AM, Hesketh CD, Shorter J. Amyloid assembly and disassembly. J Cell Sci. 2018;131:jcs189928.
Ramanan VK, Day GS. Anti-amyloid therapies for Alzheimer disease: finally, good news for patients. Mol Neurodegener. 2023;18:42.
Delrieu J, Bateman RJ, Touchon J, Sabbagh M, Cummings J. The future of AD clinical trials with the advent of anti-amyloid therapies: an CTAD Task Force Report. J Prev Alzheimers Dis. 2022;9:393–9.
Haass C, Selkoe D. If amyloid drives Alzheimer disease, why have anti-amyloid therapies not yet slowed cognitive decline? PLoS Biol. 2022;20:e3001694.
Choi Y, Lee W-S, Lee J, Park S-H, Kim S, Kim K-H, et al. Capacitive electrode-based electric field treatments on redox-toxic iron deposits in transgenic AD mouse models: the electroceutical targeting of Alzheimer’s disease feasibility study. Int J Mol Sci. 2023;24:9552.
Bateman RJ, Cummings J, Schobel S, Salloway S, Vellas B, Boada M, et al. Gantenerumab: an anti-amyloid monoclonal antibody with potential disease-modifying effects in early Alzheimer’s disease. Alzheimer’s Res Ther. 2022;14:178.
Lacorte E, Ancidoni A, Zaccaria V, Remoli G, Tariciotti L, Bellomo G, et al. Safety and efficacy of monoclonal antibodies for Alzheimer’s disease: a systematic review and meta-analysis of published and unpublished clinical trials. J Alzheimers Dis. 2022;87:101–29.
Sobral MVS, Soares VG, Gonçalves OR, de Abreu VS, Bendaham LCAR, Batista BLL, et al. Efficacy and safety of gantenerumab in the treatment of Alzheimer’s disease: a meta-analysis of randomized controlled trials. Arch Gerontol Geriatrics. 2024;1:100016.
Budd Haeberlein S, Aisen PS, Barkhof F, Chalkias S, Chen T, Cohen S, et al. Two randomized phase 3 studies of aducanumab in early Alzheimer’s disease. J Prev Alzheimers Dis. 2022;9:197–210.
Roberts RO, Aakre JA, Kremers WK, Vassilaki M, Knopman DS, Mielke MM, et al. Prevalence and outcomes of amyloid positivity among persons without dementia in a longitudinal, population-based setting. JAMA Neurol. 2018;75:970–9.
Rabin JS, Nichols E, La Joie R, Casaletto KB, Palta P, Dams-O’Connor K, et al. Cerebral amyloid angiopathy interacts with neuritic amyloid plaques to promote tau and cognitive decline. Brain. 2022;145:2823–33.
Baker KR. Light chain amyloidosis: epidemiology, staging, and prognostication. Methodist Debakey Cardiovasc J. 2022;18:27–35.
Sanchorawala V. Systemic light chain amyloidosis. N Engl J Med. 2024;390:2295–307.
Cohen E, Paulsson JF, Blinder P, Burstyn-Cohen T, Du D, Estepa G, et al. Reduced IGF-1 signaling delays age-associated proteotoxicity in mice. Cell. 2009;139:1157–69.
Davis DG, Schmitt FA, Wekstein DR, Markesbery WR. Alzheimer neuropathologic alterations in aged cognitively normal subjects. J Neuropathol Exp Neurol. 1999;58:376–88.
Shankar GM, Li S, Mehta TH, Garcia-Munoz A, Shepardson NE, Smith I, et al. Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat Med. 2008;14:837–42.
Silveira JR, Raymond GJ, Hughson AG, Race RE, Sim VL, Hayes SF, et al. The most infectious prion protein particles. Nature. 2005;437:257–61.
Samlaska C, Reber S, Murry T. Insulin-derived amyloidosis: the insulin ball, amyloidoma. JAAD Case Rep. 2020;6:351–3.
Bates G. Huntingtin aggregation and toxicity in Huntington’s disease. Lancet. 2003;361:1642–4.
Tetter S, Arseni D, Murzin AG, Buhidma Y, Peak-Chew SY, Garringer HJ, et al. TAF15 amyloid filaments in frontotemporal lobar degeneration. Nature. 2024;625:345–51.
Oki T, Izuka S, Otsuki Y, Katayama M, Nakamura T. Localized nodular pulmonary amyloidosis mimicking primary lung cancer associated with cystic airspaces: A case report. Radio Case Rep. 2024;19:3719–23.
Bigi A, Cascella R, Chiti F, Cecchi C. Amyloid fibrils act as a reservoir of soluble oligomers, the main culprits in protein deposition diseases. Bioessays. 2022;44:e2200086.
Lu C, Cai X, Zhi S, Wen X, Shen J, Ercoli T, et al. Exploring the association between cathepsin B and Parkinson’s Disease. Brain Sci. 2024;14:482.
Jones-Tabah J, He K, Senkevich K, Karpilovsky N, Deyab G, Cousineau Y, et al. The Parkinson’s disease risk gene cathepsin B promotes fibrillar alpha-synuclein clearance, lysosomal function and glucocerebrosidase activity in dopaminergic neurons. Mol Neurodegeneration. 2024;19:88.
Bohne S, Sletten K, Menard R, Buhling F, Vockler S, Wrenger E, et al. Cleavage of AL amyloid proteins and AL amyloid deposits by cathepsins B, K, and L. J Pathol. 2004;203:528–37.
Mueller-Steiner S, Zhou Y, Arai H, Roberson ED, Sun B, Chen J, et al. Antiamyloidogenic and neuroprotective functions of cathepsin B: implications for Alzheimer’s disease. Neuron. 2006;51:703–14.
Cataldo AM, Nixon RA. Enzymatically active lysosomal proteases are associated with amyloid deposits in Alzheimer brain. Proc Natl Acad Sci USA. 1990;87:3861–5.
Hook V, Toneff T, Bogyo M, Greenbaum D, Medzihradszky KF, Neveu J, et al. Inhibition of cathepsin B reduces beta-amyloid production in regulated secretory vesicles of neuronal chromaffin cells: evidence for cathepsin B as a candidate beta-secretase of Alzheimer’s disease. Biol Chem. 2005;386:931–40.
Wang C, Sun B, Zhou Y, Grubb A, Gan L. Cathepsin B degrades amyloid-beta in mice expressing wild-type human amyloid precursor protein. J Biol Chem. 2012;287:39834–41.
Perlenfein TJ, Murphy RM. A mechanistic model to predict effects of cathepsin B and cystatin C on beta-amyloid aggregation and degradation. J Biol Chem. 2017;292:21071–82.
Yamada T, Liepnieks JJ, Kluve-Beckerman B, Benson MD. Cathepsin B generates the most common form of amyloid A (76 residues) as a degradation product from serum amyloid A. Scand J Immunol. 1995;41:94–7.
McGlinchey RP, Lacy SM, Huffer KE, Tayebi N, Sidransky E, Lee JC. C-terminal alpha-synuclein truncations are linked to cysteine cathepsin activity in Parkinson’s disease. J Biol Chem. 2019;294:9973–84.
Ferree AW. Cathepsin oxidation alters alpha-synuclein processing. Front Neurol. 2019;10:530.
McGlinchey RP, Lee JC. Cysteine cathepsins are essential in lysosomal degradation of alpha-synuclein. Proc Natl Acad Sci USA. 2015;112:9322–7.
Tsujimura A, Taguchi K, Watanabe Y, Tatebe H, Tokuda T, Mizuno T, et al. Lysosomal enzyme cathepsin B enhances the aggregate forming activity of exogenous alpha-synuclein fibrils. Neurobiol Dis. 2015;73:244–53.
Leissring MA, Farris W, Chang AY, Walsh DM, Wu X, Sun X, et al. Enhanced proteolysis of beta-amyloid in APP transgenic mice prevents plaque formation, secondary pathology, and premature death. Neuron. 2003;40:1087–93.
Hook G, Reinheckel T, Ni J, Wu Z, Kindy M, Peters C, et al. Cathepsin B gene knockout improves behavioral deficits and reduces pathology in models of neurologic disorders. Pharm Rev. 2022;74:600–29.
Siddiqui AA, Merquiol E, Bruck-Haimson R, Hirbawi J, Boocholez H, Cohen I, et al. Cathepsin B promotes Aβ proteotoxicity by modulating aging regulating mechanisms. Nat Commun. 2024;15:8564.
Ghosh U, Thurber KR, Yau WM, Tycko R. Molecular structure of a prevalent amyloid-beta fibril polymorph from Alzheimer’s disease brain tissue. Proc Natl Acad Sci USA. 2021;118:e2023089118.
Iwatsubo T, Odaka A, Suzuki N, Mizusawa H, Nukina N, Ihara Y. Visualization of A beta 42(43) and A beta 40 in senile plaques with end-specific A beta monoclonals: evidence that an initially deposited species is A beta 42(43). Neuron. 1994;13:45–53.
Spillantini MG, Crowther RA, Jakes R, Hasegawa M, Goedert M. alpha-Synuclein in filamentous inclusions of Lewy bodies from Parkinson’s disease and dementia with lewy bodies. Proc Natl Acad Sci USA. 1998;95:6469–73.
Araki K, Yagi N, Aoyama K, Choong CJ, Hayakawa H, Fujimura H, et al. Parkinson’s disease is a type of amyloidosis featuring accumulation of amyloid fibrils of alpha-synuclein. Proc Natl Acad Sci USA. 2019;116:17963–9.
Pujols J, Pena-Diaz S, Lazaro DF, Peccati F, Pinheiro F, Gonzalez D, et al. Small molecule inhibits alpha-synuclein aggregation, disrupts amyloid fibrils, and prevents degeneration of dopaminergic neurons. Proc Natl Acad Sci USA. 2018;115:10481–6.
Nakamura S, Yamada M, Iijima Y, Sawada K, Hino S, Kaneko T, et al. Hemodialysis-Related Amyloidosis in the Tongue. Case Rep. Dent. 2022;2022:9098201.
Picciani B, Gornic C, Cunha KS, Janini MER, Andrade RM, Meireles VJ, et al. Lingual amyloidosis associated to long-term hemodialysis: two case reports. Spec Care Dent. 2018;38:434–7.
Pleyer C, Flesche J, Saeed F. Lysozyme amyloidosis—a case report and review of the literature. Clin Nephrol Case Stud. 2015;3:42–5.
Anker S, Hinderhofer K, Baur J, Haupt C, Rocken C, Beimler J, et al. Lysozyme amyloidosis-a report on a large German cohort and the characterisation of a novel amyloidogenic lysozyme gene variant. Amyloid. 2022;29:245–54.
Yoon MC, Solania A, Jiang Z, Christy MP, Podvin S, Mosier C, et al. Selective neutral pH inhibitor of cathepsin B designed based on cleavage preferences at cytosolic and lysosomal pH conditions. ACS Chem Biol. 2021;16:1628–43.
Gounaris AD, Slater EE. Cathepsin B from human renal cortex. Biochem J. 1982;205:295–302.
Giordano C, Sansone A, Masi A, Masci A, Mosca L, Chiaraluce R, et al. Inhibition of amyloid peptide fragment Abeta25-35 fibrillogenesis and toxicity by N-terminal beta-amino acid-containing esapeptides: is taurine moiety essential for in vivo effects? Chem Biol Drug Des. 2012;79:30–7.
Chiti F, Dobson CM. Protein misfolding, functional amyloid, and human disease. Annu Rev Biochem. 2006;75:333–66.
Xue C, Lin TY, Chang D, Guo Z. Thioflavin T as an amyloid dye: fibril quantification, optimal concentration and effect on aggregation. R Soc Open Sci. 2017;4:160696.
Sulatsky MI, Stepanenko OV, Stepanenko OV, Mikhailova EV, Kuznetsova IM, Turoverov KK, et al. Amyloid fibrils degradation: The pathway to recovery or aggravation of the disease? Front Mol Biosci. 2023;10:1208059.
Stepanenko OV, Sulatsky MI, Mikhailova EV, Stepanenko OV, Kuznetsova IM, Turoverov KK, et al. Trypsin induced degradation of amyloid fibrils. Int J Mol Sci. 2021;22:4828.
Stepanenko OV, Sulatsky MI, Mikhailova EV, Stepanenko OV, Povarova OI, Kuznetsova IM, et al. Alpha-B-crystallin effect on mature amyloid fibrils: different degradation mechanisms and changes in cytotoxicity. Int J Mol Sci. 2020;21:7659.
Sulatsky MI, Sulatskaya AI, Stepanenko OV, Povarova OI, Kuznetsova IM, Turoverov KK. Denaturant effect on amyloid fibrils: declasterization, depolymerization, denaturation and reassembly. Int J Biol Macromol. 2020;150:681–94.
Biancalana M, Makabe K, Koide A, Koide S. Molecular mechanism of thioflavin-T binding to the surface of beta-rich peptide self-assemblies. J Mol Biol. 2009;385:1052–63.
Krebs MR, Bromley EH, Donald AM. The binding of thioflavin-T to amyloid fibrils: localisation and implications. J Struct Biol. 2005;149:30–7.
Kuznetsova IM, Sulatskaya AI, Uversky VN, Turoverov KK. A new trend in the experimental methodology for the analysis of the thioflavin T binding to amyloid fibrils. Mol Neurobiol. 2012;45:488–98.
Kuznetsova IM, Sulatskaya AI, Uversky VN, Turoverov KK. Analyzing thioflavin T binding to amyloid fibrils by an equilibrium microdialysis-based technique. PloS ONE. 2012;7:e30724.
Sulatsky MI, Stepanenko OV, Stepanenko OV, Povarova OI, Kuznetsova IM, Turoverov KK, et al. Broken but not beaten: challenge of reducing the amyloids pathogenicity by degradation. J Adv Res. 2024. https://doi.org/10.1016/j.jare.2024.04.018.
Marrero-Winkens C, Sankaran C, Schatzl HM. From seeds to fibrils and back: fragmentation as an overlooked step in the propagation of prions and prion-like proteins. Biomolecules. 2020;10:1305.
Kushnirov VV, Ter-Avanesyan MD. Structure and replication of yeast prions. Cell. 1998;94:13–6.
Jarrett JT, Lansbury PT Jr. Seeding “one-dimensional crystallization” of amyloid: a pathogenic mechanism in Alzheimer’s disease and scrapie? Cell. 1993;73:1055–8.
Sulatskaya AI, Rodina NP, Polyakov DS, Sulatsky MI, Artamonova TO, Khodorkovskii MA, et al. Structural features of amyloid fibrils formed from the full-length and truncated forms of beta-2-microglobulin probed by fluorescent dye thioflavin T. Int J Mol Sci. 2018;19:2762.
Huang C, Ren G, Zhou H, Wang CC. A new method for purification of recombinant human alpha-synuclein in Escherichia coli. Protein Expr Purif. 2005;42:173–7.
Broersen K, Jonckheere W, Rozenski J, Vandersteen A, Pauwels K, Pastore A, et al. A standardized and biocompatible preparation of aggregate-free amyloid beta peptide for biophysical and biological studies of Alzheimer’s disease. Protein Eng Des Sel. 2011;24:743–50.
Kayed R, Head E, Sarsoza F, Saing T, Cotman CW, Necula M, et al. Fibril specific, conformation dependent antibodies recognize a generic epitope common to amyloid fibrils and fibrillar oligomers that is absent in prefibrillar oligomers. Mol Neurodegener. 2007;2:18.
Sulatskaya AI, Rychkov GN, Sulatsky MI, Mikhailova EV, Melnikova NM, Andozhskaya VS, et al. New evidence on a distinction between Abeta40 and Abeta42 amyloids: thioflavin T binding modes, clustering tendency, degradation resistance, and cross-seeding. Int J Mol Sci. 2022;23:5513.
Sulatskaya AI, Rodina NP, Kuznetsova IM, Turoverov KK. Different conditions of fibrillogenesis cause polymorphysm of lysozyme amyloid fibrils. J Mol Struct. 2017;1140:52–8.
Vladimirov YA, Litvin FF. Photobiology and spectroscopic methods. In: Handbook of general biophisics. 8. Moscow: High school; 1964. p. 88–91.
Fonin AV, Sulatskaya AI, Kuznetsova IM, Turoverov KK. Fluorescence of dyes in solutions with high absorbance. Inner filter effect correction. PloS ONE. 2014;9:e103878.
Sulatskaya AI, Lavysh AV, Maskevich AA, Kuznetsova IM, Turoverov KK. Thioflavin T fluoresces as excimer in highly concentrated aqueous solutions and as monomer being incorporated in amyloid fibrils. Sci Rep. 2017;7:2146.
Nishimura S, Murasugi T, Kubo T, Kaneko I, Meguro M, Marumoto S, et al. RS-4252 inhibits amyloid beta-induced cytotoxicity in HeLa cells. Pharmacol Toxicol. 2003;93:29–32.
Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods. 1983;65:55–63.
Funding
The study was funded by the Russian Science Foundation (grant No. 23-74-10092).
Author information
Authors and Affiliations
Contributions
Conceived and designed the experiments: MIS, OVS, OVS, and AIS. Performed the experiments and analyzed the data: MIS, OVS, OVS, EVM, and AIS. Wrote the manuscript: MIS, OVS, OVS, and AIS. Acquired funding: AIS. Supervised the project: AIS. All authors read and approved the final manuscript.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary information
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Sulatsky, M.I., Stepanenko, O.V., Stepanenko, O.V. et al. Cathepsin B prevents cell death by fragmentation and destruction of pathological amyloid fibrils. Cell Death Discov. 11, 61 (2025). https://doi.org/10.1038/s41420-025-02343-w
Received:
Revised:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41420-025-02343-w