Skip to main content
Redox Biology logoLink to Redox Biology
. 2024 Mar 28;72:103133. doi: 10.1016/j.redox.2024.103133

Αnti-prion effects of anthocyanins

Nikoletta Christoudia a, Nikolaos Bekas a, Eirini Kanata b, Athanasia Chatziefsthathiou a, Spyros Pettas a,b, Korina Karagianni a, Susana Margarida Da Silva Correia c, Matthias Schmitz c, Inga Zerr c, Ioannis Tsamesidis d, Konstantinos Xanthopoulos b,1, Dimitra Dafou a,⁎,1, Theodoros Sklaviadis b,1
PMCID: PMC10990977  PMID: 38565068

Abstract

Prion diseases, also known as Transmissible Spongiform Encephalopathies (TSEs), are protein-based neurodegenerative disorders (NDs) affecting humans and animals. They are characterized by the conformational conversion of the normal cellular prion protein, PrPC, into the pathogenic isoform, PrPSc. Prion diseases are invariably fatal and despite ongoing research, no effective prophylactic or therapeutic avenues are currently available. Anthocyanins (ACNs) are unique flavonoid compounds and interest in their use as potential neuroprotective and/or therapeutic agents against NDs, has increased significantly in recent years. Therefore, we investigated the potential anti-oxidant and anti-prion effects of Oenin and Myrtillin, two of the most common anthocyanins, using the most accepted in the field overexpressing PrPScin vitro model and a cell free protein aggregation model. Our results, indicate both anthocyanins as strong anti-oxidant compounds, upregulating the expression of genes involved in the anti-oxidant response, and reducing the levels of Reactive Oxygen Species (ROS), produced due to pathogenic prion infection, through the activation of the Keap1-Nrf2 pathway. Importantly, they showcased remarkable anti-prion potential, as they not only caused the clearance of pathogenic PrPSc aggregates, but also completely inhibited the formation of PrPSc fibrils in the Cerebrospinal Fluid (CSF) of patients with Creutzfeldt–Jakob disease (CJD). Therefore, Oenin and Myrtillin possess pleiotropic effects, suggesting their potential use as promising preventive and/or therapeutic agents in prion diseases and possibly in the spectrum of neurodegenerative proteinopathies.

Keywords: Prion, Anthocyanins, Anti-oxidant, Neuroprotection, Proteinopathies, PrPSc

Graphical abstract

Image 1

Highlights

  • Oenin and Myrtillin reduce ROS production in N2a22L cells.

  • Oenin and Myrtillin activate the Keap1-Nrf2 axis in N2a22L cells.

  • Oenin and Myrtillin reduce PrPSc accumulation in N2a22L cells.

  • Oenin and Myrtillin impede de novo PrPSc formation and aggregation.

  • Oenin and Myrtillin are potent compounds for intervention against prion diseases.

Abbreviations

NDs

Neurodegenerative Disorders

CJD

Creutzfeldt-Jakob disease

TSEs

Transmissible Spongiform Encephalopathies

PrPC

Cellular prion protein

PrPSc

Scrapie infectious prion protein

ER

Endoplasmic Reticulum

ROS

Reactive Oxygen Species

OX0B

β-actin

EDTA

Ethylene Diamine Tetraacetic Acid

PMSF

Phenylmethylsulfonyl Fluoride

PVDF

Polyvinylidene difluoride

RT-QuIC

Real-time quaking-induced conversion

Th-T

Thioflavin – T

AREs

Antioxidant Response Elements

AUC

Area Under the Curve

UPR

Unfolded Protein Response

1. Introduction

Prion diseases are progressive and fatal Neurodegenerative Diseases (NDs), such as Creutzfeldt–Jakob disease (CJD), that affect humans and animals [[1], [2], [3]]. The fundamental event underlying scrapie infection seems to be a conformational change in the prion protein. Transmissible Spongiform Encephalopathies (TSEs) share a common pathogenic mechanism, which involves the autocatalytic conversion of the normal prion protein, PrPC, to its disease associated variant, PrPSc. PrPC molecules are repeatedly recruited and misfolded by PrPSc, resulting in the formation of protease-resistant aggregates, known as amyloid fibrils [[1], [2], [3]]. Accumulation of PrPSc fibrils, results in Endoplasmic Reticulum (ER) stress, dysregulated calcium signaling, mitochondrial disfunction, and eventually neuronal cell death [[4], [5], [6]].

TSEs are strongly associated with oxidative stress [[7], [8], [9], [10]]. PrPC has an important anti-oxidant function, binding bivalent metal ions [[11], [12], [13], [14]] and acting as a quencher of Reactive Oxygen Species (ROS) [[15], [16], [17]]. Loss of PrPC activity leads to a marked increase in oxidation levels [8,[18], [19], [20], [21], [22], [23]]. Oxidative stress occurs early during the onset and the progression of prion diseases [20,24], and models with compromised anti-oxidation response showcase accelerated disease progression [25,26]. Conversely, reduced ROS levels are associated with increased survival and improved phenotype [22]. Furthermore, PrPSc aggregation causes an uptake in mitochondrial ROS production and decreased levels of oxidative phosphorylation (OXPHOS) [[27], [28], [29]], which further aggravates ROS formation and oxidative stress observed in prion-affected cells [23,30].

Enhancement of anti-oxidant responses emerges as a promising therapeutic approach against neurodegeneration [31,32]. Anthocyanins (ACNs) are polyphenolic derivatives of the anthocyanidin flavonoid group, and act as water soluble vacuolar pigments in various flowers, fruits and vegetables (Supplementary Fig. 1) [33]. Due to their potent anti-oxidant action, ACNs have been extensively tested as potential remedies against oxidative stress associated conditions, such as cancer [[34], [35], [36], [37]], cardiovascular diseases [[38], [39], [40], [41]] and neurodegeneration [[42], [43], [44], [45]]. Importantly, their ability to penetrate the Blood-Brain Barrier (BBB) renders them exceptional neuroprotective compounds [46].

Oenin (Malvidin-3-glucoside) and Myrtillin (Delphinidin 3-glucoside) are two of the most prevalent ACNs present in grapes and red wine [47]. While, they have previously showcased anti-oxidant and anti-inflammatory activity [[48], [49], [50], [51], [52], [53]], their potential effect in prion diseases has not yet been investigated. In this study, the anti-prion potential of Oenin and Myrtillin (Supplementary Fig. 2) is described for the first time. In scrapie-infected murine neuroblastoma N2a (ScN2a) cells, we addressed the effect of ACNs for the reduction of ROS levels through the activation of the Keap1-Nrf2 pathway, and the reduction of PrPSc aggregates in ScN2a22L cells, and also the inhibition of the formation of PrPSc fibrils in the Cerebrospinal fluid (CSF) of CJD patients. Therefore, our results highlight the strong potential of Oenin and Myrtillin against prion diseases and possibly other neurodegenerative proteinopathies.

2. Materials and methods

2.1. Cell culture and LD50 Estimation

The murine neuroblastoma cell line N2a22L has been utilized, in which the murine scrapie prion 22L strain is expressed, leading to the sustained production of Proteinase K (PK) resistance protein, PrPSc [54]. The N2a22L cell line has been widely used over the years as the most reliable model for the study of PrPSc aggregation [18,[55], [56], [57], [58], [59], [60], [61], [62], [63], [64]]. Cells were cultured in Opti-MEM (51985042, Invitrogen Waltham, MA, USA) supplemented with 10% Fetal Bovine Serum (FBS) under a 5% CO2 at 37 °C. Oenin (0911S, Extrasynthese Genay, France) and Myrtillin (0938S, Extrasynthese Genay, France) were dissolved in dimethyl sulfoxide (DMSO). Lethal Dose 50% (LD50) values were estimated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) Assay (M6494, Invitrogen, Waltham, MA, USA), following 48-h incubation [65,66] with gradually increased concentrations of Oenin and Myrtillin. Control cells were treated with DMSO in concentrations matching those delivered with the compounds. All experiments were performed in triplicates.

2.2. In vitro estimation of ROS amounts

N2a22L cells were incubated with Oenin or Myrtillin (250 μΜ) for 48 h. Cells were incubated for 30 min at room temperature with H2DCFDA (2′,7′-Dichloro-dihydro-fluorescein, D399, Invitrogen, Waltham, MA, USA) dissolved in DMSO, and fluorescence was measured using a Tecan fluorometer. Controls received DMSO at concentrations matching those delivered with the compounds. The same set of experiments were performed after pre-treatment with two different concentrations of H2O2, 6.25 μM and 200 μM for 30 min before addition of Oenin and Myrtillin. Analysis was done in triplicates and relative fluorescence was expressed as “% of maximum emission”, determined with Tecan Magellan software (https://lifesciences.tecan.com/software-magellan, accessed December 19, 2023).

2.3. RNA Isolation and qPCR

Total RNA was extracted using spin columns (740955.250, Macherey Nagel, Dueren, Germany). For cDNA synthesis, 500 ng total RNA and the TaKaRa PrimeScript RT Reagent Kit (RR037A, TAKARA, Shiga, Japan) were used. Relative expression of Heme Oxygenase 1 (HMOX1), Glutamate Cysteine Ligase Regulatory Subunit (GCLM) and Nuclear factor (erythroid – derived 2) - like 2 (NFE2L2) was estimated by qPCR, using β-actin (ACTB) for normalization. The KAPA SYBR fast qPCR kit, 20 ng cDNA and 0.1 μΜ each primer was used (Supplementary Table 1). Reactions were performed in a 7500 Fast Real-time PCR System (Applied Biosystems), in triplicates.

2.4. In vitro assessment of PrPSc aggregation

Oenin-, Myrtillin-treated and control N2a22L cells were lysed in ice-cold lysis buffer (10 mM Tris pH 7.5, 100 mM NaCl, 10 mM EDTA, 0.5% v/v Triton-X-100, 0.5% w/v sodium deoxycholate) and centrifuged (1 min, 14,000×g). Total protein in the supernatant was estimated with Bradford reagent (A6932, 0250, AppliChem, Darmstadt, Germany). One fraction of each lysate was digested with PK (1.24569.0100, Merck, Darmstadt, Germany) in 1% w/v N lauryl-sarcosine. Phenylmethylsulfonyl Fluoride (PMSF) (5 mM final concentration) was used to stop the reaction. PK treated samples (PK+) and non-PK treated samples (PK-) were resolved on 12% w/v poly-acrylamide gels, electro-transferred onto Polyvinylidene Fluoride (PVDF) membranes and subjected to Western Blot analysis using the monoclonal antibody 6H4. Chemiluminescence was used for development on X-ray films. Films were digitized and relative protein levels were estimated with ImageJ (available at https://imagej.net/ij/index.html, accessed on May 08, 2023), utilizing exposures within the linear dynamic range of the film.

For each sample, the ratio of the intensity of PrP-immunopositive bands in the PK-resistant fraction (PK+) to the intensity of total PrP in the non-PK (PK-) treated fraction was estimated and expressed relative to controls using the formula:

[PrP(RES)ACN/ PrP(TOT)ACN]*100/[PrP(RES)Cntr/PrP(TOT)Cntr]

Where PrP(RES)ACN and PrP(TOT)ACN are the intensity of PrP bands in the PK-treated and non-PK-treated fractions respectively in Oenin or Myrtillin treated samples. PrP(RES)Cntr and PrP(TOT)Cntr show the intensity of PrP bands in the PK-treated and non-PK treated controls, respectively. In order to verify that PK treatment conditions resulted in complete digestion of PrPC, cell lysates from N2a58 cells that were not prion-infected underwent a similar PK treatment and were subsequently immunolabelled for PrP. PK treatment completely digested PrPC, because no PrP immunoreactivity was found in these experiments (Supplementary Fig. 4). The immunoreactive bands detected in N2a22L cells that were treated with PK do not correspond to partially digested PrPC, but rather to PrPSc.

2.5. Cell free detection of de novo PrPSc fibrillation through RT-QuIC

Real-time quaking-induced conversion reactions, RT-QuIC [67] were performed using CSF containing PrPSc seed material from patients with confirmed sCJD diagnosis, originating from the National Reference Center for TSEs, Göttingen, Germany. 15 μL CSF (diluted 1000 times) was mixed with 85 μL reaction buffer (5 × PBS pH 6.9, 170 mM NaCl, 1 mM EDTA, 10 μM Thioflavin-T and 0.1 mg/mL recPrPC). Oenin or Myrtillin were added to a final concentration of 2.5 μM. Reactions were set in 96-well black bottom optical plates and carried out in a BMG Labtech FluoO Star OPTIMA plate reader at 42 °C for 80 h with intermittent rest and shaking cycles. Thioflavin-T (Th-T) fluorescence was measured every 30 min. Analysis was performed in triplicates.

2.6. Statistical analysis

GraphPad Prism version 8.0.2 for Windows (GraphPad Software, San Diego, CA, USA, www.graphpad.com, Accessed on May 08, 2023) was used. Non-linear regression analysis was applied to the dose-response equations for LD50 determination. Differences in gene expression and PrPSc accumulation between untreated and treated cells were estimated with unpaired, one-tailed T-tests. Data represent Standard Error of Mean (SEM) of three independent experiments and P-values of 0.05 or lower were considered statistically significant.

3. Results

3.1. Assessment of LD50 of Oenin and Myrtillin in N2a22L cell line

The viability assay showed Oenin as less toxic. Oenin LD50 values estimated at 506.8 μM as opposed to Myrtillin LD50 estimated at 293.1 μM (Supplementary Fig. 3). A concentration of 250 μM for each ACN was used for the rest of the study, in which both compounds presented no cytotoxicity to N2a22L cells, and cell treatment entailed 48-h incubation.

3.2. Oenin and Myrtillin reduced ROS levels in ScN2a22L cells

Prion diseases are associated with elevated oxidation and ROS production [[8], [9], [10],22]. Owing to their known anti-oxidant activity [41,48], it was tested whether Oenin and Myrtillin could affect ROS levels in ScN2a22L cells. Both compounds significantly reduced the endogenous ROS levels and the ROS produced after H2O2 administration (Fig. 1). It is worth noting that, Myrtillin in most cases (without H2O2 administration and with 6.25 μM) neutralized better the amount of generated ROS when compared with Oenin. These results showcased that Oenin and especially Myrtillin have strong anti-oxidant action in prion affected cells.

Fig. 1.

Fig. 1

Anti-oxidant effects of Oenin and Myrtillin in N2a22L cells. ROS levels were measured in N2a22L cells after a 48-h treatment with Oenin or Myrtillin (250 μΜ each), without or following pre-treatment with (A) 6.25 μM H2O2 and (B) 200 μM H2O2, to induce oxidative stress. Controls received DMSO at concentrations matching those delivered with the compounds. For ROS measurement, H2DCFDA, dissolved in DMSO, was added in the cell medium at a final concentration of 20 μΜ and cells were further incubated for 30 min at room temperature. Then, fluorescence was measured using a Tecan fluorometer. The % ROS was calculated based on the maximum ROS production value. (C) Oenin and Myrtillin induce the expression of Keap1-Nrf2 pathway gene targets in N2a22L cells. The Keap1-Nrf2 pathway is a key cellular defense mechanism against oxidative stress, that protects cells by reducing the risk of ROS-mediated damage through the activation of cytoprotective enzymes. More specifically, Nrf2 binds to Antioxidant Response Elements (AREs) in the promoters of anti-oxidant genes, aiming to restore redox homeostasis [68]. The expression of NFE2L2 (encoding Nrf2), GCLM and HMOX1 in Oenin and Myrtillin treated cells (250 μΜ, 48 h) is assessed in N2a22L cells, relative to controls (administered DMSO at the same concentration as those delivered with the compounds). Data represent Standard Error of Mean (SEM) of three independent experiments. Stars denote statistical significance (unpaired, one-tailed, T-test); *: p-value <0.05, **: p-value <0.01.

3.3. Oenin and Myrtillin activate the Keap1-Nrf2 pathway

Many compounds exert their anti-oxidant neuroprotective effects through Keap1-Nrf2 activation [[69], [70], [71], [72], [73], [74], [75], [76]]. Consequently, it was tested whether the observed anti-oxidant effects of Oenin and Myrtillin in N2a22L cells also depended on Keap1-Nrf2 induction. Indeed, both ACN compounds managed to trigger the expression of key Nrf2 antioxidant target genes, GCLM and HMOX1 [77]. Interestingly, Myrtillin not only presented a more profound effect in GCLM and HMOX1 levels, but also upregulated the expression of Nrf2 (NFE2L2) (Fig. 1).

3.4. Oenin and Myrtillin decrease the levels of PrPSc aggregates

The PrPSc leads to enhanced resistance against PK and higher propensity to polymerize into amyloid fibrils, the primary cause of prion diseases [[78], [79], [80], [81]]. Consequently, reducing the amount of PrPSc is of paramount importance for any potential anti-prion and neuroprotective compound. For that reason, the ability of Oenin and Myrtillin to increase the PK sensitivity of PrPSc aggregates, was tested. Both compounds significantly reduced the amount of PrPSc aggregation in N2a22L cells, providing further support for their potential anti-prion action (Fig. 2).

Fig. 2.

Fig. 2

Oenin and Myrtillin reduce PrPSc aggregation in N2a22L cells. (A) Representation of the structural rearrangement taking place during the PrPC–PrPSc conversion. During this process, the α-helix rich PrPC (in which C stands for the cellular form of the normal prion protein and is expressed in neurons and the spinal cord) is transformed into the β-sheet enriched PrPSc (Sc stands for scrapie, the prion disease of sheep and goats). This results in physio- and bio-chemical properties distinct from PrPC, including reduced solubility in mild detergents, enhanced resistance to partial proteolysis by PK. (B) Representative Western blot results for each compound, along with the densitometric analysis from three independent experiments are depicted. Cell lysates from Oenin and Myrtillin treated N2a22L cells as well as controls (administered DMSO at the same concentrations as those delivered to the ACN treated cells) were processed for PrP immunodetection. A fraction of each lysate (150 μg total protein) was treated with proteinase K (PK+, 1.25 μg PK/mg total protein) for 1 h at 37 °C, to allow the identification of the partially resistant to PK, PrPSc. Due to its conformation, PrPSc is not accessible for enzymatic treatment, except a segment at its amino-terminal site which is digested resulting in the characteristic band shift of PrP immunopositive bands towards lower molecular weights. Analysis of non-PK treated (PK-) material (50 μg) from the same sample allowed total PrP detection (PrPC and PrPSc). For PrP immunodetection the monoclonal 6H4 antibody (7500997, Invitrogen, Waltham, MA, USA) was used (0.2 μg/mL). PK activity degrades β-Actin, thus it is not visible in PK(+) samples. Blots were developed on autoradiography films using chemiluminesence. Densitometric analysis was performed with ImageJ. Bar graphs show the conversion rate of each ACN treated sample (PrPSc/Total PrP) relative to the control conversion rate (PrPSc %). Data represent Standard Error of Mean (SEM). Stars denote statistical significance (unpaired, one-tailed, T-test); *: p value < 0.05, **: p value < 0.01.

3.5. Oenin and Myrtillin inhibit the de novo PrPSc aggregation

Oenin and Myrtillin promoted the clearance of PrPSc aggregates. As a result, it was tested whether they could also block the de novo PrPSc fibrillation. For that purpose, RT-QuIC [82], a highly sensitive technique that is routinely used for the diagnosis of prion diseases and similar neurodegenerative disorders, that it is able to detect the presence of misfolded proteins with almost 100% accuracy, was utilized [[83], [84], [85]]. Consequently, it has also been deployed for the screening of anti-prion compounds [67,[86], [87], [88], [89], [90]]. Both compounds showcased remarkable anti-aggregation capacity, as they completely inhibited the formation of PrPSc fibrils at concentrations of 5 and 10 μM for both compounds, and Myrtillin maintained moderate anti-aggregation action even at 2.5μΜ (Fig. 3).

Fig. 3.

Fig. 3

Oenin and Myrtillin inhibit recPrPC fibrillation in RT-QuIC assays seeded with human PrPSc. (A) Summary of RT-QuIC steps: (1) A sample containing PrPSc (such as CSF from CJD patients) is mixed with a recombinant PrPC (recPrPC) monomers and Th-T, which specifically binds to β-sheets, leading to fluorescence. (2) The recPrPC monomers are recruited by the PrPSc oligomers. (3) Recruited recPrPC monomers are transformed into recPrPSc and the PrPSc oligomers are elongated. (4) Creation of PrPSc -recPrPSc fibrils. (5) Quaking induces fragmentation of the PrPSc fibrils. (6) The process is repeated [83,91,92]. (B) Aggregation of recPrPC in RT-QuIC was assessed in the CSF from twelve different CJD patients. Oenin and Myrtillin were added in the reaction mixture of RT-QuIC in three different concentrations (2.5, 5 and 10 μM) and the results were compared with that from CSF only and CSF with DMSO. Reactions were set using 15 μL of diluted seed material and performed at 42 °C for 80 h with intermittent rest and shaking cycles. Th-T fluorescence, as a measure of protein aggregation, was recorded every 30 min. The graph depicts combined (mean) data from the results acquired from the twelve independent patients CSFs used as seed. sCJD: positive control; RT-QuIC assays performed with no anthocyanin supplementation. Oenin, Myrtillin: RT-QuIC assays performed in the presence of Oenin or Myrtillin. Both compounds block PrP aggregates formation. (C) Quantification of Oenin and Myrtillin effects on PrP conversion and aggregation inhibition. Box plots represent the Standard Error of Mean (SEM) of the Area Under Curve (AUC) calculated for the individual fluorescence curves of each replicate reaction. AUC values were used as a measure of protein conversion and aggregation. Stars indicate statistical significance (unpaired, one-tailed, T-test). **: p value < 0.01, ***: p value < 0.001.

4. Discussion

Prion diseases belong to a group of NDs known as proteinopathies, or prion-like diseases, in which pathologic protein misfolding and accumulation, plays a crucial role in disease development and progression [[93], [94], [95], [96]]. In the case of prion diseases, this is due to the transformation of normal prion protein, PrPC, into the pathologic PrPSc [78,79]. In this study, the strong anti-prion ability of Oenin and Myrtillin are described for the first time. Treatment with Oenin and Myrtillin for just 48 h was able to significantly decrease the number of PrPSc aggregates in N2a22L cells. Additionally, both compounds completely inhibited the de novo formation of PrPSc fibrils in the CSF of CJD patients, at a concentration of 10 μM and 5 μM, in the case of Myrtillin, maintained robust anti-prion action even at 2.5 μM.

While the mechanism of their anti-aggregation action is yet to be elucidated, the Keap1-Nrf2 pathway is highly likely to be responsible. Indeed, previous studies in proteinopathies showed that Nrf2 activation inhibited the formation and/or reduced the number of existing aggregates of α-synuclein [[97], [98], [99], [100]], amyloid beta [[101], [102], [103], [104]] and tau [[105], [106], [107]], whereas Nrf2 deficiency promotes protein aggregation [[108], [109], [110]]. The Nrf2 transcription factor has long been identified as a modulator of autophagy [[111], [112], [113], [114]], and is also associated with the Unfolded Protein Response (UPR) [100,[115], [116], [117], [118], [119], [120]], which might explain its ability to reduce pathologic protein aggregation in several proteinopathies, including prion diseases [121,122]. Interestingly, activation of the UPR has been utilized as a potential treatment against CJD [123], whereas p62 mediated Nrf2 activation and subsequent upregulation of autophagy levels has been proposed as a therapeutic strategy for prion diseases [6].

Additionally, Oenin and Myrtillin could be directly interacting with the prion protein. Indeed, other flavonoids are capable of directly binding to PrPC. For example, Quercetin, interaction with PrPSc fibrils renders them vulnerable to protein degradation, leading to de-aggregation [123,124]. Moreover, Apigenin and Nepetin managed to inhibit the fibrillation of the PrP106-126 peptide and also depolymerize the already formed fibrils [125]. It is also worth noting that, Oenin and Myrtillin might exert their anti-prion action with a combination of different mechanisms. Quercetin can simultaneously bind to the C-terminal region of murine prion protein and also act as an anti-oxidant [124,126]. A similar observation was made with Curcumin, which can bind to PrPSc fibrils, as well as intermediate aggregates of the PrPC–PrPSc conversion, while also exerting anti-oxidant action [127,128].

Oxidative stress has been identified as a hallmark of neurodegeneration [32,[129], [130], [131]]. In accordance with other anti-oxidant compounds exhibiting anti-prion activity [72,124,126,128,132,133], Oenin and Myrtillin successfully decreased the levels of ROS in prion-infected cells. While Oenin didn't affect Nrf2 expression levels, it managed to activate the Keap1-Nrf2 pathway (albeit less effectively compared to Myrtillin). Indeed, the important step for the activation of Keap1-Nrf2 target genes is nuclear translocation of Nrf2 [70,73,75,114,134,135]. As a result, one potential explanation for the activity of Oenin is that it successfully triggered the nuclear translocation of Nrf2, but due to the fact that it didn't lead to upregulation of Nrf2 itself, the activation of the Keap1-Nrf2 pathway was less potent.

5. Conclusions

To summarize, our results of the current study provide promising evidence regarding the anti-prion neuroprotective potential of Oenin and Myrtillin. Both compounds are able to de-aggregate pre-existing PrPSc fibrils, and also severely inhibit the process of de novo PrPSc fibrillation. Moreover, they acted as potent anti-oxidants, decreasing ROS levels through the activation of the Keap1-Nrf2 pathway, leading to neuroprotection (Fig. 4).

Fig. 4.

Fig. 4

Oenin and Myrtillin protect cells from PrPSc mediated oxidative stress through Keap1-Nrf2 activation. Treatment with either, Oenin or Myrtillin, disrupts the Keap1/Nrf2 dimer, resulting in Nrf2 nuclear translocation. This leads to the activation of a series of anti-oxidant genes, that are related to the glutathione and thioredoxin anti-oxidant systems, NADPH regeneration, iron metabolism, quinone reduction and superoxide neutralization [75,[135], [136], [137]]. Consequently, excessive ROS production is inhibited, and cellular homeostasis is restored, inducing neuroprotection.

Funding

This work was supported by the European Regional Development Fund, 2021–2023, Investment Research Plans for Business Research and Development of Central Macedonia (Grant number: KMP6-0079465).

Financial interests

The authors declare they have no financial interest.

Non-financial interests

None.

CRediT authorship contribution statement

Nikoletta Christoudia: Writing – original draft, Visualization, Validation, Software, Methodology, Investigation, Formal analysis, Data curation, Conceptualization. Nikolaos Bekas: Writing – original draft, Visualization. Eirini Kanata: Writing – review & editing, Writing – original draft, Methodology, Investigation, Data curation. Athanasia Chatziefsthathiou: Visualization, Methodology, Data curation. Spyros Pettas: Visualization, Methodology, Data curation. Korina Karagianni: Visualization, Methodology, Data curation. Susana Margarida Da Silva Correia: Visualization, Software, Methodology, Data curation, Conceptualization. Matthias Schmitz: Writing – review & editing, Resources. Inga Zerr: Writing – review & editing, Resources, Conceptualization. Ioannis Tsamesidis: Software, Methodology, Data curation, Conceptualization. Konstantinos Xanthopoulos: Writing – review & editing, Supervision, Resources, Funding acquisition. Dimitra Dafou: Writing – review & editing, Supervision, Resources, Funding acquisition, Conceptualization. Theodoros Sklaviadis: Writing – review & editing, Supervision, Resources, Funding acquisition.

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgements

We would like to thank Sylvain Lehmann from Centre Hospitalier Universitaire de Montpellier, France for supplying both cell lines, N2a58 and N2a22L. We also appreciate the help in Western Blot from Arketa Mesi and Georgios Alexandros Charisopoulos. The research work was supported by the Hellenic Foundation for Research and Innovation (H.F.R.I) under the 3rd Call for H.F.R.I. PhD Fellowships (Fellowship Number: 6325)

Footnotes

Appendix A

Supplementary data to this article can be found online at https://doi.org/10.1016/j.redox.2024.103133.

Contributor Information

Nikoletta Christoudia, Email: cnikolett@bio.auth.gr.

Nikolaos Bekas, Email: mpekasns@bio.auth.gr.

Eirini Kanata, Email: ekanata@bio.auth.gr.

Athanasia Chatziefsthathiou, Email: chatziea@bio.auth.gr.

Spyros Pettas, Email: spyrospg@bio.auth.gr, spyrospettas@pharm.auth.gr.

Korina Karagianni, Email: korinagk@bio.auth.gr.

Matthias Schmitz, Email: susana.correia@med.uni-goettingen.de, mathias.schmitz@med.uni-goettingen.de.

Inga Zerr, Email: inga.zerr@dzne.de.

Ioannis Tsamesidis, Email: itsamesidis@auth.gr.

Konstantinos Xanthopoulos, Email: xantho@pharm.auth.gr.

Dimitra Dafou, Email: dafoud@bio.auth.gr.

Theodoros Sklaviadis, Email: sklaviad@pharm.auth.gr.

Appendix A. Supplementary data

The following is the Supplementary data to this article:

Multimedia component 1
mmc1.docx (321.8KB, docx)

Data availability

No data was used for the research described in the article.

References

  • 1.Johnson R.T. Prion diseases. Lancet Neurol. 2005;4:635–642. doi: 10.1016/S1474-4422(05)70192-7. [DOI] [PubMed] [Google Scholar]
  • 2.Goldfarb L.G., Brown P. The transmissible spongiform encephalopathies. Annu. Rev. Med. 1995;46:57–65. doi: 10.1146/annurev.med.46.1.57. [DOI] [PubMed] [Google Scholar]
  • 3.Chesebro B. Introduction to the transmissible spongiform encephalopathies or prion diseases. Br. Med. Bull. 2003;66:1–20. doi: 10.1093/bmb/66.1.1. [DOI] [PubMed] [Google Scholar]
  • 4.Prusiner S.B. Prions. Proc. Natl. Acad. Sci. U.S.A. 1998;95:13363–13383. doi: 10.1073/pnas.95.23.13363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Prusiner S.B. Novel proteinaceous infectious particles cause scrapie. Science. 1982;216:136–144. doi: 10.1126/science.6801762. [DOI] [PubMed] [Google Scholar]
  • 6.Shah S.Z.A., Zhao D., Hussain T., Sabir N., Mangi M.H., Yang L. p62-Keap1-NRF2-ARE pathway: a contentious player for selective targeting of autophagy, oxidative stress and mitochondrial dysfunction in prion diseases. Front. Mol. Neurosci. 2018;11:310. doi: 10.3389/fnmol.2018.00310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Tahir W., Zafar S., Llorens F., Arora A.S., Thüne K., Schmitz M., Gotzmann N., Kruse N., Mollenhauer B., Torres J.M., Andréoletti O., Ferrer I., Zerr I. Molecular alterations in the cerebellum of sporadic creutzfeldt–jakob disease subtypes with DJ-1 as a key regulator of oxidative stress. Mol. Neurobiol. 2018;55:517–537. doi: 10.1007/s12035-016-0294-4. [DOI] [PubMed] [Google Scholar]
  • 8.Milhavet O., McMahon H.E.M., Rachidi W., Nishida N., Katamine S., Mangé A., Arlotto M., Casanova D., Riondel J., Favier A., Lehmann S. Prion infection impairs the cellular response to oxidative stress. Proc. Natl. Acad. Sci. U.S.A. 2000;97:13937–13942. doi: 10.1073/pnas.250289197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Arlt S., Kontush A., Zerr I., Buhmann C., Jacobi C., Schröter A., Poser S., Beisiegel U. Increased lipid peroxidation in cerebrospinal fluid and plasma from patients with creutzfeldt–jakob disease. Neurobiol. Dis. 2002;10:150–156. doi: 10.1006/nbdi.2002.0496. [DOI] [PubMed] [Google Scholar]
  • 10.Guentchev M., Voigtländer T., Haberler C., Groschup M.H., Budka H. Evidence for oxidative stress in experimental prion disease. Neurobiol. Dis. 2000;7:270–273. doi: 10.1006/nbdi.2000.0290. [DOI] [PubMed] [Google Scholar]
  • 11.Walter E.D., Stevens D.J., Visconte M.P., Millhauser G.L. The prion protein is a combined zinc and copper binding protein: Zn 2+ alters the distribution of Cu 2+ coordination modes. J. Am. Chem. Soc. 2007;129:15440–15441. doi: 10.1021/ja077146j. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Watt N.T., Taylor D.R., Kerrigan T.L., Griffiths H.H., Rushworth J.V., Whitehouse I.J., Hooper N.M. Prion protein facilitates uptake of zinc into neuronal cells. Nat. Commun. 2012;3:1134. doi: 10.1038/ncomms2135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Brazier M.W., Davies P., Player E., Marken F., Viles J.H., Brown D.R. Manganese binding to the prion protein. J. Biol. Chem. 2008;283:12831–12839. doi: 10.1074/jbc.M709820200. [DOI] [PubMed] [Google Scholar]
  • 14.Singh A., Haldar S., Horback K., Tom C., Zhou L., Meyerson H., Singh N. Prion protein regulates iron transport by functioning as a ferrireductase. JAD. 2013;35:541–552. doi: 10.3233/JAD-130218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Watt N.T., Taylor D.R., Gillott A., Thomas D.A., Perera W.S.S., Hooper N.M. Reactive oxygen species-mediated β-cleavage of the prion protein in the cellular response to oxidative stress. J. Biol. Chem. 2005;280:35914–35921. doi: 10.1074/jbc.M507327200. [DOI] [PubMed] [Google Scholar]
  • 16.Malaisé M., Schätzl H.M., Bürkle A. The octarepeat region of prion protein, but not the TM1 domain, is important for the antioxidant effect of prion protein. Free Radic. Biol. Med. 2008;45:1622–1630. doi: 10.1016/j.freeradbiomed.2008.08.024. [DOI] [PubMed] [Google Scholar]
  • 17.Nadal R.C., Abdelraheim S.R., Brazier M.W., Rigby S.E.J., Brown D.R., Viles J.H. Prion protein does not redox-silence Cu2+, but is a sacrificial quencher of hydroxyl radicals. Free Radic. Biol. Med. 2007;42:79–89. doi: 10.1016/j.freeradbiomed.2006.09.019. [DOI] [PubMed] [Google Scholar]
  • 18.Provansal M., Roche S., Pastore M., Casanova D., Belondrade M., Alais S., Leblanc P., Windl O., Lehmann S. Proteomic consequences of expression and pathological conversion of the prion protein in inducible neuroblastoma N2a cells. Prion. 2010;4:292–301. doi: 10.4161/pri.4.4.13435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Brazier M.W., Lewis V., Ciccotosto G.D., Klug G.M., Lawson V.A., Cappai R., Ironside J.W., Masters C.L., Hill A.F., White A.R., Collins S. Correlative studies support lipid peroxidation is linked to PrPres propagation as an early primary pathogenic event in prion disease. Brain Res. Bull. 2006;68:346–354. doi: 10.1016/j.brainresbull.2005.09.010. [DOI] [PubMed] [Google Scholar]
  • 20.Spiers J.G., Cortina Chen H.-J., Barry T.L., Bourgognon J.-M., Steinert J.R. Redox stress and metal dys-homeostasis appear as hallmarks of early prion disease pathogenesis in mice. Free Radic. Biol. Med. 2022;192:182–190. doi: 10.1016/j.freeradbiomed.2022.09.025. [DOI] [PubMed] [Google Scholar]
  • 21.Thackray A.M., Knight R., Haswell S.J., Bujdoso R., Brown D.R. Metal imbalance and compromised antioxidant function are early changes in prion disease. Biochem. J. 2002;362:253–258. doi: 10.1042/bj3620253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Sorce S., Nuvolone M., Keller A., Falsig J., Varol A., Schwarz P., Bieri M., Budka H., Aguzzi A. The role of the NADPH oxidase NOX2 in prion pathogenesis. PLoS Pathog. 2014;10 doi: 10.1371/journal.ppat.1004531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Arnould H., Baudouin V., Baudry A., Ribeiro L.W., Ardila-Osorio H., Pietri M., Caradeuc C., Soultawi C., Williams D., Alvarez M., Crozet C., Djouadi F., Laforge M., Bertho G., Kellermann O., Launay J.-M., Schmitt-Ulms G., Schneider B. Loss of prion protein control of glucose metabolism promotes neurodegeneration in model of prion diseases. PLoS Pathog. 2021;17 doi: 10.1371/journal.ppat.1009991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Yun S.-W., Gerlach M., Riederer P., Klein M.A. Oxidative stress in the brain at early preclinical stages of mouse scrapie. Exp. Neurol. 2006;201:90–98. doi: 10.1016/j.expneurol.2006.03.025. [DOI] [PubMed] [Google Scholar]
  • 25.Jalland C.M.O., Benestad S.L., Ersdal C., Scheffler K., Suganthan R., Nakabeppu Y., Eide L., Bjørås M., Tranulis M.A. Accelerated clinical course of prion disease in mice compromised in repair of oxidative DNA damage. Free Radic. Biol. Med. 2014;68:1–7. doi: 10.1016/j.freeradbiomed.2013.11.013. [DOI] [PubMed] [Google Scholar]
  • 26.Akhtar S., Grizenkova J., Wenborn A., Hummerich H., Fernandez De Marco M., Brandner S., Collinge J., Lloyd S.E. Sod1 deficiency reduces incubation time in mouse models of prion disease. PLoS One. 2013;8 doi: 10.1371/journal.pone.0054454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Kim M.-J., Kim H.-J., Jang B., Kim H.-J., Mostafa M.N., Park S.-J., Kim Y.-S., Choi E.-K. Impairment of neuronal mitochondrial quality control in prion-induced neurodegeneration. Cells. 2022;11:2744. doi: 10.3390/cells11172744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Moon J.-H., Hong J.-M., Park S.-Y. Calcineurin activation by prion protein induces neurotoxicity via mitochondrial reactive oxygen Species. Oxid. Med. Cell. Longev. 2021;2021:1–15. doi: 10.1155/2021/5572129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Yang D., Li J., Li Z., Zhao M., Wang D., Sun Z., Wen P., Gou F., Dai Y., Ji Y., Li W., Zhao D., Yang L. Cardiolipin externalization mediates prion protein (PrP) peptide 106–126-associated mitophagy and mitochondrial dysfunction. Front. Mol. Neurosci. 2023;16 doi: 10.3389/fnmol.2023.1163981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Spiers J.G., Chen H.-J.C., Steinert J.R. Redox mechanisms and their pathological role in prion diseases: the road to ruin. PLoS Pathog. 2023;19 doi: 10.1371/journal.ppat.1011309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Morén C., deSouza R.M., Giraldo D.M., Uff C. Antioxidant therapeutic strategies in neurodegenerative diseases. Int. J. Math. Stat. 2022;23:9328. doi: 10.3390/ijms23169328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Olufunmilayo E.O., Gerke-Duncan M.B., Holsinger R.M.D. Oxidative stress and antioxidants in neurodegenerative disorders. Antioxidants. 2023;12:517. doi: 10.3390/antiox12020517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Alappat B., Alappat J. Anthocyanin pigments: beyond aesthetics. Molecules. 2020;25:5500. doi: 10.3390/molecules25235500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Chen J., Xu B., Sun J., Jiang X., Bai W. Anthocyanin supplement as a dietary strategy in cancer prevention and management: a comprehensive review. Crit. Rev. Food Sci. Nutr. 2022;62:7242–7254. doi: 10.1080/10408398.2021.1913092. [DOI] [PubMed] [Google Scholar]
  • 35.Nascimento R.D.P.D., Machado A.P.D.F. The preventive and therapeutic effects of anthocyanins on colorectal cancer: a comprehensive review based on up-to-date experimental studies. Food Res. Int. 2023;170 doi: 10.1016/j.foodres.2023.113028. [DOI] [PubMed] [Google Scholar]
  • 36.Rabelo A.C.S., Guerreiro C.D.A., Shinzato V.I., Ong T.P., Noratto G. Anthocyanins reduce cell invasion and migration through akt/mTOR downregulation and apoptosis activation in triple-negative breast cancer cells: a systematic review and meta-analysis. Cancers. 2023;15:2300. doi: 10.3390/cancers15082300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Lin B., Gong C., Song H., Cui Y. Effects of anthocyanins on the prevention and treatment of cancer. Br. J. Pharmacol. 2017;174:1226–1243. doi: 10.1111/bph.13627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Krga I., Milenkovic D. Anthocyanins: from sources and bioavailability to cardiovascular-health benefits and molecular mechanisms of action. J. Agric. Food Chem. 2019;67:1771–1783. doi: 10.1021/acs.jafc.8b06737. [DOI] [PubMed] [Google Scholar]
  • 39.Reis J.F., Monteiro V.V.S., De Souza Gomes R., Do Carmo M.M., Da Costa G.V., Ribera P.C., Monteiro M.C. Action mechanism and cardiovascular effect of anthocyanins: a systematic review of animal and human studies. J. Transl. Med. 2016;14:315. doi: 10.1186/s12967-016-1076-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Laudani S., Godos J., Di Domenico F.M., Barbagallo I., Randazzo C.L., Leggio G.M., Galvano F., Grosso G. Anthocyanin effects on vascular and endothelial health: evidence from clinical trials and role of gut microbiota metabolites. Antioxidants. 2023;12:1773. doi: 10.3390/antiox12091773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Merecz-Sadowska A., Sitarek P., Kowalczyk T., Zajdel K., Jęcek M., Nowak P., Zajdel R. Food anthocyanins: malvidin and its glycosides as promising antioxidant and anti-inflammatory agents with potential health benefits. Nutrients. 2023;15:3016. doi: 10.3390/nu15133016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Zaa C.A., Marcelo Á.J., An Z., Medina-Franco J.L., Velasco-Velázquez M.A. Anthocyanins: molecular aspects on their neuroprotective activity. Biomolecules. 2023;13:1598. doi: 10.3390/biom13111598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Suresh S., Begum R.F., Singh S, C. V A. Anthocyanin as a therapeutic in Alzheimer's disease: a systematic review of preclinical evidences. Ageing Res. Rev. 2022;76 doi: 10.1016/j.arr.2022.101595. [DOI] [PubMed] [Google Scholar]
  • 44.Winter A.N., Bickford P.C. Anthocyanins and their metabolites as therapeutic agents for neurodegenerative disease. Antioxidants. 2019;8:333. doi: 10.3390/antiox8090333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Ullah R., Khan M., Shah S.A., Saeed K., Kim M.O. Natural antioxidant anthocyanins—a hidden therapeutic candidate in metabolic disorders with major focus in neurodegeneration. Nutrients. 2019;11:1195. doi: 10.3390/nu11061195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Li P., Feng D., Yang D., Li X., Sun J., Wang G., Tian L., Jiang X., Bai W. Protective effects of anthocyanins on neurodegenerative diseases. Trends Food Sci. Technol. 2021;117:205–217. doi: 10.1016/j.tifs.2021.05.005. [DOI] [Google Scholar]
  • 47.He F., Liang N.-N., Mu L., Pan Q.-H., Wang J., Reeves M.J., Duan C.-Q. Anthocyanins and their variation in red wines I. Monomeric anthocyanins and their color expression. Molecules. 2012;17:1571–1601. doi: 10.3390/molecules17021571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Sun B., Li F., Zhang X., Wang W., Shao J., Zheng Y. Delphinidin-3- O -glucoside, an active compound of Hibiscus sabdariffa calyces, inhibits oxidative stress and inflammation in rabbits with atherosclerosis. Pharmaceut. Biol. 2022;60:247–254. doi: 10.1080/13880209.2021.2017469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Xie X., Zhao R., Shen G.X. Influence of delphinidin-3-glucoside on oxidized low-density lipoprotein-induced oxidative stress and apoptosis in cultured endothelial cells. J. Agric. Food Chem. 2012;60:1850–1856. doi: 10.1021/jf204461z. [DOI] [PubMed] [Google Scholar]
  • 50.Hu J., Li X., Wu N., Zhu C., Jiang X., Yuan K., Li Y., Sun J., Bai W. Anthocyanins prevent AAPH-induced steroidogenesis disorder in leydig cells by counteracting oxidative stress and StAR abnormal expression in a structure-dependent manner. Antioxidants. 2023;12:508. doi: 10.3390/antiox12020508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Yacout S.M., Gaillard E.R. The anthocyanins, Oenin and callistephin, protect RPE cells against oxidative stress. Photochem. Photobiol. 2017;93:590–599. doi: 10.1111/php.12683. [DOI] [PubMed] [Google Scholar]
  • 52.Herrera-Balandrano D.D., Chai Z., Hutabarat R.P., Beta T., Feng J., Ma K., Li D., Huang W. Hypoglycemic and hypolipidemic effects of blueberry anthocyanins by AMPK activation: in vitro and in vivo studies. Redox Biol. 2021;46 doi: 10.1016/j.redox.2021.102100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Huang W.-Y., Wu H., Li D.-J., Song J.-F., Xiao Y.-D., Liu C.-Q., Zhou J.-Z., Sui Z.-Q. Protective effects of blueberry anthocyanins against H 2 O 2 -induced oxidative injuries in human retinal pigment epithelial cells. J. Agric. Food Chem. 2018;66:1638–1648. doi: 10.1021/acs.jafc.7b06135. [DOI] [PubMed] [Google Scholar]
  • 54.Nishida N., Harris D.A., Vilette D., Laude H., Frobert Y., Grassi J., Casanova D., Milhavet O., Lehmann S. Successful transmission of three mouse-adapted scrapie strains to murine neuroblastoma cell lines overexpressing wild-type mouse prion protein. J. Virol. 2000;74:320–325. doi: 10.1128/JVI.74.1.320-325.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Taguchi Y., Lu L., Marrero-Winkens C., Otaki H., Nishida N., Schatzl H.M. Disulfide-crosslink scanning reveals prion–induced conformational changes and prion strain–specific structures of the pathological prion protein PrPSc. J. Biol. Chem. 2018;293:12730–12740. doi: 10.1074/jbc.RA117.001633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Ishibashi D., Nakagaki T., Ishikawa T., Atarashi R., Watanabe K., Cruz F.A., Hamada T., Nishida N. Structure-based drug discovery for prion disease using a novel binding simulation. EBioMedicine. 2016;9:238–249. doi: 10.1016/j.ebiom.2016.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Pankiewicz J.E., Lizińczyk A.M., Franco L.A., Diaz J.R., Martá-Ariza M., Sadowski M.J. Absence of Apolipoprotein E is associated with exacerbation of prion pathology and promotes microglial neurodegenerative phenotype. Acta Neuropathol Commun. 2021;9:157. doi: 10.1186/s40478-021-01261-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Uchiyama K., Hara H., Chida J., Pasiana A.D., Imamura M., Mori T., Takatsuki H., Atarashi R., Sakaguchi S. Ethanolamine is a new anti-prion compound. Int. J. Math. Stat. 2021;22 doi: 10.3390/ijms222111742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Takatsuki H., Imamura M., Mori T., Atarashi R. Pentosan polysulfate induces low-level persistent prion infection keeping measurable seeding activity without PrP-res detection in Fukuoka-1 infected cell cultures. Sci. Rep. 2022;12:7923. doi: 10.1038/s41598-022-12049-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Wagner W., Reuter A., Hüller P., Löwer J., Wessler S. Peroxiredoxin 6 promotes upregulation of the prion protein (PrP) in neuronal cells of prion-infected mice. Cell Commun. Signal. 2012;10:38. doi: 10.1186/1478-811X-10-38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Prior M., Lehmann S., Sy M.-S., Molloy B., McMahon H.E.M. Cyclodextrins inhibit replication of scrapie prion protein in cell culture. J. Virol. 2007;81:11195–11207. doi: 10.1128/JVI.02559-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Féraudet C., Morel N., Simon S., Volland H., Frobert Y., Créminon C., Vilette D., Lehmann S., Grassi J. Screening of 145 anti-PrP monoclonal antibodies for their capacity to inhibit PrPSc replication in infected cells. J. Biol. Chem. 2005;280:11247–11258. doi: 10.1074/jbc.M407006200. [DOI] [PubMed] [Google Scholar]
  • 63.Alais S., Simoes S., Baas D., Lehmann S., Raposo G., Darlix J.L., Leblanc P. Mouse neuroblastoma cells release prion infectivity associated with exosomal vesicles. Biol. Cell. 2008;100:603–618. doi: 10.1042/BC20080025. [DOI] [PubMed] [Google Scholar]
  • 64.Ayrolles-Torro A., Imberdis T., Torrent J., Toupet K., Baskakov I.V., Poncet-Montange G., Grégoire C., Roquet-Baneres F., Lehmann S., Rognan D., Pugnière M., Verdier J.-M., Perrier V. Oligomeric-induced activity by thienyl pyrimidine compounds traps prion infectivity. J. Neurosci. 2011;31:14882–14892. doi: 10.1523/JNEUROSCI.0547-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Rooprai H.K., Lawrence P., Keshavarz S., Yashod P., Gullan R.W., Selway R.P., Davies D. DRAQ7 as an alternative to MTT assay for measuring viability of glioma cells treated with polyphenols. Anticancer Res. 2020;40:5427–5436. doi: 10.21873/anticanres.14553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Valadez-Vega C., Lugo-Magaña O., Mendoza-Guzmán L., Villagómez-Ibarra J.R., Velasco-Azorsa R., Bautista M., Betanzos-Cabrera G., Morales-González J.A., Madrigal-Santillán E.O. Antioxidant activity and anticarcinogenic effect of extracts from bouvardia ternifolia (cav.) schltdl. Life. 2023;13:2319. doi: 10.3390/life13122319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Schmitz M., Cramm M., Llorens F., Candelise N., Müller-Cramm D., Varges D., Schulz-Schaeffer W.J., Zafar S., Zerr I. Application of an in vitro-amplification assay as a novel pre-screening test for compounds inhibiting the aggregation of prion protein scrapie. Sci. Rep. 2016;6 doi: 10.1038/srep28711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Yu C., Xiao J.-H. The keap1-Nrf2 system: a mediator between oxidative stress and aging. Oxid. Med. Cell. Longev. 2021;2021:1–16. doi: 10.1155/2021/6635460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Shah S.A., Amin F.U., Khan M., Abid M.N., Rehman S.U., Kim T.H., Kim M.W., Kim M.O. Anthocyanins abrogate glutamate-induced AMPK activation, oxidative stress, neuroinflammation, and neurodegeneration in postnatal rat brain. J. Neuroinflammation. 2016;13:286. doi: 10.1186/s12974-016-0752-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Chao C.-C., Huang C.-L., Cheng J.-J., Chiou C.-T., Lee I.-J., Yang Y.-C., Hsu T.-H., Yei C.-E., Lin P.-Y., Chen J.-J., Huang N.-K. SRT1720 as an SIRT1 activator for alleviating paraquat-induced models of Parkinson's disease. Redox Biol. 2022;58 doi: 10.1016/j.redox.2022.102534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Martín-Montañez E., Valverde N., Ladrón De Guevara-Miranda D., Lara E., Romero-Zerbo Y.S., Millon C., Boraldi F., Ávila-Gámiz F., Pérez-Cano A.M., Garrido-Gil P., Labandeira-Garcia J.L., Santin L.J., Pavia J., Garcia-Fernandez M. Insulin-like growth factor II prevents oxidative and neuronal damage in cellular and mice models of Parkinson's disease. Redox Biol. 2021;46 doi: 10.1016/j.redox.2021.102095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Karagianni K., Pettas S., Kanata E., Lioulia E., Thune K., Schmitz M., Tsamesidis I., Lymperaki E., Xanthopoulos K., Sklaviadis T., Dafou D. Carnosic Acid and carnosol display antioxidant and anti-prion properties in in vitro and cell-free models of prion diseases. Antioxidants. 2022;11:726. doi: 10.3390/antiox11040726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Sotolongo K., Ghiso J., Rostagno A. Nrf2 activation through the PI3K/GSK-3 axis protects neuronal cells from Aβ-mediated oxidative and metabolic damage. Alz Res Therapy. 2020;12:13. doi: 10.1186/s13195-019-0578-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Fragoulis A., Siegl S., Fendt M., Jansen S., Soppa U., Brandenburg L.-O., Pufe T., Weis J., Wruck C.J. Oral administration of methysticin improves cognitive deficits in a mouse model of Alzheimer's disease. Redox Biol. 2017;12:843–853. doi: 10.1016/j.redox.2017.04.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Amin F.U., Shah S.A., Badshah H., Khan M., Kim M.O. Anthocyanins encapsulated by PLGA@PEG nanoparticles potentially improved its free radical scavenging capabilities via p38/JNK pathway against Aβ1–42-induced oxidative stress. J. Nanobiotechnol. 2017;15:12. doi: 10.1186/s12951-016-0227-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Wang C.-Y., Zhang Q., Xun Z., Yuan L., Li R., Li X., Tian S.-Y., Xin N., Xu Y. Increases of iASPP-Keap1 interaction mediated by syringin enhance synaptic plasticity and rescue cognitive impairments via stabilizing Nrf2 in Alzheimer's models. Redox Biol. 2020;36 doi: 10.1016/j.redox.2020.101672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Brackhan M., Arribas-Blazquez M., Lastres-Becker I. Aging, NRF2, and TAU: a perfect match for neurodegeneration? Antioxidants. 2023;12:1564. doi: 10.3390/antiox12081564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Baral P.K., Yin J., Aguzzi A., James M.N.G. Transition of the prion protein from a structured cellular form (PrP C) to the infectious scrapie agent (PrP Sc) Protein Sci. 2019;28:2055–2063. doi: 10.1002/pro.3735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Wille H., Requena J. The structure of PrPSc prions. Pathogens. 2018;7:20. doi: 10.3390/pathogens7010020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Riesner D. Biochemistry and structure of PrPC and PrPSc. Br. Med. Bull. 2003;66:21–33. doi: 10.1093/bmb/66.1.21. [DOI] [PubMed] [Google Scholar]
  • 81.Wang F., Wang X., Abskharon R., Ma J. Prion infectivity is encoded exclusively within the structure of proteinase K-resistant fragments of synthetically generated recombinant PrPSc. Acta Neuropathol Commun. 2018;6:30. doi: 10.1186/s40478-018-0534-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Schmitz M., Cramm M., Llorens F., Müller-Cramm D., Collins S., Atarashi R., Satoh K., Orrù C.D., Groveman B.R., Zafar S., Schulz-Schaeffer W.J., Caughey B., Zerr I. The real-time quaking-induced conversion assay for detection of human prion disease and study of other protein misfolding diseases. Nat. Protoc. 2016;11:2233–2242. doi: 10.1038/nprot.2016.120. [DOI] [PubMed] [Google Scholar]
  • 83.Zerr I. RT-QuIC for detection of prodromal α-synucleinopathies. Lancet Neurol. 2021;20:165–166. doi: 10.1016/S1474-4422(21)00036-3. [DOI] [PubMed] [Google Scholar]
  • 84.Zerr I. Laboratory diagnosis of creutzfeldt–jakob disease. N. Engl. J. Med. 2022;386:1345–1350. doi: 10.1056/NEJMra2119323. [DOI] [PubMed] [Google Scholar]
  • 85.Saijo E., Ghetti B., Zanusso G., Oblak A., Furman J.L., Diamond M.I., Kraus A., Caughey B. Ultrasensitive and selective detection of 3-repeat tau seeding activity in Pick disease brain and cerebrospinal fluid. Acta Neuropathol. 2017;133:751–765. doi: 10.1007/s00401-017-1692-z. [DOI] [PubMed] [Google Scholar]
  • 86.Kim D.H., Kim J., Lee H., Lee D., Im S.M., Kim Y.E., Yoo M., Cheon Y.-P., Bartz J.C., Son Y.-J., Choi E.-K., Kim Y.-S., Jeon J.-H., Kim H.S., Lee S., Ryou C., Nam T. Synthesis and anti-prion aggregation activity of acylthiosemicarbazide analogues. J. Enzym. Inhib. Med. Chem. 2023;38 doi: 10.1080/14756366.2023.2191164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Ferreira N.C., Ascari L.M., Hughson A.G., Cavalheiro G.R., Góes C.F., Fernandes P.N., Hollister J.R., Da Conceição R.A., Silva D.S., Souza A.M.T., Barbosa M.L.C., Lara F.A., Martins R.A.P., Caughey B., Cordeiro Y. A promising antiprion trimethoxychalcone binds to the globular domain of the cellular prion protein and changes its cellular location. Antimicrob. Agents Chemother. 2018;62 doi: 10.1128/AAC.01441-17. e01441-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Colini Baldeschi A., Zattoni M., Vanni S., Nikolic L., Ferracin C., La Sala G., Summa M., Bertorelli R., Bertozzi S.M., Giachin G., Carloni P., Bolognesi M.L., De Vivo M., Legname G. Innovative non-PrP-targeted drug strategy designed to enhance prion clearance. J. Med. Chem. 2022;65:8998–9010. doi: 10.1021/acs.jmedchem.2c00205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Shi S., Wagner J., Mitteregger-Kretzschmar G., Ryazanov S., Leonov A., Griesinger C., Giese A. Quantitative real-time quaking-induced conversion allows monitoring of disease-modifying therapy in the urine of prion-infected mice. J. Neuropathol. Exp. Neurol. 2015;74:924–933. doi: 10.1097/NEN.0000000000000233. [DOI] [PubMed] [Google Scholar]
  • 90.Imberdis T., Heeres J.T., Yueh H., Fang C., Zhen J., Rich C.B., Glicksman M., Beeler A.B., Harris D.A. Identification of anti-prion compounds using a novel cellular assay. J. Biol. Chem. 2016;291:26164–26176. doi: 10.1074/jbc.M116.745612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Poleggi A., Baiardi S., Ladogana A., Parchi P. The use of real-time quaking-induced conversion for the diagnosis of human prion diseases. Front. Aging Neurosci. 2022;14 doi: 10.3389/fnagi.2022.874734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Candelise N., Baiardi S., Franceschini A., Rossi M., Parchi P. Towards an improved early diagnosis of neurodegenerative diseases: the emerging role of in vitro conversion assays for protein amyloids. Acta Neuropathol Commun. 2020;8:117. doi: 10.1186/s40478-020-00990-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Jucker M., Walker L.C. Self-propagation of pathogenic protein aggregates in neurodegenerative diseases. Nature. 2013;501:45–51. doi: 10.1038/nature12481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Golde T.E., Borchelt D.R., Giasson B.I., Lewis J. Thinking laterally about neurodegenerative proteinopathies. J. Clin. Invest. 2013;123:1847–1855. doi: 10.1172/JCI66029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Brundin P., Melki R., Kopito R. Prion-like transmission of protein aggregates in neurodegenerative diseases. Nat. Rev. Mol. Cell Biol. 2010;11:301–307. doi: 10.1038/nrm2873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Frost B., Diamond M.I. Prion-like mechanisms in neurodegenerative diseases. Nat. Rev. Neurosci. 2010;11:155–159. doi: 10.1038/nrn2786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Baxter P.S., Márkus N.M., Dando O., He X., Al-Mubarak B.R., Qiu J., Hardingham G.E. Targeted de-repression of neuronal Nrf2 inhibits α-synuclein accumulation. Cell Death Dis. 2021;12:218. doi: 10.1038/s41419-021-03507-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Gan L., Vargas M.R., Johnson D.A., Johnson J.A. Astrocyte-Specific overexpression of Nrf2 delays motor pathology and synuclein aggregation throughout the CNS in the alpha-synuclein mutant (A53T) mouse model. J. Neurosci. 2012;32:17775–17787. doi: 10.1523/JNEUROSCI.3049-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Bi M., Du X., Xiao X., Dai Y., Jiao Q., Chen X., Zhang L., Jiang H. Deficient immunoproteasome assembly drives gain of α-synuclein pathology in Parkinson's disease. Redox Biol. 2021;47 doi: 10.1016/j.redox.2021.102167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Skibinski G., Hwang V., Ando D.M., Daub A., Lee A.K., Ravisankar A., Modan S., Finucane M.M., Shaby B.A., Finkbeiner S. Nrf2 mitigates LRRK2- and α-synuclein–induced neurodegeneration by modulating proteostasis. Proc. Natl. Acad. Sci. U.S.A. 2017;114:1165–1170. doi: 10.1073/pnas.1522872114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Jiwaji Z., Tiwari S.S., Avilés-Reyes R.X., Hooley M., Hampton D., Torvell M., Johnson D.A., McQueen J., Baxter P., Sabari-Sankar K., Qiu J., He X., Fowler J., Febery J., Gregory J., Rose J., Tulloch J., Loan J., Story D., McDade K., Smith A.M., Greer P., Ball M., Kind P.C., Matthews P.M., Smith C., Dando O., Spires-Jones T.L., Johnson J.A., Chandran S., Hardingham G.E. Reactive astrocytes acquire neuroprotective as well as deleterious signatures in response to Tau and Aß pathology. Nat. Commun. 2022;13:135. doi: 10.1038/s41467-021-27702-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Zhang X., Wang J., Gong G., Ma R., Xu F., Yan T., Wu B., Jia Y. Spinosin inhibits aβ 1-42 production and aggregation via activating Nrf2/HO-1 pathway. Biomolecules & Therapeutics. 2020;28:259–266. doi: 10.4062/biomolther.2019.123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Abou El-ezz D., Maher A., Sallam N., El-brairy A., Kenawy S. Trans-cinnamaldehyde modulates hippocampal Nrf2 factor and inhibits amyloid beta aggregation in LPS-induced neuroinflammation mouse model. Neurochem. Res. 2018;43:2333–2342. doi: 10.1007/s11064-018-2656-y. [DOI] [PubMed] [Google Scholar]
  • 104.Eftekharzadeh B., Maghsoudi N., Khodagholi F. Stabilization of transcription factor Nrf2 by tBHQ prevents oxidative stress-induced amyloid β formation in NT2N neurons. Biochimie. 2010;92:245–253. doi: 10.1016/j.biochi.2009.12.001. [DOI] [PubMed] [Google Scholar]
  • 105.Jo C., Gundemir S., Pritchard S., Jin Y.N., Rahman I., Johnson G.V.W. Nrf2 reduces levels of phosphorylated tau protein by inducing autophagy adaptor protein NDP52. Nat. Commun. 2014;5:3496. doi: 10.1038/ncomms4496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Kim S., Choi K.J., Cho S.-J., Yun S.-M., Jeon J.-P., Koh Y.H., Song J., Johnson G.V.W., Jo C. Fisetin stimulates autophagic degradation of phosphorylated tau via the activation of TFEB and Nrf2 transcription factors. Sci. Rep. 2016;6 doi: 10.1038/srep24933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Tang M., Ji C., Pallo S., Rahman I., Johnson G.V.W. Nrf2 mediates the expression of BAG3 and autophagy cargo adaptor proteins and tau clearance in an age-dependent manner. Neurobiol. Aging. 2018;63:128–139. doi: 10.1016/j.neurobiolaging.2017.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Lastres-Becker I., Ulusoy A., Innamorato N.G., Sahin G., Rábano A., Kirik D., Cuadrado A. α-Synuclein expression and Nrf2 deficiency cooperate to aggravate protein aggregation, neuronal death and inflammation in early-stage Parkinson's disease. Hum. Mol. Genet. 2012;21:3173–3192. doi: 10.1093/hmg/dds143. [DOI] [PubMed] [Google Scholar]
  • 109.Joshi G., Gan K.A., Johnson D.A., Johnson J.A. Increased Alzheimer's disease–like pathology in the APP/PS1ΔE9 mouse model lacking Nrf2 through modulation of autophagy. Neurobiol. Aging. 2015;36:664–679. doi: 10.1016/j.neurobiolaging.2014.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Rojo A.I., Pajares M., Rada P., Nuñez A., Nevado-Holgado A.J., Killik R., Van Leuven F., Ribe E., Lovestone S., Yamamoto M., Cuadrado A. NRF2 deficiency replicates transcriptomic changes in Alzheimer's patients and worsens APP and TAU pathology. Redox Biol. 2017;13:444–451. doi: 10.1016/j.redox.2017.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Tang Z., Hu B., Zang F., Wang J., Zhang X., Chen H. Nrf2 drives oxidative stress-induced autophagy in nucleus pulposus cells via a Keap1/Nrf2/p62 feedback loop to protect intervertebral disc from degeneration. Cell Death Dis. 2019;10:510. doi: 10.1038/s41419-019-1701-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Pajares M., Rojo A.I., Arias E., Díaz-Carretero A., Cuervo A.M., Cuadrado A. Transcription factor NFE2L2/NRF2 modulates chaperone-mediated autophagy through the regulation of LAMP2A. Autophagy. 2018;14:1310–1322. doi: 10.1080/15548627.2018.1474992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Pajares M., Jiménez-Moreno N., García-Yagüe Á.J., Escoll M., De Ceballos M.L., Van Leuven F., Rábano A., Yamamoto M., Rojo A.I., Cuadrado A. Transcription factor NFE2L2/NRF2 is a regulator of macroautophagy genes. Autophagy. 2016;12:1902–1916. doi: 10.1080/15548627.2016.1208889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Jena K.K., Kolapalli S.P., Mehto S., Nath P., Das B., Sahoo P.K., Ahad A., Syed G.H., Raghav S.K., Senapati S., Chauhan S., Chauhan S. TRIM16 controls assembly and degradation of protein aggregates by modulating the p62‐NRF2 axis and autophagy. EMBO J. 2018;37 doi: 10.15252/embj.201798358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Zimmermann K., Baldinger J., Mayerhofer B., Atanasov A.G., Dirsch V.M., Heiss E.H. Activated AMPK boosts the Nrf2/HO-1 signaling axis—a role for the unfolded protein response. Free Radic. Biol. Med. 2015;88:417–426. doi: 10.1016/j.freeradbiomed.2015.03.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Mukaigasa K., Tsujita T., Nguyen V.T., Li L., Yagi H., Fuse Y., Nakajima-Takagi Y., Kato K., Yamamoto M., Kobayashi M. Nrf2 activation attenuates genetic endoplasmic reticulum stress induced by a mutation in the phosphomannomutase 2 gene in zebrafish. Proc. Natl. Acad. Sci. U.S.A. 2018;115:2758–2763. doi: 10.1073/pnas.1714056115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Cullinan S.B., Zhang D., Hannink M., Arvisais E., Kaufman R.J., Diehl J.A. Nrf2 is a direct PERK substrate and effector of PERK-dependent cell survival. Mol. Cell Biol. 2003;23:7198–7209. doi: 10.1128/MCB.23.20.7198-7209.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Cullinan S.B., Diehl J.A. Coordination of ER and oxidative stress signaling: the PERK/Nrf2 signaling pathway. Int. J. Biochem. Cell Biol. 2006;38:317–332. doi: 10.1016/j.biocel.2005.09.018. [DOI] [PubMed] [Google Scholar]
  • 119.Dong L., Xu M., Li Y., Xu W., Wu C., Zheng H., Xiao Z., Sun G., Ding L., Li X., Li W., Zhou L., Xia Q. SMURF1 attenuates endoplasmic reticulum stress by promoting the degradation of KEAP1 to activate NRF2 antioxidant pathway. Cell Death Dis. 2023;14:361. doi: 10.1038/s41419-023-05873-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Bruch J., Xu H., Rösler T.W., De Andrade A., Kuhn P., Lichtenthaler S.F., Arzberger T., Winklhofer K.F., Müller U., Höglinger G.U. PERK activation mitigates tau pathology in vitro and in vivo. EMBO Mol. Med. 2017;9:371–384. doi: 10.15252/emmm.201606664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Pajares M., Cuadrado A., Rojo A.I. Modulation of proteostasis by transcription factor NRF2 and impact in neurodegenerative diseases. Redox Biol. 2017;11:543–553. doi: 10.1016/j.redox.2017.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Zhang Z., Zhang L., Zhou L., Lei Y., Zhang Y., Huang C. Redox signaling and unfolded protein response coordinate cell fate decisions under ER stress. Redox Biol. 2019;25 doi: 10.1016/j.redox.2018.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Moreno J.A., Halliday M., Molloy C., Radford H., Verity N., Axten J.M., Ortori C.A., Willis A.E., Fischer P.M., Barrett D.A., Mallucci G.R. Oral treatment targeting the unfolded protein response prevents neurodegeneration and clinical disease in prion-infected mice. Sci. Transl. Med. 2013;5 doi: 10.1126/scitranslmed.3006767. [DOI] [PubMed] [Google Scholar]
  • 124.Yu K.-H., Jheng C.-P., Lee C.-I. Quercetin binding accelerates prion fibrillation into proteinase sensitive and loosely structured amyloids. Biomed. Pharmacother. 2022;151 doi: 10.1016/j.biopha.2022.113177. [DOI] [PubMed] [Google Scholar]
  • 125.Huo Y., Zhao C., Wang Y., Wang S., Mu T., Du W. Roles of Apigenin and Nepetin in the assembly behavior and cytotoxicity of prion neuropeptide PrP106-126. ACS Chem. Neurosci. 2023 doi: 10.1021/acschemneuro.3c00417. acschemneuro.3c00417. [DOI] [PubMed] [Google Scholar]
  • 126.Yu K.-H., Lee C.-I. Quercetin disaggregates prion fibrils and decreases fibril-induced cytotoxicity and oxidative stress. Pharmaceutics. 2020;12:1081. doi: 10.3390/pharmaceutics12111081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Hafner‐Bratkovič I., Gašperšič J., Šmid L.M., Bresjanac M., Jerala R. Curcumin binds to the α‐helical intermediate and to the amyloid form of prion protein – a new mechanism for the inhibition of PrP Sc accumulation. J. Neurochem. 2008;104:1553–1564. doi: 10.1111/j.1471-4159.2007.05105.x. [DOI] [PubMed] [Google Scholar]
  • 128.Lin C.-F., Yu K.-H., Jheng C.-P., Chung R., Lee C.-I. Curcumin reduces amyloid fibrillation of prion protein and decreases reactive oxidative stress. Pathogens. 2013;2:506–519. doi: 10.3390/pathogens2030506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Mehta N., Marwah P., Njus D. Are proteinopathy and oxidative stress two sides of the same coin? Cells. 2019;8:59. doi: 10.3390/cells8010059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Barnham K.J., Masters C.L., Bush A.I. Neurodegenerative diseases and oxidative stress. Nat. Rev. Drug Discov. 2004;3:205–214. doi: 10.1038/nrd1330. [DOI] [PubMed] [Google Scholar]
  • 131.Kawamata H., Manfredi G. Proteinopathies and OXPHOS dysfunction in neurodegenerative diseases. JCB (J. Cell Biol.) 2017;216:3917–3929. doi: 10.1083/jcb.201709172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Zhang X., Zhao D., Wu W., Ali Shah S.Z., Lai M., Yang D., Li J., Guan Z., Li W., Gao H., Zhao H., Zhou X., Yang L. Melatonin regulates mitochondrial dynamics and alleviates neuron damage in prion diseases. Aging. 2020;12:11139–11151. doi: 10.18632/aging.103328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Yang W., Chen C., Chen J., Xia Y., Hu C., Wang L., Wu Y.-Z., Shi Q., Chen Z.-B., Dong X.-P. PrPSc inhibition and cellular protection of DBL on a prion-infected cultured cell via multiple pathways. Mol. Neurobiol. 2022;59:3310–3321. doi: 10.1007/s12035-022-02729-2. [DOI] [PubMed] [Google Scholar]
  • 134.Sun Y., Xu L., Zheng D., Wang J., Liu G., Mo Z., Liu C., Zhang W., Yu J., Xing C., He L., Zhuang C. A potent phosphodiester Keap1-Nrf2 protein-protein interaction inhibitor as the efficient treatment of Alzheimer's disease. Redox Biol. 2023;64 doi: 10.1016/j.redox.2023.102793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Mizunoe Y., Kobayashi M., Sudo Y., Watanabe S., Yasukawa H., Natori D., Hoshino A., Negishi A., Okita N., Komatsu M., Higami Y. Trehalose protects against oxidative stress by regulating the Keap1–Nrf2 and autophagy pathways. Redox Biol. 2018;15:115–124. doi: 10.1016/j.redox.2017.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Villavicencio Tejo F., Quintanilla R.A. Contribution of the Nrf2 pathway on oxidative damage and mitochondrial failure in Parkinson and alzheimer's disease. Antioxidants. 2021;10:1069. doi: 10.3390/antiox10071069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Moratilla-Rivera I., Sánchez M., Valdés-González J.A., Gómez-Serranillos M.P. Natural products as modulators of Nrf2 signaling pathway in neuroprotection. Int. J. Math. Stat. 2023;24:3748. doi: 10.3390/ijms24043748. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Multimedia component 1
mmc1.docx (321.8KB, docx)

Data Availability Statement

No data was used for the research described in the article.


Articles from Redox Biology are provided here courtesy of Elsevier

RESOURCES