Abstract
Because old age is the greatest risk factor for Alzheimer’s disease (AD), it is critical to target the pathological events that link aging to AD in order to develop an efficient treatment that acts upon the primary causes of the disease. One such event might be the activation of oxytosis/ferroptosis, a unique cell death mechanism characterized by mitochondrial dysfunction and lethal lipid peroxidation. Here, a comprehensive library of > 900 natural compounds was screened for protection against oxytosis/ferroptosis in nerve cells with the goal of better understanding the chemical nature of inhibitors of oxytosis/ferroptosis. Although the compounds tested spanned structurally diverse chemical classes from animal, microbial, plant and synthetic origins, a small set of very potent anti-oxytotic/ferroptotic compounds was identified that was highly enriched in plant quinones. The ability of these compounds to protect against oxytosis/ferroptosis strongly correlated with their ability to protect against in vitro ischemia and intracellular amyloid-beta toxicity in nerve cells, indicating that aspects of oxytosis/ferroptosis also underly other toxicities that are relevant to AD. Importantly, the anti-oxytotic/ferroptotic character of the quinone compounds relied on their capacity to target and directly prevent lipid peroxidation in a manner that required the reducing activity of cellular redox enzymes, such as NAD(P)H:quinone oxidoreductase 1 (NQO1) and ferroptosis suppressor protein 1 (FSP1). Because some of the compounds increased the production of total reactive oxygen species while decreasing lipid peroxidation, it appears that the pro-oxidant character of a compound can coexist with an inhibitory effect on lipid peroxidation and, consequently, still prevent oxytosis/ferroptosis. These findings have significant implications for the understanding of oxytosis/ferroptosis and open new approaches to the development of future neurotherapies.
Keywords: Natural products, quinones, aging, lipid peroxidation, oxytosis/ferroptosis, neurodegenerative disease, drug discovery
Graphical Abstract

1. Introduction
There are no drugs for Alzheimer’s disease (AD) that prevent or slow down its progression. This may be due to a lack of therapies that target the primary drivers of the disease. Given that old age is the greatest risk factor for AD [1], our labs have been focused on identifying the pathological processes associated with aging that are detrimental to brain function and may be driving AD. Our efforts have led to the identification of a unique mechanism of neurodegeneration called oxytosis/ferroptosis [2, 3].
Oxytosis/ferroptosis is a form of non-apoptotic regulated cell death characterized by glutathione (GSH) depletion and dysregulated production of reactive oxygen species (ROS) from mitochondria that results in lethal lipid peroxidation [2–5]. All of these changes are detected in the aging brain and, to a larger extent, in AD [4]. Importantly, oxytosis/ferroptosis can manifest itself over a lengthy time period before the cells die, thereby offering a window for therapeutic intervention. We have shown that inhibitors of oxytosis/ferroptosis are protective in transgenic mouse models of AD and prevent the development of dementia in the SAMP8 mouse model of accelerated aging [6–9].
At the molecular level, oxytosis/ferroptosis can be triggered by glutamate which inhibits cystine uptake via system xc− and subsequently depletes intracellular GSH [2, 4, 5]. This leads to inhibition of the GSH-dependent enzyme GSH peroxidase 4 (GPX4) and activation of lipoxygenases (LOXs) [10]. GPX4 can also be directly inhibited by the chemical RSL3. Based on these two insults and using a nerve cell line-based assay to screen compounds for their protection against oxytosis/ferroptosis, we have successfully identified several drug candidates for AD [3, 11].
Here, we used this assay to screen a commercial library of > 900 natural compounds. We identified a set of potent inhibitors of oxytosis/ferroptosis, most of which were previously unreported as anti-oxytotic/ferroptotic molecules. Importantly, our data indicate that oxytosis/ferroptosis may be a common pathway in other toxicities relevant to dementia and provide clues about the chemistry of anti-oxytotic/ferroptotic compounds.
2. Materials and methods
All reagents were obtained from Sigma-Aldrich (St. Louis, MO, United States), unless otherwise stated. The library of natural compounds (cat. No. HY-L021) was purchased from MedChemExpress (NJ, United States).
2.1. Cell culture
HT22 mouse hippocampal nerve cells and MC65 human neuroblastoma cells were cultured in high-glucose Dulbecco’s modified Eagle’s medium (DMEM) (Invitrogen, Carlsbad, CA, United States) supplemented with 10% fetal calf serum (FCS) (Hyclone, Logan, UT, United States), and incubated at 37°C in an atmosphere with 10% CO2.
2.2. Phenotypic screening assays
2.2.1. Oxytosis/ferroptosis (HT22 cells)
5×103 HT22 mouse hippocampal nerve cells were plated per well in 96 well plates. After 24 h, the medium was exchanged with fresh medium and 10 mM glutamate or 200 nM RSL3 were added alone or in combination with the natural compounds at the indicated concentrations. 24 h later, the cellular viability was measured by the 3-(4, 5-dimethylthiazolyl-2)- 2,5-diphenyltetrazolium bromide (MTT) assay, as previously described [12]. Cell survival was confirmed by visual inspection of the wells. After the primary screening, compounds yielding ≥ 65% cell survival against at least one of the insults were selected to be tested at a wider range of concentrations (58 compounds), and those compounds displaying ≥ 75% protection against both insults in this secondary screening were selected for further analysis (20 compounds).
2.2.2. Intracellular Aβ toxicity (MC65 cells)
MC65 cells were regularly grown in high glucose DMEM supplemented with 10% FCS and 2 μg/ml tetracycline [13]. For the assay, cells were dissociated, plated at 4×105 cells per 35 mm tissue culture dish and grown for 24 h. The next day, the cells were washed with PBS and placed in Opti-minimal essential media (Opti-MEM, Invitrogen) in the presence (no induction) or absence (APP-C99 induced) of 2 μg/ml tetracycline in combination with the natural compounds. At day 3, the control cells in the absence of tetracycline were dead, and cell viability was determined by the MTT assay and confirmed by visual inspection.
2.2.3. Protection against energy loss (HT22 cells)
HT22 cells were seeded onto 96 well plates as described in the oxytosis/ferroptosis assay. The medium was exchanged 24 h later with fresh medium and the cells were treated with 20 μM IAA alone (which results in 90–95% cell death) or in combination with the compounds at the indicated concentrations, as previously described [14]. After 2 h, the medium was replaced with fresh medium without IAA but containing the compounds. 24 h later, the cellular viability was measured by the MTT assay.
2.3. Measurement of total glutathione (tGSH)
For measurement of tGSH, 3×105 HT22 cells were plated in 60 mm dishes. After 24 h, the medium was exchanged with fresh medium, and glutamate and compounds were added. The cells were treated for 24 h and then scraped into ice-cold PBS, and 10% sulfosalicylic acid was added at a final concentration of 3.3%. GSH was determined by the recycling assay based on the reduction of 5,5-dithiobis(2-nitrobenzoic acid) with glutathione reductase and NADPH [15] and normalized to protein recovered from the acid-precipitated pellet by treatment with 0.2 N NaOH at 37 °C overnight and measured by the bicinchoninic acid assay (Pierce, Rockford, IL, USA).
2.4. Measurement of ROS and lipid peroxidation levels by flow cytometry
7.5×103 HT22 cells per well were seeded in 96 well plates. After 24 h, the compounds were added in the presence or absence of glutamate (10 mM). The concentration of each compound was chosen based on the lowest dose yielding maximal protection. After 6 h, the media was aspirated and 100 μl per well of BODIPY™ 581/591 C11 (cat No. D3861, Invitrogen) (1μM) or CellROX™ Green (cat No. C10444, Invitrogen) (1μM) were added in the presence of the corresponding treatments, to detect lipid peroxidation or total ROS levels, respectively. The cells were incubated for 30 minutes, washed and trypsinized. The fluorescence was measured at 4°C using a MACSQuant® VYB Flow Cytometer (Miltenyi Biotec, Germany). The data were normalized to the emission of control cells treated with DMSO.
2.5. Fe2+-binding capacity
Ferrozine is a specific reagent which forms a magenta-colored complex (absorption maximum at 562 nm) with ferrous (Fe2+) ions [16]. Briefly, 5 μl of a ferrous chloride (FeCl2) solution were mixed with 5 μl of compound in a 96-well plate at a 1:2 molar ratio and the mixture was incubated for 2 minutes at room temperature. 100 μl of 50 mM HEPES (pH 6.8) and 50 μl of 5mM ferrozine were then added to the mixture (50 μM final compound concentration). Absorbance (A562) was measured immediately after addition of ferrozine using a plate spectrophotometer. Concentration of free Fe2+ ions corresponds linearly to the A562. For normalization, the A562 yielded by an equimolar solution of the standard iron-chelator deferiprone or by a control solution without compound were considered as maximum and minimum iron binding capacity, respectively.
2.6. Autoxidation of egg-phosphatidylcholine (PC) liposomes
STY-BODIPY (1.5 μM) (cat No. 27089, Cayman Chemical Co, MI, United States) and liposomes of egg-PC (1 mM) (cat No. 840051P, Avanti Polar Lipids Inc, AL, United States) in TBS (pH 7.4) were added to an opaque 96-well polypropylene plate, followed by the addition of the indicated compounds (10 μM). The autoxidation was initiated by the addition of V-70 (0.5 mM) (cat No. 001-70078, Fujifilm Wako, Japan). For the enzymatic experiments, recombinant NQO1 (cat No. MBS201141, MyBioSource, CA, United States) (50 nM), recombinant FSP1 (cat No. 29611-50, Cayman Chemical) (50 nM), NADH (75 μM), the indicated quinones (5 μM), or a combination thereof, were added to the wells containing STY-BODIPY and liposomes prior to the addition of V-70. A lower compound concentration was used here to avoid rapid consumption of NADH by the most reactive quinones. Data were acquired by excitation of STY-BODIPY at 488 nm and emission was measured at 518 nm every 15 min at 37°C. Data were transformed into [ox-STY-BODIPY] by taking the raw fluorescence values of the saturated curve of control DMSO and dividing them by the initial concentration of reduced STY-BODIPY (1.5 μM). To obtain a unique number related to the data, the area under the curve (AUC) was calculated using Graph Pad Prism 6.
2.7. Enzymatic assays
In a 96 well plate, recombinant NQO1 or FSP1 (50 nM) were mixed with 75 μM NADH in TBS buffer, pH 7.4. The reactions were initiated by adding the different quinones at 5 μM. The plate was mixed and the change in absorbance at 340 nm was measured every 15 min at 37°C. Results are expressed as concentration of NADH over time.
2.8. Statistical analysis
The half maximal effective concentration (EC50) was determined from sigmoidal dose response curves using GraphPad Prism 6. All experiments were done at least in triplicate and repeated at least three times. Multiple groups were compared using one-way ANOVA followed by Dunnett’s correction. GraphPad Prism 6 was used for the statistical analyses. Data are expressed as mean ± SEM, and significance of difference is indicated as *P<0.05, **P<0.01 and ***P<0.001.
3. Results
3.1. Primary screening identifies novel potent anti-oxytotic/ferroptotic compounds
In order to identify novel anti-oxytotic/ferroptotic compounds, we screened a commercial library (HY-L021, MedChemExpress) of 903 chemically diverse compounds from a variety of biological sources including animals, microorganisms and plants (Table S1). The rationale for using this diversity was to not only increase the chances of identifying potent compounds but also to provide insight into the chemical nature of protection against oxytosis/ferroptosis.
All compounds were screened for their ability to protect HT22 cells from oxytosis/ferroptosis induced by RSL3 and glutamate, and a number of them were found to be protective at the concentrations tested (1 μM and 10 μM; Table S1). 58 compounds with ≥ 65% protection (at either 1 μM or 10 μM) against at least one of the insults were identified and re-tested at a wider range of concentrations (0.1 μM, 0.5 μM, 1 μM and 5 μM) (Table S2). This cutoff represents about two thirds of cell survival, which is a reliable level of protection that allows a good selection of compounds with good probability of displaying potent anti-oxytotic/ferroptotic activity.
In order to better estimate the anti-oxytotic/ferroptotic potency and proceed to a more detailed analysis, the range of concentrations tested was widened and the number of anti-oxytotic/ferroptotic compounds narrowed down to a manageable set of 20 compounds that protected ≥ 75% against both glutamate and RSL3 (Table 1 and Fig. 1A). The half maximal effective concentrations (EC50) of these 20 compounds against RSL3 and glutamate showed a strong correlation between the protection against both insults (Table 1 and Fig. 1B).
Table 1. EC50s (nM) of the top 20 anti-oxytotic/ferroptotic compounds for protection against insults relevant to AD.
RSL3 and glutamate were used to induce oxytosis/ferroptosis in HT22 cells. IAA was used to induce in vitro ischemia in HT22 cells. Aβ toxicity was induced in MC65 cells by removal of tetracycline.
| Compound | EC50 vs RSL3 (nM) | EC50 vs glutamate (nM) | EC50 vs IAA (nM) | EC50 vs Aβ (nM) |
|---|---|---|---|---|
| β-lapachone | 3 ± 1 | 15 ± 8 | 5 ± 1 | 5 ± 1 |
| DHIT I | 9 ± 5 | 59 ± 6 | 62 ± 18 | 10 ± 5 |
| DHT I | 38 ± 3 | 113 ± 20 | 26 ± 15 | 12 ± 7 |
| Triptophenolide | 52 ± 18 | 2330 ± 589 | 562 ± 105 | 40 ± 19 |
| Tanshinone I | 122 ± 35 | 361 ± 32 | 291 ± 70 | 100 ± 59 |
| Idebenone | 176 ± 33 | 182 ± 65 | 464 ± 187 | 651 ± 334 |
| Cryptotanshinone | 221 ± 14 | 738 ± 39 | 586 ± 108 | 19 ± 20 |
| Lipoic acid | 270 ± 80 | 1501 ± 131 | 2727 ± 465 | 181 ± 103 |
| Icaritin | 485 ± 87 | 663 ± 130 | 1330 ± 95 | 850 ± 651 |
| Gossypol | 579 ± 199 | 1596 ± 83 | 1729 ± 547 | 30 ± 37 |
| L-Ascorbic acid | 614 ± 82 | 742 ± 72 | 1333 ± 111 | - |
| Baicalein | 660 ± 158 | 1240 ± 76 | 1844 ± 182 | 253 ± 34 |
| Catharanthine | 906 ± 164 | 2418 ± 299 | 849 ± 19 | 2398 ± 1305 |
| Honokiol | 911 ± 211 | 3650 ± 148 | 2803 ± 19 | 279 ± 30 |
| Carnosol | 933 ± 104 | 1423 ± 88 | 645 ± 242 | 609 ± 42 |
| 3,3’ -Diindolylmethane | 1086 ± 187 | 2714 ± 520 | 1798 ± 316 | 402 ± 39 |
| Menaquinone-4 | 1317 ± 41 | 1173 ± 56 | 2747 ± 946 | 80 ± 65 |
| Dopamine | 1494 ± 360 | 1686 ± 217 | 4247 ± 60 | 3010 ± 944 |
| Cynaroside | 1152 ± 179 | 2439 ± 101 | 3856 ± 359 | 1203 ± 729 |
| Fisetin | 3844 ± 242 | 3771 ± 179 | 3430 ± 23 | 2501 ± 1173 |
Figure 1. Top 20 compounds and other quinones in the library.

(A) Chemical structures of the top 20 compounds. (B) Correlation between the protection against RSL3 (200 nM) and glutamate (10 mM) for the 20 most protective compounds (logEC50s, nM). (C) Chemical structures of other quinones in the library.
3.2. Source and chemical profiling of the most protective compounds show an enrichment in plant-derived secondary metabolites
To investigate the relationship between the biological source of a compound and its anti-oxytotic/ferroptotic activity, we compared the sources of the top 20 protective compounds with the sources of the compounds from the entire library (Fig. 2A). The percentage of plant-derived compounds greatly increased from ~56% in the whole library to ~85% in the top 20 protective compounds. This observation suggests that plants may be a better source of anti-oxytotic/ferroptotic compounds than other sources.
Figure 2. Source and chemical classification of the anti-oxytotic/ferroptotic compounds.

(A) Pie charts showing the percentage of compounds obtained from the indicated sources. (B) Pie charts showing the percentage of plant-derived compounds belonging to the indicated chemical classes.
We then focused on the different classes of plant compounds. With the exception of quinones, the other chemical classes, including flavonoids, alkaloids, terpenoids and lignans, were represented among the most protective plant-derived compounds at similar percentages to those in the whole library (Fig. 2B). Quinone-related compounds were highly enriched in the top 20, going from ~3% of the whole library to ~35% of the top 20 protective compounds. Although four of these quinones (tanshinone I, cryptotanshinone, dihydrotanshinone I (DHT I) and dihydroisotanshinone I (DHIT I)) are derived from the same plant (Salvia miltiorrhiza) and are structurally similar (Fig. 1A), three other quinones were identified among the best 20 compounds (β-lapachone, idebenone and menaquinone-4 (vitamin K2)). This observation suggests that the class of quinones may present increased potential for compounds with anti-oxytotic/ferroptotic activity. In fact, all the quinones in the library, including those not present in the top 20 (with the exception of rhein and chrysophanol), displayed some degree of protection against oxytosis/ferroptosis (Table S3 and Fig. 1C).
Physicochemical parameters are useful for predicting the potential of a molecule to be delivered to different tissues in an organism, thus providing a preliminary estimate of how suitable a compound might be as a drug candidate. Due to the constraints of the blood-brain barrier, this type of analysis is especially relevant for drugs targeting the central nervous system (CNS) [17, 18]. Notably, most of the top 20 compounds of the library have excellent estimated physicochemical properties that are within the range of successful CNS drugs (Table 2).
Table 2. Physicochemical properties of the top 20 protective compounds.
Mw, molecular weight; ClogP, general lipophilicity; tPSA, topological polar surface area; HBD, hydrogen bond donor; HBA, hydrogen bond receptor; ClogD, lipophilicity of ionizable compounds; pKa, acid dissociation constant; MPO, multiparameter optimization score; CNS, central nervous system.
| Compound | Mw | ClogP | tPSA (Å2) | HBD (n.OH,NH) | HBA (n.O,N) | ClogD (pH 7.4) | pKa (most basic center) | Pfizer CNS MPO score |
|---|---|---|---|---|---|---|---|---|
| Successful CNS drugs | ≤ 360 | ≤ 5 | ≤ 90 | ≤ 3 | ≤ 7 | ≤ 2 | ≤ 8 | 0 to 6 |
| β-lapachone | 242 | 1.71 | 43 | 0 | 3 | 2.77 | 0.0 | 5.6 |
| DHIT I | 278 | 4.40 | 43 | 0 | 3 | 4.22 | 0.0 | 4.3 |
| DHT I | 278 | 2.43 | 43 | 0 | 3 | 3.52 | 0.0 | 5.2 |
| Triptophenolide | 312 | 3.64 | 47 | 1 | 3 | 4.25 | 9.8 | 3.6 |
| Tanshinone I | 276 | 4.80 | 43 | 0 | 3 | 4.24 | 0.0 | 4.1 |
| Idebenone | 338 | 3.42 | 73 | 1 | 5 | 3.41 | 16.0 | 3.9 |
| Cryptotanshinone | 296 | 3.37 | 43 | 0 | 3 | 4.14 | 0.0 | 4.8 |
| Lipoic acid | 206 | 2.39 | 88 | 1 | 2 | −0.13 | 4.9 | 5.8 |
| Icaritin | 368 | 4.65 | 96 | 3 | 6 | 3.12 | 8.1 | 3.5 |
| Gossypol | 519 | 5.76 | 156 | 6 | 8 | 5.16 | 8.7 | 0.7 |
| L-Ascorbic acid | 176 | −1.76 | 107 | 4 | 6 | −4.99 | 4.2 | 4.4 |
| Baicalein | 270 | 3.00 | 87 | 3 | 5 | 1.60 | 6.7 | 5.2 |
| Catharanthine | 336 | 3.64 | 45 | 1 | 4 | 3.76 | 6.9 | 4.6 |
| Honokiol | 266 | 4.49 | 40 | 2 | 2 | 4.13 | 9.1 | 3.2 |
| Carnosol | 330 | 3.16 | 67 | 2 | 4 | 3.84 | 7.6 | 4.5 |
| 3,3’ -Diindolylmethane | 246 | 4.19 | 32 | 2 | 2 | 4.83 | 0.0 | 3.5 |
| Menaquinone-4 | 445 | 10.56 | 34 | 0 | 2 | 9.55 | 0.0 | 3.1 |
| Dopamine | 153 | 0.17 | 66 | 4 | 3 | −2.18 | 8.6 | 4.7 |
| Cynaroside | 448 | 0.81 | 186 | 7 | 11 | −1.21 | 8.0 | 3.4 |
| Fisetin | 286 | 1.24 | 107 | 4 | 6 | 1.38 | 7.6 | 4.4 |
3.3. Anti-oxytotic/ferroptotic activity correlates with protection against other insults relevant to AD
In order to further characterize the therapeutic potential of the top 20 protective compounds, these were tested in two additional cell culture models relevant to AD, one based on intracellular amyloid-beta (Aβ) toxicity and the other based on energy depletion.
A significant body of evidence indicates that Aβ accumulates within neurons of AD patients well before the appearance of the Aβ plaques themselves, and that this accumulation may play a central role in driving the disease [19–22]. To determine the effects of the top 20 anti-oxytotic/ferroptotic compounds on intracellular Aβ toxicity we used the MC65 nerve cell model. The MC65 is a human nerve cell line that expresses the C99 fragment of the amyloid precursor protein (APP) under the control of a tetracycline-sensitive promoter [23]. When tetracycline is withdrawn, cells express C99 which is then converted to Aβ by γ-secretase and the cells die within several days due to Aβ accumulation and aggregation within cells [24]. We found that the protection by the top 20 compounds against Aβ toxicity correlated with their protection against oxytosis/ferroptosis in HT22 cells (Table 1 and Fig. 3A).
Figure 3. Protection against oxytosis/ferroptosis correlates with protection against additional insults relevant to AD.

Correlation between protection against glutamate/RSL3 in HT22 cells and protection against Aβ toxicity in MC65 cells (A) or in vitro ischemia in HT22 cells (B) for the top 20 protective compounds.
Energy metabolism in the brain decreases with age and to larger extent in AD [25]. A breakdown in neuronal energy production leading to decreases in the levels of adenosine triphosphate (ATP) is associated with nerve cell damage and death in AD [26]. The loss of ATP can be mimicked using an in vitro ischemia model [27]. To induce ischemia, HT22 cells were treated with iodoacetic acid (IAA), a well-known irreversible inhibitor of the glycolytic enzyme glyceraldehyde 3-phosphate dehydrogenase (GAPDH) [28], and the effects of the top 20 anti-oxytotic/ferroptotic compounds in this model were evaluated. Notably, similar to the Aβ toxicity in MC65 cells, all of the compounds prevented IAA-induced cell death in a manner that highly correlated with their protection against oxytosis/ferroptosis (Table 1 and Fig. 3B). Taken together, these data are consistent with recent studies showing that the oxytosis/ferroptosis cell death pathway may underlie, at least in part, both the Aβ- and IAA-induced toxicities [13, 14, 29].
3.4. Inhibition of lipid peroxidation, but not modulation of GSH levels or total ROS production, is a common feature of the best anti-oxytotic/ferroptotic quinones
Since the group of the most potent anti-oxytotic/ferroptotic compounds was enriched in quinones, we hypothesized that these could share a protective mechanism of action. To begin investigating this hypothesis, we analyzed the effects of the top 20 compounds on the main steps of oxytosis/ferroptosis including the levels of GSH, ROS and lipid peroxidation.
To address changes in GSH metabolism, the levels of GSH were measured upon treatment with each compound in the presence or absence of glutamate (Fig. 4A). The concentration used for each compound in these assays was chosen based on the minimal concentration yielding the maximal protection against oxytosis/ferroptosis. Since we observed a strong correlation between the protection against glutamate and RSL3, the subsequent experiments were carried out only with glutamate. It was found that, although a few compounds maintained GSH levels in HT22 cells treated with glutamate (carnosol, cynaroside and fisetin), and some increased basal GSH levels in the absence of glutamate (DHIT I, DHT I, lipoic acid, carnosol, cynaroside and fisetin), most had little to no effect.
Figure 4. Modulation of GSH, total ROS and lipid peroxidation in HT22 cells exposed to oxytosis/ferroptosis.

Total GSH (A), total ROS (B) and lipid peroxidation (C) levels in HT22 cells treated with the top 20 protective compounds in the presence or absence of glutamate (10 mM). Graphs show the percentage of total GSH, fluorescent oxidized C11-BODIPY 581/591 (lipid peroxidation), fluorescent oxidized CellROX (total ROS) normalized to control DMSO treatment. The concentration of each compound tested in these assays was chosen based on the lowest dose yielding maximal protection against oxytosis/ferroptosis in HT22 cells. §§§p < 0.001 relative to control DMSO. *p < 0.05; **p < 0.01; ***p < 0.001 relative to control DMSO. #p < 0.05; ##p < 0.01; ###p < 0.001 relative to glutamate DMSO.
Next, the ability of the 20 best anti-oxytotic/ferroptotic compounds to modulate ROS production was determined by assessing the levels of total ROS in HT22 cells treated with the compounds in the presence or absence of glutamate. CellRox was used to measure intracellular levels of total ROS by flow cytometry. We found that about half of the compounds did not prevent the increase in ROS induced by glutamate (Fig. 4B). In fact, most of the quinones tested actually increased the levels of ROS on their own in the absence of glutamate.
Given the lack of a clear protective mechanism common to the quinones associated with regulation of GSH or total ROS levels, we then measured the effects of the compounds on lipid peroxidation. To do that, C11 BODIPY 581/591 was used to determine the levels of lipid peroxidation by flow cytometry. As seen in Fig. 4C, all of the top 20 compounds showed a strong prevention of the increase in lipid peroxidation induced by glutamate. This effect was most noticeable with the quinones, which also significantly decreased the basal levels of lipid peroxidation in the absence of glutamate.
Taken together, these data indicate that the anti-oxytotic/ferroptotic activity of the top 20 compounds analyzed, especially the quinones, is unrelated to the modulation of either ROS or GSH levels, but rather is likely dependent on their strong anti-lipid peroxidation activity.
3.5. Cell-free redox profiling shows no correlation with protection against oxytosis/ferroptosis in cells
To further understand the mechanism(s) underlying the effects of the compounds on lipid peroxidation, we next assessed the ability of the top 20 anti-oxytotic/ferroptotic compounds to chelate iron and/or directly inhibit lipid peroxidation in cell-free systems in order to exclude effects on other cellular processes.
The term ferroptosis was first introduced to define the iron-dependent generation of lipid peroxides and consequent cell death, and it was shown that iron chelators were able to prevent this toxicity [14, 30]. As such, we began by testing the iron-chelation capacity of the top 20 protective compounds. To do this, the compounds were incubated with ferrous chloride (FeCl2) and ferrozine solutions. The absorbance of this solution is inversely proportional to the iron (Fe2+)-binding capacity of the tested compound. We observed different degrees of Fe2+-binding capacity among the compounds but found no correlation with their respective EC50s against oxytosis/ferroptosis (Table 3). In fact, the majority of the top 20 compounds showed little to no ability to chelate Fe2+. Hence, the iron-binding capacity of these compounds appears to be unrelated to their ability to prevent oxytosis/ferroptosis in HT22 cells.
Table 3. Cell-free profiling of the top 20 anti-oxytotic/ferroptotic compounds.
LPO (AUC), lipid peroxidation (area under the curve).
| Compound | Fe2+-binding capacity (%) | LPO (AUC) |
|---|---|---|
| β-lapachone | 50.6 ± 7.4 | 0.97 ± 0.05 |
| DHIT I | 2.3 ± 1.1 | 0.81 ± 0.03 |
| DHT I | 6.7 ± 3.4 | 0.66 ± 0.09 |
| Triptophenolide | 6.7 ± 3.0 | 0.44 ± 0.03 |
| Tanshinone I | 3.7 ± 2.2 | 0.85 ± 0.04 |
| Idebenone | 7.6 ± 3.1 | 0.91 ± 0.05 |
| Cryptotanshinone | 71.3 ± 5.3 | 0.73 ± 0.03 |
| Lipoic acid | 7.5 ± 4.5 | 0.65 ± 0.02 |
| Icaritin | ~0 | 0.55 ± 0.06 |
| Gossypol | 8.2 ± 3.7 | 0.10 ± 0.02 |
| L-Ascorbic acid | 3.1 ± 1.5 | 0.35 ± 0.03 |
| Baicalein | 19.3 ± 5.2 | 0.83 ± 0.02 |
| Catharanthine | 40.1 ± 3.0 | 0.21 ± 0.04 |
| Honokiol | 8.7 ± 6.2 | 0.62 ± 0.02 |
| Carnosol | 10.5 ± 3.0 | 0.14 ± 0.04 |
| 3,3’ -Diindolylmethane | 8.2 ± 3.7 | 0.41 ± 0.03 |
| Menaquinone-4 | 1.1 ± 0.8 | 0.94 ± 0.15 |
| Dopamine | 10.2 ± 4.3 | 0.27 ± 0.03 |
| Cynaroside | 22.4 ± 6.9 | 0.31 ± 0.03 |
| Fisetin | 27.4 ± 6.1 | 0.17 ± 0.04 |
| Deferiprone | 82.7 ± 4.1 | - |
| Trolox | - | - |
Given that all of the top 20 protective compounds consistently prevented lipid peroxidation in cells but did not display Fe2+-binding capacity, we then asked whether their protection could be a consequence of direct inhibition of lipid peroxidation. To test this, we analyzed the ability of the compounds to inhibit lipid peroxidation in a cell-free assay based on the autoxidation of egg-phosphatidylcholine (PC) liposomes. Unexpectedly, although most of the top 20 protective compounds showed reduced lipid peroxidation in HT22 cells (Fig. 4), their effects on lipid peroxidation in the egg-PC liposomes did not correlate with the cell-based assay, with the best compounds, namely the quinones, presenting the weakest activity (Table 3 and Fig. 5).
Figure 5. Effects on lipid peroxidation in a cell-free liposome-based system.

Co-autoxidation of STY-BODIPY (1.5 μM) and the polyunsaturated lipids of egg-phosphatidylcholine liposomes (1 mM) in the presence of the indicated compounds (10 μM). Lipid autoxidation was initiated using V-70 (0.5 mM) and monitored by measuring the fluorescence increase over time at 37 °C. Graphs show one of n = 3 representative experiments.
The lack of correlation between the effects of the quinones on lipid peroxidation in cells and the cell-free assay prompted us to ask whether the cellular environment could be important for their activity. Quinones are unique chemical entities known to be dynamic redox species in their ability to shift between oxidized and reduced forms and are classified as bioreductive compounds as their activity depends on one- or two-electron reduction catalyzed by various quinone reductases that are present in cells [31–33]. The natural state for most of them is the oxidized form with conjugated cyclic systems [34]. Therefore, the low activity observed for the quinones in the egg-PC-autoxidation cell-free assay could be explained by the absence of the reduced form of the quinones given the lack of oxidoreductases in a cell-free system. To address this possibility, we next tested whether enzymatic reduction of the quinones could alter their anti-lipid peroxidation activity in the cell-free assay.
3.6. Enzymatic reduction of quinones is required for their anti-lipid peroxidation activity in cell-free systems
The ability of NAD(P)H:quinone oxidoreductase 1 (NQO1) to reduce endogenous and exogenous quinones in different models has been known for a long time [35, 36]. More recently, AIFM2, another oxidoreductase that has been renamed as ferroptosis-suppressor protein 1 (FSP1), has been shown to contribute to the anti-oxytotic/ferroptotic activity of coenzyme Q10 (ubiquinone) [37–39], an endogenous quinone.
To determine whether the most protective quinones can be enzymatically reduced in a cell-free environment, the quinones were incubated with recombinant NQO1 or FSP1 using NADH as the immediate electron donor. Since NADH absorbs at 340 nm, the decay of its absorbance was used to determine the enzymatic reduction of the quinones. With the exceptions of tanshinone I and menaquinone 4, all quinones were able to decrease NADH levels over time when incubated with either NQO1 or FSP1 (Fig. 6A).
Figure 6. Enzymatic reduction of quinones increases their anti-lipid peroxidation activity.

(A) The indicated compounds (5 μM) were mixed with NADH (75 μM) alone (CTRL) or with NQO1 or FSP1 (50 nM) and NADH in TBS pH 7.4. NADH levels were monitored by absorbance at 340 nm. (B) Co-autoxidation of STY-BODIPY (1.5 μM) and the polyunsaturated lipids of egg-phosphatidylcholine liposomes (1 mM) in the presence of the indicated compounds (5 μM) and NADH (75 μM) alone (CTRL) or with NQO1 or FSP1 (50 nM) and NADH. Lipid autoxidation was initiated using V-70 (0.5 mM) and monitored by measuring the fluorescence increase over time at 37°C. Graphs show one of n = 3 representative experiments.
To test whether this reduction of the quinones enhances their anti-lipid peroxidation activity in a cell-free system, the quinones were retested in the egg-PC-liposome-based assay in the presence of NQO1 or FSP1 and NADH. All quinones that were substrates of these proteins as identified by the NADH decay assay now displayed an increase in anti-lipid peroxidation activity in the liposomes in the presence of NQO1 or FSP1 (Fig. 6B). However, consistent with the NADH decay assay, the anti-lipid peroxidation activity of tanshinone I and menaquinone 4 was not affected by the presence of the enzymes (Fig. 6B). Since these two quinones both strongly prevent lipid peroxidation in cells, it is possible that they are reduced by other oxidoreductase enzymes [40–42], which may be required for their anti-lipid peroxidation activity.
Altogether, these data support the idea that quinones are a class of chemical compounds with the potential to strongly inhibit oxytosis/ferroptosis by directly targeting lipid peroxidation, but that they require endogenous cellular redox systems for their activity.
Discussion
Our study highlights the importance of plant-derived compounds for the identification and development of novel, potent inhibitors of oxytosis/ferroptosis. Plants display a tremendous array of biochemicals that have evolved to deal with diverse physiological stresses, some of which are relevant to human medicine [17]. Plant primary metabolites (PPMs) are involved in metabolic processes that are required for basic life functions, including cell division, development and reproduction. On the other hand, plant secondary metabolites (PSMs) are not involved in basic functions but rather are produced to deal with abiotic stresses (e.g., UV radiation, drought, heat and soil toxicities) as well as to mediate interactions with antagonists (e.g., herbivores, pathogens and neighboring plants) and mutualists (e.g., fungi, bacteria and pollinators) [43]. Importantly, as in oxytosis/ferroptosis, high levels of lipid peroxides are generated as by-products of plant metabolism, which can occur both enzymatically via LOXs and non-enzymatically by the action of ROS and transition metal ions [17]. In fact, oxytosis/ferroptosis-like cell death has been shown to occur in plants upon stress [44]. These observations are consistent with the high prevalence of PSMs among the best anti-oxytotic/ferroptotic compounds identified in the present study.
To our knowledge, only 9 of the best 20 compounds identified (Cryptotanshinone, idebenone, menaquinone 4, dopamine, lipoic acid, ascorbic acid, baicalein, 3,3’-diindolylmethane and fisetin) have been previously described as anti-oxytotic/ferroptotic molecules [45–56]. Triptophenolide and cynaroside (luteoloside) have been shown to exert anti-oxidant activity [57, 58]. Icaritin, gossypol, carnosol and honokiol have been described as both pro- and anti-oxidant in different studies [59–67]. β-lapachone, DHIT I, DHT I and tanshinone I have been shown to induce ROS production leading to apoptosis and ferroptosis, particularly in cancer cells [68–71].
Four of the best top 20 compounds (DHIT I, DHT I, tanshinone I and cryptotanshinone) are diterpenoid quinones found in the Chinese medicinal herb Danshen (Salvia miltiorrhiza) [72]. Six other compounds in the library (baicalin, danshensu, rosmarinic acid, salvianolic acid, stigmasterol and ursolic acid) are also found in Danshen [72], but offered little to no protection against oxytosis/ferroptosis in HT22 cells. Danshen, a perennial herb native to China, has been traditionally used for centuries to treat cardiovascular diseases [73], but its bioactive constituents possess a variety of pharmacological activities including anti-tumor, anti-oxidant, anti-microbial, anti-viral and anti-inflammatory activities [72]. Importantly, Danshen and several of its constituents have been tested in cellular and animal models of AD and found to be neuroprotective [74, 75]. It will be interesting to investigate in future studies whether there is potential in Danshen to treat AD based on the anti-oxytotic/ferroptotic activity of some of its secondary metabolites, as identified in our screening.
We found that, from all the plant-derived compounds, quinones were enriched and had the highest activity against oxytosis/ferroptosis in the library. Quinones can be found in natural products other than plants, including fungi and bacteria, and have been shown to play a key role in the redox homeostasis of mitochondria and cell membranes [42, 76, 77]. Although recent studies have reported the involvement of ubiquinone in the regulation of oxytosis/ferroptosis [37, 38], the role of other quinones in this process remains poorly understood. Quinones are usually reduced by cellular reductases in either one- or two-electron reductions that require NADH or NADPH [42]. One-electron reduction yields a semiquinone radical (Q•−), which reacts rapidly with O2 to form superoxide anion and eventually H2O2. Therefore, in the redox environment of biological systems, one-electron reduction of quinones can lead to ROS production [42, 78].
Two-electron reduction yields the corresponding dihydroquinone (QH2). Several two-electron quinone reductases, including NQO1 and FSP1, have been identified [76, 79, 80]. It is thought that the enzymatic action of NQO1 overrides semiquinone formation by one-electron quinone reductases [79], conferring a quinone-detoxifying role to this enzyme [81]. However, there are also reports showing that NQO1 is necessary to activate quinone toxicity. This property has been investigated as a potential anti-cancer approach using quinones, including β-lapachone and tanshinone, given that NQO1 is frequently overexpressed in a variety of tumors [36, 82–85]. Based on these observations and studies suggesting that the antioxidant role of the QH2 form of the endogenous ubiquinone is specifically involved in preventing lipid peroxidation versus other types of ROS, it is possible that the QH2 forms generated by NQO1 and FPS1 are responsible for the anti-oxytotic/ferroptotic activity of the quinones analyzed here via prevention of lipid peroxidation while still leading to an increase in ROS [41, 86–88]. It is likely that, in addition to NQO1 and FPS1, other oxidoreductases participate in the reducing process of the quinones in HT22 cells. This is suggested by the observations with tanshinone I and menaquinone. Future studies should aim at the identification of these enzymes, which evidently will be of value for further understanding the oxytosis/ferroptosis pathway.
In addition, due to the natural tendency of QH2 for auto-oxidation and the redox cycling that is established between quinone, semiquinone and QH2, the use of intracellular NADH and NADPH by redox enzymes in this cycle can further exacerbate the production of ROS [89]. The variable instability of the reduced form of some of the quinones analyzed here could explain the lack of correlation between the respective rate of NADH consumption and the observed anti-lipid peroxidation activity in the cell-free systems in the presence of NQO1 or FSP1. Moreover, these observations could explain the increased levels of ROS under basal conditions that we observed upon treatment with the most protective quinones. The fact that these quinones act differently on ROS and lipid peroxidation shows that a compound can be pro-oxidant while still inhibiting lipid peroxidation, highlighting the importance of therapeutically targeting lipid peroxidation in this cell death mechanism. However, although there has been interest in testing quinones as therapeutics against neurodegenerative diseases with positive results, namely idebenone and β-lapachone [90–93], indicating promising therapeutic potential, their use in the clinic for this purpose must be viewed with caution given the possibility of generating ROS, which can damage other molecules (protein, DNA) [94, 95]. It should be added that confirmation of the top 20 compounds as potential AD drug candidates, the ones that have not been studied as such yet, will require proper testing in animal models of the disease.
Several studies suggest that pathological Aβ accumulation in AD can be initiated within neuronal cells [96–99]. Our lab has recently shown that intracellular Aβ accumulation induces oxytosis/ferroptosis in MC65 nerve cells while reducing GSH levels as well as increasing lipid peroxidation and mitochondrial ROS production [13, 14]. In fact, intracellular Aβ accumulation has been associated with the disruption of a variety of cellular functions that overlap with those most affected during oxytosis/ferroptosis such as autophagy or mitochondrial function [100–102]. Our results in MC65 cells further support the idea that the cell death caused by the toxic intracellular accumulation of Aβ can be prevented by treating the cells with compounds that target lipid peroxidation and prevent oxytosis/ferroptosis.
A growing body of evidence shows that features of oxytosis/ferroptosis are also found in other neurological disorders including Parkinson’s disease, Huntington’s disease and amyotrophic lateral sclerosis [3, 4, 103, 104]. For example, decreases in GSH along with iron accumulation and lipid peroxidation have been reported in the regions of the nervous system that are preferentially affected in all of these diseases [103–107]. Notably, iron plays an important role in oxytosis/ferroptosis, as iron chelators prevent oxytosis/ferroptosis-mediated cell death [3, 30]. It is thought that iron can not only generate ROS via the Fenton reaction but also promote the activation of the non-heme iron-containing LOX enzymes [108]. However, our data show that neither iron chelation nor GSH modulation are responsible for the protective effects of most of the anti-oxytotic/ferroptotic compounds identified in this study. In fact, the best compounds are much stronger inhibitors of oxytosis/ferroptosis than the other compounds tested that exhibited iron chelation activity. This observation indicates that phenotypic screening using cells exposed to RSL3 or glutamate is substantially more efficient at identifying potent inhibitors of oxytosis/ferroptosis than selecting compounds based on their capacity to chelate iron.
In addition to the MC65 assay, our lab has been using other cell-based assays for the identification of neuroprotective compounds which represent additional toxicities relevant to dementia [109]. This is the case of energy loss in the brain, which can be mimicked in cultured HT22 cells using IAA [110]. We and others have shown that GSH levels are decreased and lipid peroxidation is enhanced in this cellular model of in vitro ischemia [111, 112]. Our observations that the protection against oxytosis/ferroptosis by the best 20 compounds strongly correlated with the protection against IAA and intracellular Aβ toxicities not only support a role for oxytosis/ferroptosis in these two pathological processes relevant to aging and dementia, but also further reinforce using a phenotypic screening approach based on oxytosis/ferroptosis insults to identify new neuroprotective drugs that have a broad range of beneficial activities.
Conclusions
In summary, our study shows that plants represent an excellent source of natural anti-oxytotic/ferroptotic compounds, and that this potential may favor specific chemical classes that directly target lipid peroxidation in cells. Once identified, these compounds could either be tested as drug candidates, if their physicochemical and safety properties allow, or used as lead compounds for further improvement via medicinal chemistry. Given the growing central role that oxytosis/ferroptosis is being reported to play in a diversity of human diseases that extend beyond the CNS, the value of having new therapeutics that target the pathway cannot be overstated. In addition, our data highlight the necessity for using cell-based assays as opposed to solely relying on cell-free assays, which do not account for the redox systems present in cells that may be required for the activity of certain classes of compounds. Finally, the value of using oxytosis/ferroptosis as a screening strategy is further substantiated by the fact that anti-oxytotic/ferroptotic compounds can also act upon other pathological processes that are critical for aging and neurodegeneration, such as energy loss and Aβ toxicity.
Supplementary Material
Table S1. Anti-oxytotic/ferroptotic activity of all the compounds in the library.
Table S2. Anti-oxytotic/ferroptotic activity of the best 58 compounds in the library.
Highlights.
Plants display tremendous potential as source of anti-oxytotic/ferroptotic drugs
Plant-derived quinones have highest activity among all chemical classes
Pro-oxidant compounds can prevent lipid peroxidation and oxytosis/ferroptosis
Anti-lipid peroxidation activity of quinones depends on cellular oxidoreductases
Acknowledgments
This work was supported by the Shiley-Marcos Alzheimer’s Disease Research Center at University of California San Diego (A.C.), an Innovation Award from the Salk Institute (A.C.), the Shiley Foundation (D.S.C.), the Paul F. Glenn Center for Biology of Aging Research at the Salk Institute (Z.L.) and the National Institutes of Health grant R01AG069206 (P.M.).
Abbreviations:
- Aβ
β-amyloid
- ABTS
2,2’-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid)
- AD
Alzheimer’s disease
- AIFM2
apoptosis inducing factor mitochondria associated 2
- ATP
adenosine triphosphate
- CNS
central nervous system
- Cys
cysteine
- DHT I
dihydrotanshinone I
- DHIT I
dihydroisotanshinone I
- EC50
half maximal effective concentration
- FSP1
ferroptosis suppressor protein 1
- GAPDH
glyceraldehyde 3-phosphate dehydrogenase
- Glu
glutamate
- GPX4
glutathione peroxidase 4
- GSH
glutathione
- HD
Huntington’s disease
- IAA
iodoacetic acid
- LOX
lipoxygenase
- NAD
nicotinamide adenine dinucleotide
- NADH
nicotinamide adenine dinucleotide hydrogen
- NQO1
NAD(P)H:quinone oxidoreductase 1
- PC
phosphatidylcholine
- PD
Parkinson’s disease
- PK
pharmacokinetics
- PPM
plant primary metabolites
- PSM
plant secondary metabolites
- QH2
dihydroquinone
- Q•−
semiquinone radical
- ROS
reactive oxygen species
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflicts of interest
The authors declare no conflict of interest.
References
- 1.Masters CL, Bateman R, Blennow K, Rowe CC, Sperling RA, and Cummings JL (2015). Alzheimer’s disease. Nat Rev Dis Primers 1, 15056. [DOI] [PubMed] [Google Scholar]
- 2.Tan S, Schubert D, and Maher P (2001). Oxytosis: A novel form of programmed cell death. Curr Top Med Chem 1, 497–506. [DOI] [PubMed] [Google Scholar]
- 3.Maher P, Currais A, and Schubert D (2020). Using the Oxytosis/Ferroptosis Pathway to Understand and Treat Age-Associated Neurodegenerative Diseases. Cell Chem Biol 27, 1456–1471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Lewerenz J, Ates G, Methner A, Conrad M, and Maher P (2018). Oxytosis/Ferroptosis-(Re-) Emerging Roles for Oxidative Stress-Dependent Non-apoptotic Cell Death in Diseases of the Central Nervous System. Front Neurosci 12, 214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Tan S, Sagara Y, Liu Y, Maher P, and Schubert D (1998). The regulation of reactive oxygen species production during programmed cell death. J Cell Biol 141, 1423–1432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Ates G, Goldberg J, Currais A, and Maher P (2020). CMS121, a fatty acid synthase inhibitor, protects against excess lipid peroxidation and inflammation and alleviates cognitive loss in a transgenic mouse model of Alzheimer’s disease. Redox Biol 36, 101648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Currais A, Farrokhi C, Dargusch R, Armando A, Quehenberger O, Schubert D, and Maher P (2018). Fisetin Reduces the Impact of Aging on Behavior and Physiology in the Rapidly Aging SAMP8 Mouse. J Gerontol A Biol Sci Med Sci 73, 299–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Currais A, Goldberg J, Farrokhi C, Chang M, Prior M, Dargusch R, Daugherty D, Armando A, Quehenberger O, Maher P, et al. (2015). A comprehensive multiomics approach toward understanding the relationship between aging and dementia. Aging (Albany NY) 7, 937–955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Goldberg J, Currais A, Prior M, Fischer W, Chiruta C, Ratliff E, Daugherty D, Dargusch R, Finley K, Esparza-Molto PB, et al. (2018). The mitochondrial ATP synthase is a shared drug target for aging and dementia. Aging Cell 17(2):e12715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Li Y, Maher P, and Schubert D (1997). A role for 12-lipoxygenase in nerve cell death caused by glutathione depletion. Neuron 19, 453–463. [DOI] [PubMed] [Google Scholar]
- 11.Schubert D, and Maher P (2012). An alternative approach to drug discovery for Alzheimer’s disease dementia. Future Med Chem 4, 1681–1688. [DOI] [PubMed] [Google Scholar]
- 12.Davis JB, and Maher P (1994). Protein kinase C activation inhibits glutamate-induced cytotoxicity in a neuronal cell line. Brain Res 652, 169–173. [DOI] [PubMed] [Google Scholar]
- 13.Huang L, McClatchy DB, Maher P, Liang Z, Diedrich JK, Soriano-Castell D, Goldberg J, Shokhirev M, Yates JR 3rd, Schubert D, et al. (2020). Intracellular amyloid toxicity induces oxytosis/ferroptosis regulated cell death. Cell Death Dis 11, 828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Soriano-Castell D, Currais A, and Maher P (2021). Defining a pharmacological inhibitor fingerprint for oxytosis/ferroptosis. Free Radic Biol Med 171, 219–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Maher P (2018). Potentiation of glutathione loss and nerve cell death by the transition metals iron and copper: Implications for age-related neurodegenerative diseases. Free Radic Biol Med 115, 92–104. [DOI] [PubMed] [Google Scholar]
- 16.Mladenka P, Macakova K, Filipsky T, Zatloukalova L, Jahodar L, Bovicelli P, Silvestri IP, Hrdina R, and Saso L (2011). In vitro analysis of iron chelating activity of flavonoids. J Inorg Biochem 105, 693–701. [DOI] [PubMed] [Google Scholar]
- 17.Soriano-Castell D, Liang Z, Maher P, and Currais A (2021). The search for anti-oxytotic/ferroptotic compounds in the plant world. Br J Pharmacol 178, 3611–3626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Srinivas N, Maffuid K, and Kashuba ADM (2018). Clinical Pharmacokinetics and Pharmacodynamics of Drugs in the Central Nervous System. Clin Pharmacokinet 57, 1059–1074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Gouras GK, Tampellini D, Takahashi RH, and Capetillo-Zarate E (2010). Intraneuronal beta-amyloid accumulation and synapse pathology in Alzheimer’s disease. Acta Neuropathol 119, 523–541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.LaFerla FM, Green KN, and Oddo S (2007). Intracellular amyloid-beta in Alzheimer’s disease. Nat Rev Neurosci 8, 499–509. [DOI] [PubMed] [Google Scholar]
- 21.Takahashi RH, Nagao T, and Gouras GK (2017). Plaque formation and the intraneuronal accumulation of beta-amyloid in Alzheimer’s disease. Pathol Int 67, 185–193. [DOI] [PubMed] [Google Scholar]
- 22.Wirths O, Multhaup G, and Bayer TA (2004). A modified beta-amyloid hypothesis: intraneuronal accumulation of the beta-amyloid peptide--the first step of a fatal cascade. J Neurochem 91, 513–520. [DOI] [PubMed] [Google Scholar]
- 23.Sopher BL, Fukuchi K, Smith AC, Leppig KA, Furlong CE, and Martin GM (1994). Cytotoxicity mediated by conditional expression of a carboxyl-terminal derivative of the beta-amyloid precursor protein. Brain Res Mol Brain Res 26, 207–217. [DOI] [PubMed] [Google Scholar]
- 24.Currais A, Farrokhi C, Dargusch R, Goujon-Svrzic M, and Maher P (2016). Dietary glycemic index modulates the behavioral and biochemical abnormalities associated with autism spectrum disorder. Mol Psychiatry 21, 426–436. [DOI] [PubMed] [Google Scholar]
- 25.Currais A (2015). Ageing and inflammation - A central role for mitochondria in brain health and disease. Ageing Res Rev 21, 30–42. [DOI] [PubMed] [Google Scholar]
- 26.Saxena U (2012). Bioenergetics failure in neurodegenerative diseases: back to the future. Expert Opin Ther Targets 16, 351–354. [DOI] [PubMed] [Google Scholar]
- 27.Maher P, Salgado KF, Zivin JA, and Lapchak PA (2007). A novel approach to screening for new neuroprotective compounds for the treatment of stroke. Brain Res 1173, 117–125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Sabri MI, and Ochs S (1971). Inhibition of glyceraldehyde-3-phosphate dehydrogenase in mammalian nerve by iodoacetic acid. J Neurochem 18, 1509–1514. [DOI] [PubMed] [Google Scholar]
- 29.Gunesch S, Soriano-Castell D, Lamer S, Schlosser A, Maher P, and Decker M (2020). Development and Application of a Chemical Probe Based on a Neuroprotective Flavonoid Hybrid for Target Identification Using Activity-Based Protein Profiling. ACS Chem Neurosci 11, 3823–3837. [DOI] [PubMed] [Google Scholar]
- 30.Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al. (2012). Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149, 1060–1072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Molina Portela MP, and Stoppani AO (1996). Redox cycling of beta-lapachone and related o-naphthoquinones in the presence of dihydrolipoamide and oxygen. Biochem Pharmacol 51, 275283. [DOI] [PubMed] [Google Scholar]
- 32.Martinez-Cifuentes M, Salazar R, Ramirez-Rodriguez O, Weiss-Lopez B, and Araya-Maturana R (2017). Experimental and Theoretical Reduction Potentials of Some Biologically Active ortho-Carbonyl para-Quinones. Molecules 22(4), 577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Iyanagi T, and Yamazaki I (1970). One-electron-transfer reactions in biochemical systems. V. Difference in the mechanism of quinone reduction by the NADH dehydrogenase and the NAD(P)H dehydrogenase (DT-diaphorase). Biochim Biophys Acta 216, 282–294. [DOI] [PubMed] [Google Scholar]
- 34.Benito PB, M. MJA a. (2005). Biological Activity of Quinones. Studies in natural products chemistry 30, 303–366. [Google Scholar]
- 35.Brunmark A, Cadenas E, Lind C, Segura-Aguilar J, and Ernster L (1987). DT-diaphorase-catalyzed two-electron reduction of quinone epoxides. Free Radic Biol Med 3, 181–188. [DOI] [PubMed] [Google Scholar]
- 36.Cresteil T, and Jaiswal AK (1991). High levels of expression of the NAD(P)H:quinone oxidoreductase (NQO1) gene in tumor cells compared to normal cells of the same origin. Biochem Pharmacol 42, 1021–1027. [DOI] [PubMed] [Google Scholar]
- 37.Bersuker K, Hendricks JM, Li Z, Magtanong L, Ford B, Tang PH, Roberts MA, Tong B, Maimone TJ, Zoncu R, et al. (2019). The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature 575, 688–692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Doll S, Freitas FP, Shah R, Aldrovandi M, da Silva MC, Ingold I, Goya Grocin A, Xavier da Silva TN, Panzilius E, Scheel CH, et al. (2019). FSP1 is a glutathione-independent ferroptosis suppressor. Nature 575, 693–698. [DOI] [PubMed] [Google Scholar]
- 39.Hadian K (2020). Ferroptosis Suppressor Protein 1 (FSP1) and Coenzyme Q10 Cooperatively Suppress Ferroptosis. Biochemistry 59, 637–638. [DOI] [PubMed] [Google Scholar]
- 40.O’Brien PJ (1991). Molecular mechanisms of quinone cytotoxicity. Chem Biol Interact 80, 1–41. [DOI] [PubMed] [Google Scholar]
- 41.Cadenas E, Hochstein P, and Ernster L (1992). Pro- and antioxidant functions of quinones and quinone reductases in mammalian cells. Adv Enzymol Relat Areas Mol Biol 65, 97–146. [DOI] [PubMed] [Google Scholar]
- 42.Monks TJ, and Jones DC (2002). The metabolism and toxicity of quinones, quinonimines, quinone methides, and quinone-thioethers. Curr Drug Metab 3, 425–438. [DOI] [PubMed] [Google Scholar]
- 43.Kessler A (2015). The information landscape of plant constitutive and induced secondary metabolite production. Curr Opin Insect Sci 8, 47–53. [DOI] [PubMed] [Google Scholar]
- 44.Distefano AM, Martin MV, Cordoba JP, Bellido AM, D’Ippolito S, Colman SL, Soto D, Roldan JA, Bartoli CG, Zabaleta EJ, et al. (2017). Heat stress induces ferroptosis-like cell death in plants. J Cell Biol 216, 463–476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Avci B, Gunaydin C, Guvenc T, Yavuz CK, Kuruca N, and Bilge SS (2021). Idebenone Ameliorates Rotenone-Induced Parkinson’s Disease in Rats Through Decreasing Lipid Peroxidation. Neurochem Res 46, 513–522. [DOI] [PubMed] [Google Scholar]
- 46.Gao H, Bai Y, Jia Y, Zhao Y, Kang R, Tang D, and Dai E (2018). Ferroptosis is a lysosomal cell death process. Biochem Biophys Res Commun 503, 1550–1556. [DOI] [PubMed] [Google Scholar]
- 47.Ito K, Eguchi Y, Imagawa Y, Akai S, Mochizuki H, and Tsujimoto Y (2017). MPP+ induces necrostatin-1- and ferrostatin-1-sensitive necrotic death of neuronal SH-SY5Y cells. Cell Death Discov 3, 17013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Li J, Lin JC, Wang H, Peterson JW, Furie BC, Furie B, Booth SL, Volpe JJ, and Rosenberg PA (2003). Novel role of vitamin k in preventing oxidative injury to developing oligodendrocytes and neurons. J Neurosci 23, 5816–5826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Li J, Wang H, and Rosenberg PA (2009). Vitamin K prevents oxidative cell death by inhibiting activation of 12-lipoxygenase in developing oligodendrocytes. J Neurosci Res 87, 1997–2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Li Q, Li QQ, Jia JN, Sun QY, Zhou HH, Jin WL, and Mao XY (2019). Baicalein Exerts Neuroprotective Effects in FeCl3-Induced Posttraumatic Epileptic Seizures via Suppressing Ferroptosis. Front Pharmacol 10, 638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Liu L, Yang S, and Wang H (2021). alpha-Lipoic acid alleviates ferroptosis in the MPP(+) -induced PC12 cells via activating the PI3K/Akt/Nrf2 pathway. Cell Biol Int 45, 422–431. [DOI] [PubMed] [Google Scholar]
- 52.Lorincz T, Holczer M, Kapuy O, and Szarka A (2019). The Interrelationship of Pharmacologic Ascorbate Induced Cell Death and Ferroptosis. Pathol Oncol Res 25, 669–679. [DOI] [PubMed] [Google Scholar]
- 53.Maher P (2020). Modulation of the Neuroprotective and Anti-inflammatory Activities of the Flavonol Fisetin by the Transition Metals Iron and Copper. Antioxidants (Basel) 9(11), 1113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Wang D, Peng Y, Xie Y, Zhou B, Sun X, Kang R, and Tang D (2016). Antiferroptotic activity of non-oxidative dopamine. Biochem Biophys Res Commun 480, 602–607. [DOI] [PubMed] [Google Scholar]
- 55.Xie Y, Song X, Sun X, Huang J, Zhong M, Lotze MT, Zeh HJR, Kang R, and Tang D (2016). Identification of baicalein as a ferroptosis inhibitor by natural product library screening. Biochem Biophys Res Commun 473, 775–780. [DOI] [PubMed] [Google Scholar]
- 56.Zhang YH, Wang DW, Xu SF, Zhang S, Fan YG, Yang YY, Guo SQ, Wang S, Guo T, Wang ZY, et al. (2018). alpha-Lipoic acid improves abnormal behavior by mitigation of oxidative stress, inflammation, ferroptosis, and tauopathy in P301S Tau transgenic mice. Redox Biol 14, 535–548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Cai Y, Luo Q, Sun M, and Corke H (2004). Antioxidant activity and phenolic compounds of 112 traditional Chinese medicinal plants associated with anticancer. Life Sci 74, 2157–2184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Sun X, Sun GB, Wang M, Xiao J, and Sun XB (2011). Protective effects of cynaroside against H(2)O(2)-induced apoptosis in H9c2 cardiomyoblasts. J Cell Biochem 112, 2019–2029. [DOI] [PubMed] [Google Scholar]
- 59.Xu Y, Lu X, Zhang L, Wang L, Zhang G, Yao J, and Sun C (2021). Icaritin activates Nrf2/Keap1 signaling to protect neuronal cells from oxidative stress. Chem Biol Drug Des 97, 111–120. [DOI] [PubMed] [Google Scholar]
- 60.Zhou C, Chen Z, Lu X, Wu H, Yang Q, and Xu D (2016). Icaritin activates JNK-dependent mPTP necrosis pathway in colorectal cancer cells. Tumour Biol 37, 3135–3144. [DOI] [PubMed] [Google Scholar]
- 61.Hanus J, Zhang H, Chen DH, Zhou Q, Jin P, Liu Q, and Wang S (2015). Gossypol Acetic Acid Prevents Oxidative Stress-Induced Retinal Pigment Epithelial Necrosis by Regulating the FoxO3/Sestrin2 Pathway. Mol Cell Biol 35, 1952–1963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.El-Sharaky AS, Wahby MM, Bader El-Dein MM, Fawzy RA, and El-Shahawy IN (2009). Mutual anti-oxidative effect of gossypol acetic acid and gossypol-iron complex on hepatic lipid peroxidation in male rats. Food Chem Toxicol 47, 2735–2741. [DOI] [PubMed] [Google Scholar]
- 63.Santana AT, Guelfi M, Medeiros HC, Tavares MA, Bizerra PF, and Mingatto FE (2015). Mechanisms involved in reproductive damage caused by gossypol in rats and protective effects of vitamin E. Biol Res 48, 43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Saleh SR, Attia R, and Ghareeb DA (2018). The Ameliorating Effect of Berberine-Rich Fraction against Gossypol-Induced Testicular Inflammation and Oxidative Stress. Oxid Med Cell Longev 2018, 1056173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Al Dhaheri Y, Attoub S, Ramadan G, Arafat K, Bajbouj K, Karuvantevida N, AbuQamar S, Eid A, and Iratni R (2014). Carnosol induces ROS-mediated beclin1-independent autophagy and apoptosis in triple negative breast cancer. PLoS One 9, e109630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Samarghandian S, Azimi-Nezhad M, Borji A, Samini M, and Farkhondeh T (2017). Protective effects of carnosol against oxidative stress induced brain damage by chronic stress in rats. BMC Complement Altern Med 17, 249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Lin C, Zhang X, Su Z, Xiao J, Lv M, Cao Y, and Chen Y (2019). Carnosol Improved Lifespan and Healthspan by Promoting Antioxidant Capacity in Caenorhabditis elegans. Oxid Med Cell Longev 2019, 5958043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Park EJ, Choi KS, and Kwon TK (2011). beta-Lapachone-induced reactive oxygen species (ROS) generation mediates autophagic cell death in glioma U87 MG cells. Chem Biol Interact 189, 37–44. [DOI] [PubMed] [Google Scholar]
- 69.Wu CY, Yang YH, Lin YS, Chang GH, Tsai MS, Hsu CM, Yeh RA, Shu LH, Cheng YC, and Liu HT (2021). Dihydroisotanshinone I induced ferroptosis and apoptosis of lung cancer cells. Biomed Pharmacother 139, 111585. [DOI] [PubMed] [Google Scholar]
- 70.Tan S, Hou X, and Mei L (2020). Dihydrotanshinone I inhibits human glioma cell proliferation via the activation of ferroptosis. Oncol Lett 20, 122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Nizamutdinova IT, Lee GW, Son KH, Jeon SJ, Kang SS, Kim YS, Lee JH, Seo HG, Chang KC, and Kim HJ (2008). Tanshinone I effectively induces apoptosis in estrogen receptor-positive (MCF-7) and estrogen receptor-negative (MDA-MB-231) breast cancer cells. Int J Oncol 33, 485–491. [PubMed] [Google Scholar]
- 72.Wang B-Q (2010). Salvia miltiorrhiza: Chemical and pharmacological review of a medicinal plant. Journal of Medicinal Plants Research 4, 2813–2820. [Google Scholar]
- 73.Li ZM, Xu SW, and Liu PQ (2018). Salvia miltiorrhizaBurge (Danshen): a golden herbal medicine in cardiovascular therapeutics. Acta Pharmacol Sin 39, 802–824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Chong CM, Su H, Lu JJ, and Wang Y (2019). The effects of bioactive components from the rhizome of Salvia miltiorrhiza (Danshen) on the characteristics of Alzheimer’s disease. Chin Med 14, 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Howes MR, Fang R, and Houghton PJ (2017). Effect of Chinese Herbal Medicine on Alzheimer’s Disease. Int Rev Neurobiol 135, 29–56. [DOI] [PubMed] [Google Scholar]
- 76.Thor H, Smith MT, Hartzell P, Bellomo G, Jewell SA, and Orrenius S (1982). The metabolism of menadione (2-methyl-1,4-naphthoquinone) by isolated hepatocytes. A study of the implications of oxidative stress in intact cells. J Biol Chem 257, 12419–12425. [PubMed] [Google Scholar]
- 77.Liang Z, Currais A, Soriano-Castell D, Schubert D, and Maher P (2021). Natural products targeting mitochondria: emerging therapeutics for age-associated neurological disorders. Pharmacol Ther 221, 107749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Bolton JL, Trush MA, Penning TM, Dryhurst G, and Monks TJ (2000). Role of quinones in toxicology. Chem Res Toxicol 13, 135–160. [DOI] [PubMed] [Google Scholar]
- 79.Lind C, Hochstein P, and Ernster L (1982). DT-diaphorase as a quinone reductase: a cellular control device against semiquinone and superoxide radical formation. Arch Biochem Biophys 216, 178–185. [DOI] [PubMed] [Google Scholar]
- 80.Elguindy MM, and Nakamaru-Ogiso E (2015). Apoptosis-inducing Factor (AIF) and Its Family Member Protein, AMID, Are Rotenone-sensitive NADH:Ubiquinone Oxidoreductases (NDH-2). J Biol Chem 290, 20815–20826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Cadenas E (1995). Antioxidant and prooxidant functions of DT-diaphorase in quinone metabolism. Biochem Pharmacol 49, 127–140. [DOI] [PubMed] [Google Scholar]
- 82.Glorieux C, Sandoval JM, Dejeans N, Ameye G, Poirel HA, Verrax J, and Calderon PB (2016). Overexpression of NAD(P)H:quinone oxidoreductase 1 (NQO1) and genomic gain of the NQO1 locus modulates breast cancer cell sensitivity to quinones. Life Sci 145, 57–65. [DOI] [PubMed] [Google Scholar]
- 83.Yang Y, Zhou X, Xu M, Piao J, Zhang Y, Lin Z, and Chen L (2017). beta-lapachone suppresses tumour progression by inhibiting epithelial-to-mesenchymal transition in NQO1-positive breast cancers. Sci Rep 7, 2681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Kim DW, and Cho JY (2018). NQO1 is Required for beta-Lapachone-Mediated Downregulation of Breast-Cancer Stem-Cell Activity. Int J Mol Sci 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Zhang Y, Jiang P, Ye M, Kim SH, Jiang C, and Lu J (2012). Tanshinones: sources, pharmacokinetics and anti-cancer activities. Int J Mol Sci 13, 13621–13666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Maroz A, Anderson RF, Smith RA, and Murphy MP (2009). Reactivity of ubiquinone and ubiquinol with superoxide and the hydroperoxyl radical: implications for in vivo antioxidant activity. Free Radic Biol Med 46, 105–109. [DOI] [PubMed] [Google Scholar]
- 87.Jaber S, and Polster BM (2015). Idebenone and neuroprotection: antioxidant, pro-oxidant, or electron carrier? J Bioenerg Biomembr 47, 111–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Ross D, and Siegel D (2021). The diverse functionality of NQO1 and its roles in redox control. Redox Biol 41, 101950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Gauthier LD, Greenstein JL, O’Rourke B, and Winslow RL (2013). An integrated mitochondrial ROS production and scavenging model: implications for heart failure. Biophys J 105, 2832–2842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Weyer G, Babej-Dolle RM, Hadler D, Hofmann S, and Herrmann WM (1997). A controlled study of 2 doses of idebenone in the treatment of Alzheimer’s disease. Neuropsychobiology 36, 73–82. [DOI] [PubMed] [Google Scholar]
- 91.Huang Y, Ma M, Zhu X, Li M, Guo M, Liu P, He Z, and Fu Q (2021). Effectiveness of idebenone nanorod formulations in the treatment of Alzheimer’s disease. J Control Release 336, 169–180. [DOI] [PubMed] [Google Scholar]
- 92.Park JS, Leem YH, Park JE, Kim DY, and Kim HS (2019). Neuroprotective Effect of beta-Lapachone in MPTP-Induced Parkinson’s Disease Mouse Model: Involvement of Astroglial p-AMPK/Nrf2/HO-1 Signaling Pathways. Biomol Ther (Seoul) 27, 178–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Mokarizadeh N, Karimi P, Erfani M, Sadigh-Eteghad S, Fathi Maroufi N, and Rashtchizadeh N (2020). beta-Lapachone attenuates cognitive impairment and neuroinflammation in beta-amyloid induced mouse model of Alzheimer’s disease. Int Immunopharmacol 81, 106300. [DOI] [PubMed] [Google Scholar]
- 94.Wiseman H, and Halliwell B (1996). Damage to DNA by reactive oxygen and nitrogen species: role in inflammatory disease and progression to cancer. Biochem J 313 (Pt 1), 17–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Forster MJ, Dubey A, Dawson KM, Stutts WA, Lal H, and Sohal RS (1996). Age-related losses of cognitive function and motor skills in mice are associated with oxidative protein damage in the brain. Proc Natl Acad Sci U S A 93, 4765–4769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Gyure KA, Durham R, Stewart WF, Smialek JE, and Troncoso JC (2001). Intraneuronal abeta-amyloid precedes development of amyloid plaques in Down syndrome. Arch Pathol Lab Med 125, 489–492. [DOI] [PubMed] [Google Scholar]
- 97.Gouras GK, Tsai J, Naslund J, Vincent B, Edgar M, Checler F, Greenfield JP, Haroutunian V, Buxbaum JD, Xu H, et al. (2000). Intraneuronal Abeta42 accumulation in human brain. Am J Pathol 156, 15–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Hartmann T (1999). Intracellular biology of Alzheimer’s disease amyloid beta peptide. Eur Arch Psychiatry Clin Neurosci 249, 291–298. [DOI] [PubMed] [Google Scholar]
- 99.Wilson CA, Doms RW, and Lee VM (1999). Intracellular APP processing and A beta production in Alzheimer disease. J Neuropathol Exp Neurol 58, 787–794. [DOI] [PubMed] [Google Scholar]
- 100.Brewer GJ, Herrera RA, Philipp S, Sosna J, Reyes-Ruiz JM, and Glabe CG (2020). Age-Related Intraneuronal Aggregation of Amyloid-beta in Endosomes, Mitochondria, Autophagosomes, and Lysosomes. J Alzheimers Dis 73, 229–246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Caspersen C, Wang N, Yao J, Sosunov A, Chen X, Lustbader JW, Xu HW, Stern D, McKhann G, and Yan SD (2005). Mitochondrial Abeta: a potential focal point for neuronal metabolic dysfunction in Alzheimer’s disease. FASEB J 19, 2040–2041. [DOI] [PubMed] [Google Scholar]
- 102.Bayer TA, and Wirths O (2010). Intracellular accumulation of amyloid-Beta - a predictor for synaptic dysfunction and neuron loss in Alzheimer’s disease. Front Aging Neurosci 2, 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Ren JX, Sun X, Yan XL, Guo ZN, and Yang Y (2020). Ferroptosis in Neurological Diseases. Front Cell Neurosci 14, 218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Li J, Cao F, Yin HL, Huang ZJ, Lin ZT, Mao N, Sun B, and Wang G (2020). Ferroptosis: past, present and future. Cell Death Dis 11, 88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Hagemeier J, Geurts JJ, and Zivadinov R (2012). Brain iron accumulation in aging and neurodegenerative disorders. Expert Rev Neurother 12, 1467–1480. [DOI] [PubMed] [Google Scholar]
- 106.Hare D, Ayton S, Bush A, and Lei P (2013). A delicate balance: Iron metabolism and diseases of the brain. Front Aging Neurosci 5, 34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Reed TT (2011). Lipid peroxidation and neurodegenerative disease. Free Radic Biol Med 51, 1302–1319. [DOI] [PubMed] [Google Scholar]
- 108.Chen X, Yu C, Kang R, and Tang D (2020). Iron Metabolism in Ferroptosis. Front Cell Dev Biol 8, 590226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Prior M, Chiruta C, Currais A, Goldberg J, Ramsey J, Dargusch R, Maher PA, and Schubert D (2014). Back to the future with phenotypic screening. ACS Chem Neurosci 5, 503–513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Winkler BS, Sauer MW, and Starnes CA (2003). Modulation of the Pasteur effect in retinal cells: implications for understanding compensatory metabolic mechanisms. Exp Eye Res 76, 715–723. [DOI] [PubMed] [Google Scholar]
- 111.Maher P, and Hanneken A (2008). Flavonoids protect retinal ganglion cells from ischemia in vitro. Exp Eye Res 86, 366–374. [DOI] [PubMed] [Google Scholar]
- 112.Taylor BM, Fleming WE, Benjamin CW, Wu Y, Mathews WR, and Sun FF (1996). The mechanism of cytoprotective action of lazaroids I: Inhibition of reactive oxygen species formation and lethal cell injury during periods of energy depletion. J Pharmacol Exp Ther 276, 1224–1231. [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Table S1. Anti-oxytotic/ferroptotic activity of all the compounds in the library.
Table S2. Anti-oxytotic/ferroptotic activity of the best 58 compounds in the library.
