Next Article in Journal
Indole Diterpene Derivatives from the Aspergillus flavus GZWMJZ-288, an Endophytic Fungus from Garcinia multiflora
Previous Article in Journal
Effect of Surface Pt Doping on the Reactivity of Au(111) Surfaces towards Methanol Dehydrogenation: A First-Principles Density Functional Theory Investigation
Previous Article in Special Issue
Antifungal Constituents of Piper crocatum and Their Activities as Ergosterol Biosynthesis Inhibitors Discovered via In Silico Study Using ADMET and Drug-Likeness Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Effects of Natural Products on Enzymes Involved in Ferroptosis: Regulation and Implications

by
Hua-Li Zuo
1,2,*,
Hsi-Yuan Huang
1,2,
Yang-Chi-Dung Lin
1,2,
Kun-Meng Liu
3,
Ting-Syuan Lin
1,2,
Yi-Bing Wang
1,2 and
Hsien-Da Huang
1,2,*
1
School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
2
Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
3
Center for Medical Artificial Intelligence, Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao 266112, China
*
Authors to whom correspondence should be addressed.
Molecules 2023, 28(23), 7929; https://doi.org/10.3390/molecules28237929
Submission received: 30 September 2023 / Revised: 18 November 2023 / Accepted: 30 November 2023 / Published: 4 December 2023
(This article belongs to the Special Issue Discovery of Enzyme Inhibitors from Natural Products II)

Abstract

:
Ferroptosis is a form of regulated cell death that is characterized by the accumulation of iron-dependent lipid peroxides. The regulation of ferroptosis involves both non-enzymatic reactions and enzymatic mechanisms. Natural products have demonstrated potential effects on various enzymes, including GPX4, HO-1, NQO1, NOX4, GCLC, and GCLM, which are mainly involved in glutathione metabolic pathway or oxidative stress regulation, and ACSL3 and ACSL4, which mainly participate in lipid metabolism, thereby influencing the regulation of ferroptosis. In this review, we have provided a comprehensive overview of the existing literature pertaining to the effects of natural products on enzymes involved in ferroptosis and discussed their potential implications for the prevention and treatment of ferroptosis-related diseases. We also highlight the potential challenge that the majority of research has concentrated on investigating the impact of natural products on the expression of enzymes involving ferroptosis while limited attention is given to the regulation of enzyme activity. This observation underscores the considerable potential and scope for exploring the influence of natural products on enzyme activity.

1. Introduction

Several instances of ferroptosis were documented prior to 2012, when a comprehensive molecular comprehension of ferroptosis was established. Ferroptosis, a type of regulated cell death (RCD), is governed by both non-enzymatic (Fenton reaction) and enzymatic mechanisms [1]. It is morphologically, genetically, and biochemically distinct from other forms of RCD, such as apoptosis, necroptosis, and autophagy. This process relies on iron and is characterized by a reduction in glutathione levels and the accumulation of lipid peroxides, which are lipids that have undergone oxidative degradation [2,3]. The intricate process of ferroptosis is governed by a multitude of biological mechanisms, encompassing numerous enzymes and pertinent molecules such as iron, amino acids, and polyunsaturated fatty acids (PUFAs). Additionally, the interplay between glutathione (GSH), NADPH, coenzyme Q10 (CoQ10), and phospholipids further contributes to the regulation of ferroptosis, impacting various cellular metabolic pathways including redox homeostasis, iron homeostasis, and lipid metabolism [4].
Ferroptosis plays a pivotal role in tumor suppression and can reverse drug resistance in cancer therapy [5,6], and it has also been implicated in various non-cancer pathological conditions, such as neurodegeneration [7], ischemia-reperfusion injury [8], and COVID-19 [9].
Having grown rapidly in recent years, ferroptosis has gradually established an interaction with natural products, which are conventional resources for drug discovery and modification. Enzymatic reactions are interconnected, and the delicate equilibrium between them ultimately determines whether a cell undergoes ferroptosis or manages to survive. Previous studies have demonstrated the therapeutic potential of natural products in the treatment and prevention of various diseases, with their effectiveness attributed to their ability to induce apoptosis, inhibit angiogenesis, suppress cell growth, regulate inflammation, and modulate oxidative stress [10,11,12]. In addition, recent research has also begun to establish a connection between ferroptosis and natural products [13]. Consequently, it is crucial to comprehend the molecular mechanisms underlying ferroptosis and identify potential modulators derived from natural products to advance the development of novel therapeutic strategies. Botanical drugs contain a rich source of naturally bioactive compounds considered conventional sources for drug discovery and modification and can affect various cellular processes, including ferroptosis [14].
This review aims to provide a comprehensive overview of the existing literature regarding the impact of natural products on the enzymatic activities associated with ferroptosis. Specifically, we focus on the enzymes GPX4, HO-1, NQO1, NOX4, GCLC, and GCLM, which primarily contribute to glutathione metabolism and regulation of oxidative stress; and ACSL3, and ACSL4, which mainly participate in lipid metabolism. We also highlight the potential applications and challenges, current advances of the emerging regulatory mechanisms and disease relevance of ferroptosis.

2. Results and Discussion

2.1. Search Results and Study Inclusion

The search results were combined and duplicates removed to include 278 papers reporting the natural products’ activity on enzymes involved in ferroptosis. After removing the sixty-five papers that were not research articles, and four papers were not concluded in the life sciences, we read the abstracts of the remaining two-hundred-and-nine articles for relevance, specifically focusing on the enzymes associated with ferroptosis as indicated in Table 1, based on prior knowledge. Subsequently, a more thorough analysis led to the exclusion of 147 articles due to their lack of relevance, and one article was excluded due to unavailability of the full text. As a result of this literature screening process, a total of 61 full-text studies were deemed suitable for inclusion in this review (Figure 1).

2.2. Study Characteristics

The included 61 studies mainly reported the activity of natural products on the enzymes included GPX4, HO-1 (encoded by HMOX1), ACSL4, GCLC, GCLM, NQO1, NOX4, and ACSL3 (Figure 2). GPX4 attracts considerable scholarly interest, possibly due to the fact that the concept of ferroptosis is predominantly defined as a form of cell demise dependent on iron and triggered by oxidative stress-induced dysfunction of phospholipids. GPX4, being a phospholipid hydroperoxidase, plays a crucial role in preventing lipid peroxidation and irreversible cellular demise when inactivated [38].

2.3. The Molecular Mechanisms of Ferroptosis

The molecular regulatory mechanisms governing ferroptosis are intricate, entailing various biological processes, including extrinsic: iron metabolism, transport of component of GSH; and intrinsic: lipid metabolism/peroxidation iron metabolism [39]. The initiation of the extrinsic pathway is facilitated by the regulation of transporters, specifically those responsible for iron and lipid transport. These transporters govern the cellular absorption of iron and the synthesis of PUFAs. The accumulation of iron and PUFAs subsequently triggers lipid peroxidation, which plays a pivotal role in the occurrence of ferroptosis. In contrast, the intrinsic pathway primarily arises from the inhibition of intracellular antioxidant enzymes, such as GPX4, which play a crucial role in safeguarding cells against oxidative harm by converting lipid hydroperoxides into their respective alcohols.

2.4. Enzymes-Mediated Phospholipid Peroxidation

PUFAs are prone to oxidative attack at the carbon-carbon double bonds of lipids, a process known as “lipid peroxidation”, and play a crucial role in the initiation and execution of ferroptosis [40,41]. The biosynthesis and modification of PUFAs involve the actions of two enzymes, namely ACSL4 and LPCAT3, which facilitate the incorporation of PUFA into membrane phospholipids (PL-PUFA) and the formation of phosphatidylethanolamine (PEs)-containing PL-PUFA [29]. In detail, ACSL4 catalyzes the conversion of free PUFAs, such as arachidonic acid (AA) and adrenic acid (AdA), into their corresponding CoA esters. Subsequently, LPCAT3 incorporates these PUFA-CoAs into phospholipids, leading to the production of PEs enriched with PUFA. These PEs are particularly vulnerable to peroxidation [6]. In the presence of oxidative or energetic stress, these enzymes facilitate the synthesis of PUFA-containing phospholipids and trigger ferroptosis as a means to sustain redox homeostasis.
Although the precise mechanisms by which PL-PUFAs (PE) initiate lipid peroxidation remain incompletely understood, the influence on the formation of phospholipid hydroperoxide derivatives of polyunsaturated fatty acids (PUFA-OOHs) is widely recognized to arise from both non-enzymatic and enzymatic processes [31]. These derivatives have been observed to disrupt the structural integrity of cellular membranes, disturb their characteristics, induce oxidative harm to cellular structures, and ultimately lead to ferroptotic cell death.
Furthermore, the generation of PUFA-OOHs can be facilitated by the production of reactive oxygen species (ROS) through the Fenton reaction, wherein iron acts as a catalyst to generate alkoxyl and peroxyl radicals, thereby promoting the synthesis of PUFA-OOHs (elaborated upon subsequently). Consequently, intracellular and intercellular signaling processes, along with diverse metabolic stresses, can modulate ferroptosis by regulating cellular oxidative stress and levels of ROS. For instance, the activation of antioxidant defense systems, such as peroxiredoxins, superoxide dismutase, catalase, GPXs, and Nrf2 (HO-1 regulated by Nrf2), or the production of non-enzymatic antioxidants (e.g., coenzyme Q10, tetrahydrobiopterin), can effectively reduce ROS and inhibit the occurrence of ferroptosis. Additionally, PL-PUFAs can undergo direct oxidation by lipoxygenases (LOXs), which are non-heme iron-dependent dioxygenases, and by prostaglandin-endoperoxide synthase (PTGS/COX), resulting in the generation of PUFA-OOHs and the initiation of ferroptosis.

2.5. The Effects of Natural Products on the Enzymes Involving Ferroptosis

A wide range of phytochemicals exhibiting various biological activities have been employed as dietary interventions for the purpose of managing human diseases. Specifically, certain bioactive components derived from different plant sources have been recognized as influential regulators of ferroptosis, both in vitro and in vivo. The natural products’ functions and their molecular mechanisms regulating enzymes involving ferroptosis were summarized, as shown in Table 2.

2.6. The Effects of Natural Products on the Enzymes Involving Glutathione Metabolic Pathway

2.6.1. GPX4

GPX4, a phospholipid hydroperoxidase, is crucial in safeguarding cells from membrane lipid peroxidation. It is classified within the glutathione peroxidase family, encompassing eight recognized mammalian isoenzymes (GPX1-8). The primary function of GPX4 involves the reduction of hydrogen peroxide, organic hydroperoxides, and lipid peroxides, utilizing reduced glutathione as a substrate. This enzymatic activity is essential for cellular defense against oxidative stress [40]. The equilibrium between reduced glutathione (GSH) and oxidized glutathione (GSSG) is a crucial intracellular antioxidant system that plays a critical role in cell survival. Disruption of this balance leads to the accumulation of ROS and ultimately results in cell death. It is worth noting GPX4 utilizes GSH as a cofactor to convert potentially harmful PUFA-OOHs into non-toxic lipid alcohols (PUFA-OHs). Consequently, the inhibition of GPX4 through direct pharmacological inhibitors, or the depletion of GSH induces excessive lipid peroxidation and triggers ferroptotic cell death.
In previously reported studies, the most focused diseases, included various cancers (breast cancer, colorectal cancer, gastric cancer, liver cancer, lung cancer, esophageal cancer), myocardial infarction/ischemic stroke, Alzheimer’s disease (AD), or insulin secretion dysfunction, etc. Additionally, most of the natural products’ modulation on GPX4 was evaluated by mRNA or protein expression level. For disease treatment, the mRNA or protein levels of GPX4 are expected to be lower or down-regulated, when designed natural products’ treatment is given, consistent with most of the relevant research, as detailed information listed in Table 2.
What is more, there were some, though only a small part of these studies, that also investigated the on-target enzymatic activity or direct binding potential. The cellular glutathione peroxidase assay kit was used to evaluate the enzymatic activity of GPX4 to investigate the effects of glycyrrhetinic acid (GA) extracted from Licorice (Glycyrrhiza glabra) on the promotion of ROS and reactive nitrogen species (RNS) production. This activation was achieved through the activation of NADPH oxidases and inducible nitric oxide synthase (iNOS), while simultaneously decreasing the production of glutathione (GSH) and the activity of glutathione peroxidase (GPX4). Consequently, these actions led to the exacerbation of lipid peroxidation and the initiation of ferroptosis in triple-negative breast cancer (TNBC) cells [52]. Another study was conducted to examine the impact of a small molecule (DMOCPTL) on GPX4 for the treatment of TNBC. This study utilized an on-target binding assay to evaluate the activity of DMOCPTL. The assay investigating cell death mechanisms revealed DMOCPTL predominantly induces ferroptosis and apoptosis to exert its anti-TNBC effect, accomplished through the ubiquitination of GPX4. The probe employed in the DMOCPTL assay furnished evidence DMOCPTL induced GPX4 ubiquitination by directly interacting with the GPX4 protein [16].

2.6.2. HO-1

HO-1, which is regulated by the transcription factor Nrf2, acts as a critical mediator in ferroptosis, catalyzes the degradation of heme, a pro-oxidant molecule, into biliverdin/bilirubin, carbon monoxide, and ferrous iron. The key role of HO-1 in heme metabolism makes it an important enzyme in ferroptosis. Initially, the release of ferrous iron during this process has the potential to participate in the Fenton reaction, resulting in the generation of ROS. These ROS can subsequently be counteracted by glutathione, a prominent cellular antioxidant. Furthermore, the biliverdin synthesized by HO-1 undergoes swift conversion into bilirubin through the action of biliverdin reductase. Both biliverdin and bilirubin possess robust antioxidant properties, thereby aiding in the safeguarding of cells against oxidative stress. Hence, although HO-1 does not participate directly in the synthesis or recycling of glutathione, it assumes a pivotal role in preserving cellular redox equilibrium and safeguarding cells from oxidative stress, functions that are also fundamental to the glutathione metabolic pathway [100].
However, recent research has revealed a potential downside to HO-1, as its dual functionality appeared to be contingent upon the levels of cellular iron and ROS. In instances of excessive cellular iron and ROS, HO-1 transitions from a protective agent to a perpetrator. This shift is supported by evidence from experiments involving HO-1 knockdown mice and the use of HO-1 inhibitors, which demonstrate HO-1 facilitates erastin-induced ferroptosis and is linked to iron bioavailability [101,102].
Similar to the studies on GPX4, for natural products’ regulation on HO-1, the research also mainly conducted to evaluate the stimulations’ affection on mRNA and protein expression level, mainly up-regulated for treatment or beneficial effects.

2.6.3. NQO1

NQO1 has enzymatic functions catalyzing and reverting a wide variety of compounds, including quinones, nitroaromatic compounds, imidazoles, and iron ions [103,104], and plays a role in ferroptosis.
Study from Xu’s group indicated plumbagin, a potent bioactive compound present in roots, stems, and leaves of the genus Plumbago, could target NQO1/GPX4-mediated ferroptosis to suppress in vitro and in vivo glioma growth [105]. Also, a recent study from Chen’s group demonstrated 2-methoxy-6-acetyl-7-methyljuglone (MAM), a natural naphthoquinone, induced ferroptosis triggered by NQO1 in both chemotherapeutic drug (cisplatin) resistant and targeted drug (AZD9291) resistant NSCLC cells [106].These suggested NQO1 could be a potential target for developing new therapies for gliomas, drug resistant cancers, or other conditions where ferroptosis played a role.
In addition, some research has also shown NQO1 could suppress ferroptosis when it functioned as a coenzyme for tanshinone, a compound from Salvia miltiorrhiza (Danshen). Tanshinone helps NQO1 detoxify lipid peroxyl radicals and inhibit ferroptosis both in vitro and in vivo [107]. And, some other quinones derived from Salvia miltiorrhiza or Tabebuia impetiginosa, which may benefit Alzheimer’s disease (AD), were identified as substrates of these proteins through the NADH decay assay. Notably, the presence of NQO1 or FSP1 resulted in an observed augmentation of anti-lipid peroxidation activity in the liposomes [99].

2.6.4. NOX4

The generation of superoxide anion and reactive oxygen species by NADPH oxidase (NOX), through the consumption of NADPH, represents a significant contributor to oxidative stress. The overexpression of NOX results in the depletion of NADPH, the accumulation of intracellular oxidative stress, and heightened vulnerability to ferroptosis. Conversely, NOX expression or activity inhibition can confer resistance or insensitivity to ferroptosis upon cells. NOX4 is one of the members of the NADPH oxidase family. Research has shown NOX4 is associated with both immunity and ferroptosis [108]. In the context of specific diseases, NOX4 has been found to promote ferroptosis of astrocytes by oxidative stress-induced lipid peroxidation, and a previous study reported a high expression of NOX4 was associated with poor prognosis in colon cancer, indicating NOX4 may act as a biomarker for colon cancer ferroptosis [109]. But the studies on the effect of natural products on NOX4 are limited.
Tectorigenin, a main component derived from Belamcanda chinensis, has proved to possess multiple pharmacological activities, including improving kidney injury caused by ferroptosis by mediating the oxidative stress response. In the in vitro investigation on chronic kidney disease treatment, tectorigenin stimulation significantly inhibited the transcription and protein level of NOX4, indicating tectorigenin could confer resistance or insensitivity to ferroptosis [81].

2.6.5. GCLC and GCLM

GCLC and GCLM are two subunits of glutamate-cysteine ligase (GCL), the enzyme responsible for limiting the rate of glutathione (GSH) synthesis, a crucial antioxidant in cellular function [102]. Prior research has demonstrated the knockout of GCLC in mice resulted in a complete deficiency of GSH and embryonic mortality, while the knockout of GCLM led to a significant reduction in cellular GSH levels.
The reported natural products’ regulation on GCLC and GCLM mainly focused on cancer treatment (including colorectal cancer, esophageal cancer, and gastric cancer), or diabetic retinopathy (DR). For treating colorectal cancer, andrographolide, or andrographis extract, derived from Andrographis paniculata, all showed potential in treating colorectal cancer, potential mechanisms including upregulation of GCLC and GCLM at mRNA level, both in vitro and in vivo [95,96]. Andrographolide also showed potential in gastric cancer in vitro by altering HMOX1, GCLC, and GCLM gene expression [93]. In the context of esophageal cancer treatment, it has been observed Oridonin (Ori) possessed the capability to establish a covalent bond with cysteine, resulting in the formation of a conjugate referred to as oridonin-cysteine (Ori-Cys). This interaction leads to the inhibition of glutathione synthesis, which aligns with the observed decline in the enzymatic activity of GCLC. Consequently, there is a reduction in the intracellular ratio of GSH/GSSG, accompanied by a significant decrease in the enzymatic activity of GPX4 [80]. For diabetic retinopathy (DR), astragaloside-IV (AS-IV) could increase the expression of GPX4, GCLC, and GCLM in vitro [86].

2.7. The Effects of Natural Products on Enzymes Involving Lipid Metabolism

ACSL3 and ACSL4 are enzymes that belong to the long-chain fatty acyl CoA synthetase family (ACSLs), which contribute significantly to lipid metabolism [30]. ACSL4 is a positive regulator in ferroptosis. On the other hand, ACSL3 contributes to cancer cells acquiring resistance to ferroptosis. This means that while ACSL4 promotes ferroptosis, ACSL3 helps cancer cells resist this process [110].
Epimedium koreanum Nakai, one of the species of Epimedii Folium, is widely used worldwide as an herbal supplement but has a severe propensity for hepatotoxicity. The in vivo study investigating the mechanism of its hepatotoxicity identified the ferroptosis-promoting protein ACSL4 was significantly up-regulated, and GPX4 and System Xc were significantly down-regulated, indicating the mechanism of hepatotoxicity of Epimedium koreanum Nakai may related to the induction of ferroptosis in hepatocytes [82].
Icariin (ICA), a compound derived from Epimedii, may be responsible for the plant’s toxicity, the results obtained from both in vitro and in vivo experiments demonstrated ICA exerted a significant down-regulatory effect on the protein expression of PPAR-α. Additionally, it down-regulated the expression of APOB, ACSL3, ACSL4, and up-regulated 5/12/15-HETE, indicating the involvement of a lipid metabolism disorder in the nephrotoxicity induced by ICA. Furthermore, ICA was observed to enhance the accumulation of iron and levels of lipid peroxidation (LPO), while simultaneously reducing the activity of GPX4 [83]. In the contrast, ICA also shared the beneficial effect on myocardial hypoxia/reoxygenation (H/R) injury, since the in vitro study demonstrated a significant reduction in mRNA expressions and protein levels of Nrf2 and HO-1 following H/R stimulation, which were subsequently elevated with increasing concentrations of ICA. Additionally, the expression of ACSL4 decreased with increasing ICA concentration, while the expression of GPX4 increased [76].
Except for above research focused on the regulation of the mRNA or protein expression on ACSL3 or ACSL4, there is also a study investigating the enzymatic activity of ACSL4. Silibinin (SIL), a compound derived from Silybum marianum, exhibited a protective effect on HepG2 cells against ferroptosis, which was found to be dependent on ACSL4. To validate this interaction, biophysical assays and a SIL derivatized chemical probe were employed, confirming the binding capability of SIL to ACSL4. Subsequent enzymatic assays demonstrated SIL effectively inhibited the enzymatic activity of ACSL4, consequently attenuating the ACSL4-mediated ferroptosis [98].

3. Materials and Methods

3.1. Data Retrieval

The Web of ScienceTM (ClarivateTM, London, UK) platform was used for literature survey to achieve a dataset of studies reporting the effects of natural products on enzymes contributing to ferroptosis. The search query was performed in WOS core collection based on keywords: ferroptosis, enzyme, and herbal product. Then, we conducted the search strategy: All Fields (ferroptosis) AND All Fields (herb* OR herbal-products OR herbal-supplements OR natural-supplements OR botanical-supplements OR phytotherapy OR dietary-supplementation OR plant-extract OR traditional-medicine OR natural-product OR Botanical*), and in life/medical science focused.

3.2. Screening and Eligibility

According to the selection criteria outlined in the preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines [111,112], publications were retrieved from the WOS database, and all search results were compiled and systematically reviewed, with each source being carefully evaluated and labeled as either included or excluded in an excel spreadsheet. Initially, duplicates were identified and removed. Subsequently, eligibility criteria were established prior to the commencement of this review. As shown in Table 3.

3.3. Annotated Bibliography

Upon completion of the initial screening process, all papers meeting the eligibility criteria were downloaded. Subsequently, we meticulously examined the abstract or entire text of each research article to ascertain its continued relevance. Furthermore, we annotated each eligible research article by manually incorporating the retrieved information, encompassing the natural products investigated in each paper (i.e., ingredients, including what Chinese medicine or natural product the compound is derived from), herbal extracts, or formulae/preparations.), the tested enzymes, the cell line, the animal model used in each study, the biomolecules involved in each study, the specific mechanism of action discovered in each study, and the diseases that concerned each study. The statistical data were then generated to describe the current status of research in this field, such as the disease, natural products that were commonly concerned, and the enzymes that were most likely affected.

4. Conclusions

Ferroptosis is characterized as a controlled mechanism of membrane degradation resulting from the impairment of the lipid redox protection system and the disruption of iron homeostasis. Notably, ferroptosis possesses a dual nature, capable of either eliminating pathological cells to uphold bodily equilibrium and continuity, or inflicting harm upon normal cells, leading to illness. Exogenous stimuli and substances, including various natural products, exert distinct effects on the promotion or prevention of ferroptosis depending on the specific pathological context.
Up to now, the majority of research has concentrated on investigating the impact of natural products on the expression of enzymes involving ferroptosis, while limited attention has been given to the regulation of enzyme activity. This observation underscores the considerable potential and scope for exploring the influence of natural products on enzyme activity. Through extensive screening and analysis of various natural products, this review has provided several promising substances that exhibit noteworthy regulatory effects, elucidating their underlying mechanisms and modes of action. These findings offer novel insights and a foundation for future advancements and applications in the field of natural product development and utilization.

Author Contributions

Conceptualization, H.-L.Z. and H.-D.H.; methodology, H.-L.Z.; data analysis, H.-L.Z. and K.-M.L.; validation, H.-Y.H. and Y.-C.-D.L.; data curation, H.-L.Z., T.-S.L. and Y.-B.W.; writing—original draft preparation, H.-L.Z.; writing—review and editing, H.-L.Z., K.-M.L., T.-S.L., Y.-B.W., H.-Y.H., Y.-C.-D.L. and H.-D.H.; supervision, H.-D.H.; project administration, H.-L.Z.; funding acquisition, H.-L.Z., K.-M.L. and H.-D.H. All authors have read and agreed to the published version of the manuscript.

Funding

This work was financially supported by Key Program of Guangdong Basic and Applied Basic Research Fund (Guangdong–Shenzhen Joint Fund), grant number 2020B1515120069, the Natural Science Foundation of Shandong Province, grant number ZR2022QG051, the science and technology of traditional Chinese medicine project of Shandong Province, grant number Q-2023045, the China Postdoctoral Science Foundation, grant number 2021M693103. This research was also supported by funding from Shenzhen City and Longgang District for the Warshel Institute for Computational Biology, and Guangdong Young Scholar Development Fund of Shenzhen Ganghong Group Co., Ltd.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are contained within the article.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Tang, D.; Chen, X.; Kang, R.; Kroemer, G. Ferroptosis: Molecular mechanisms and health implications. Cell Res. 2021, 31, 107–125. [Google Scholar] [CrossRef] [PubMed]
  2. Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef] [PubMed]
  3. Li, J.; Cao, F.; Yin, H.L.; Huang, Z.J.; Lin, Z.T.; Mao, N.; Sun, B.; Wang, G. Ferroptosis: Past, present and future. Cell Death Dis. 2020, 11, 88. [Google Scholar] [CrossRef] [PubMed]
  4. Dixon, S.J.; Stockwell, B.R. The Hallmarks of Ferroptosis. Annu. Rev. Cancer Biol. 2019, 3, 35–54. [Google Scholar] [CrossRef]
  5. Zhang, C.; Liu, X.Y.; Jin, S.D.; Chen, Y.; Guo, R.H. Ferroptosis in cancer therapy: A novel approach to reversing drug resistance. Mol. Cancer 2022, 21, 47. [Google Scholar] [CrossRef] [PubMed]
  6. Lei, G.; Zhuang, L.; Gan, B.Y. Targeting ferroptosis as a vulnerability in cancer. Nat. Rev. Cancer 2022, 22, 381–396. [Google Scholar] [CrossRef]
  7. Ren, J.X.; Sun, X.; Yan, X.L.; Guo, Z.N.; Yang, Y. Ferroptosis in neurological diseases. Front. Cell. Neurosci. 2020, 14, 218. [Google Scholar] [CrossRef]
  8. Zhou, L.X.; Han, S.T.; Guo, J.Y.; Qiu, T.; Zhou, J.Q.; Shen, L. Ferroptosis—A New Dawn in the Treatment of Organ Ischemia–Reperfusion Injury. Cells 2022, 11, 3653. [Google Scholar] [CrossRef]
  9. Peleman, C.; Van Coillie, S.; Ligthart, S.; Choi, S.M.; De Waele, J.; Depuydt, P.; Benoit, D.; Schaubroeck, H.; Francque, S.M.; Dams, K. Ferroptosis and pyroptosis signatures in critical COVID-19 patients. Cell Death Differ. 2023, 30, 2066–2077. [Google Scholar] [CrossRef]
  10. Wang, Y.; Zhong, J.; Bai, J.J.; Tong, R.S.; An, F.F.; Jiao, P.C.; He, L.; Zeng, D.W.; Long, E.W.; Yan, J.F. The application of natural products in cancer therapy by targeting apoptosis pathways. Curr. Drug Metab. 2018, 19, 739–749. [Google Scholar] [CrossRef]
  11. Arulselvan, P.; Fard, M.T.; Tan, W.S.; Gothai, S.; Fakurazi, S.; Norhaizan, M.E.; Kumar, S.S. Role of antioxidants and natural products in inflammation. Oxidative Med. Cell. Longev. 2016, 2016, 5276130. [Google Scholar] [CrossRef]
  12. Rehman, M.U.; Wali, A.F.; Ahmad, A.; Shakeel, S.; Rasool, S.; Ali, R.; Rashid, S.M.; Madkhali, H.; Ganaie, M.A.; Khan, R. Neuroprotective strategies for neurological disorders by natural products: An update. Curr. Neuropharmacol. 2019, 17, 247–267. [Google Scholar] [CrossRef]
  13. Zheng, K.; Dong, Y.; Yang, R.; Liang, Y.F.; Wu, H.Q.; He, Z.D. Regulation of ferroptosis by bioactive phytochemicals: Implications for medical nutritional therapy. Pharmacol. Res. 2021, 168, 105580. [Google Scholar] [CrossRef] [PubMed]
  14. Kibble, M.; Saarinen, N.; Tang, J.; Wennerberg, K.; Mäkelä, S.; Aittokallio, T. Network pharmacology applications to map the unexplored target space and therapeutic potential of natural products. Nat. Prod. Rep. 2015, 32, 1249–1266. [Google Scholar] [CrossRef]
  15. Ma, T.Y.; Du, J.T.; Zhang, Y.F.; Wang, Y.Y.; Wang, B.X.; Zhang, T.H. GPX4-independent ferroptosis—A new strategy in disease’s therapy. Cell Death Discov. 2022, 8, 434. [Google Scholar] [CrossRef]
  16. Ding, Y.H.; Chen, X.P.; Liu, C.; Ge, W.Z.; Wang, Q.; Hao, X.; Wang, M.M.; Chen, Y.; Zhang, Q. Identification of a small molecule as inducer of ferroptosis and apoptosis through ubiquitination of GPX4 in triple negative breast cancer cells. J. Hematol. Oncol. 2021, 14, 1–21. [Google Scholar] [CrossRef]
  17. Amos, A.; Amos, A.; Wu, L.R.; Xia, H. The Warburg effect modulates DHODH role in ferroptosis: A review. Cell Commun. Signal. 2023, 21, 100. [Google Scholar] [CrossRef] [PubMed]
  18. Kuang, F.M.; Liu, J.; Tang, D.L.; Kang, R. Oxidative damage and antioxidant defense in ferroptosis. Front. Cell Dev. Biol. 2020, 8, 586578. [Google Scholar] [CrossRef] [PubMed]
  19. Park, M.W.; Cha, H.W.; Kim, J.; Kim, J.H.; Yang, H.; Yoon, S.; Boonpraman, N.; Yi, S.S.; Yoo, I.D.; Moon, J.S. NOX4 promotes ferroptosis of astrocytes by oxidative stress-induced lipid peroxidation via the impairment of mitochondrial metabolism in Alzheimer’s diseases. Redox Biol. 2021, 41, 101947. [Google Scholar] [CrossRef]
  20. Lin, Z.; Liu, J.; Kang, R.; Yang, M.H.; Tang, D.L. Lipid metabolism in ferroptosis. Adv. Biol. 2021, 5, 2100396. [Google Scholar] [CrossRef]
  21. Gnanapradeepan, K.; Basu, S.; Barnoud, T.; Budina-Kolomets, A.; Kung, C.-P.; Murphy, M.E. The p53 tumor suppressor in the control of metabolism and ferroptosis. Front. Endocrinol. 2018, 9, 124. [Google Scholar] [CrossRef]
  22. Wei, R.R.; Zhao, Y.Q.; Wang, J.; Yang, X.; Li, S.L.; Wang, Y.Y.; Yang, X.Z.; Fei, J.M.; Hao, X.J.; Zhao, Y.H. Tagitinin C induces ferroptosis through PERK-Nrf2-HO-1 signaling pathway in colorectal cancer cells. Int. J. Biol. Sci. 2021, 17, 2703. [Google Scholar] [CrossRef]
  23. Yang, J.W.; Mo, J.; Dai, J.J.; Ye, C.Q.; Cen, W.; Zheng, X.Z.; Jiang, L.; Ye, L.C. Cetuximab promotes RSL3-induced ferroptosis by suppressing the Nrf2/HO-1 signalling pathway in KRAS mutant colorectal cancer. Cell Death Dis. 2021, 12, 1079. [Google Scholar] [CrossRef]
  24. Kang, Y.P.; Mockabee-Macias, A.; Jiang, C.; Falzone, A.; Prieto-Farigua, N.; Stone, E.; Harris, I.S.; DeNicola, G.M. Non-canonical Glutamate-Cysteine Ligase Activity Protects against Ferroptosis. Cell Metab. 2021, 33, 174–189. [Google Scholar] [CrossRef]
  25. Hayano, M.; Yang, W.S.; Corn, C.K.; Pagano, N.C.; Stockwell, B.R. Loss of cysteinyl-tRNA synthetase (CARS) induces the transsulfuration pathway and inhibits ferroptosis induced by cystine deprivation. Cell Death Differ. 2016, 23, 270–278. [Google Scholar] [CrossRef]
  26. Liu, Y.; Wu, D.; Fu, Q.L.; Hao, S.J.; Gu, Y.Z.; Zhao, W.; Chen, S.Y.; Sheng, F.Y.; Xu, Y.L.; Chen, Z.Q.; et al. CHAC1 as a Novel Contributor of Ferroptosis in Retinal Pigment Epithelial Cells with Oxidative Damage. Int. J. Mol. Sci. 2023, 24, 1582. [Google Scholar] [CrossRef]
  27. Chen, X.; Li, J.B.; Kang, R.; Klionsky, D.J.; Tang, D.L. Ferroptosis: Machinery and regulation. Autophagy 2021, 17, 2054–2081. [Google Scholar] [CrossRef] [PubMed]
  28. Xuan, Y.; Wang, H.; Yung, M.M.; Chen, F.; Chan, W.S.; Chan, Y.S.; Tsui, S.K.; Ngan, H.Y.; Chan, K.K.; Chan, D.W. SCD1/FADS2 fatty acid desaturases equipoise lipid metabolic activity and redox-driven ferroptosis in ascites-derived ovarian cancer cells. Theranostics 2022, 12, 3534–3552. [Google Scholar] [CrossRef] [PubMed]
  29. Yuan, Z.H.; Liu, T.; Wang, H.; Xue, L.X.; Wang, J.J. Fatty acids metabolism: The bridge between ferroptosis and ionizing radiation. Front. Cell Dev. Biol. 2021, 9, 675617. [Google Scholar] [CrossRef]
  30. Yang, Y.F.; Zhu, T.; Wang, X.; Xiong, F.; Hu, Z.M.; Qiao, X.H.; Yuan, X.; Wang, D.Q. ACSL3 and ACSL4, distinct roles in ferroptosis and cancers. Cancers 2022, 14, 5896. [Google Scholar] [CrossRef] [PubMed]
  31. Kagan, V.E.; Mao, G.W.; Qu, F.; Angeli, J.P.F.; Doll, S.; Croix, C.S.; Dar, H.H.; Liu, B.; Tyurin, V.A.; Ritov, V.B. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat. Chem. Biol. 2017, 13, 81–90. [Google Scholar] [CrossRef]
  32. Xie, Y.; Hou, W.; Song, X.; Yu, Y.; Huang, J.; Sun, X.; Kang, R.; Tang, D. Ferroptosis: Process and function. Cell Death Differ. 2016, 23, 369–379. [Google Scholar] [CrossRef] [PubMed]
  33. Grant, G.A. D-3-Phosphoglycerate dehydrogenase. Front. Mol. Biosci. 2018, 5, 110. [Google Scholar] [CrossRef] [PubMed]
  34. Meng, Q.F.; Zhang, Y.H.; Hao, S.M.; Sun, H.H.; Liu, B.; Zhou, H.L.; Wang, Y.S.; Xu, Z.X. Recent findings in the regulation of G6PD and its role in diseases. Front. Pharmacol. 2022, 13, 932154. [Google Scholar] [CrossRef]
  35. Lei, P.X.; Bai, T.; Sun, Y.L. Mechanisms of ferroptosis and relations with regulated cell death: A review. Front. Physiol. 2019, 10, 139. [Google Scholar] [CrossRef] [PubMed]
  36. Forcina, G.C.; Dixon, S.J. GPX4 at the crossroads of lipid homeostasis and ferroptosis. Proteomics 2019, 19, 1800311. [Google Scholar] [CrossRef]
  37. Picón, D.F.; Skouta, R. Unveiling the therapeutic potential of squalene synthase: Deciphering its biochemical mechanism, disease implications, and intriguing ties to ferroptosis. Cancers 2023, 15, 3731. [Google Scholar] [CrossRef] [PubMed]
  38. Maiorino, M.; Conrad, M.; Ursini, F. GPx4, lipid peroxidation, and cell death: Discoveries, rediscoveries, and open issues. Antioxid. Redox Signal. 2018, 29, 61–74. [Google Scholar] [CrossRef] [PubMed]
  39. Stockwell, B.R. Ferroptosis turns 10: Emerging mechanisms, physiological functions, and therapeutic applications. Cell 2022, 185, 2401–2421. [Google Scholar] [CrossRef]
  40. Yang, W.S.; SriRamaratnam, R.; Welsch, M.E.; Shimada, K.; Skouta, R.; Viswanathan, V.S.; Cheah, J.H.; Clemons, P.A.; Shamji, A.F.; Clish, C.B. Regulation of ferroptotic cancer cell death by GPX4. Cell 2014, 156, 317–331. [Google Scholar] [CrossRef]
  41. Yang, W.S.; Kim, K.J.; Gaschler, M.M.; Patel, M.; Shchepinov, M.S.; Stockwell, B.R. Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc. Natl. Acad. Sci. USA 2016, 113, E4966–E4975. [Google Scholar] [CrossRef]
  42. Xie, Y.C.; Song, X.X.; Sun, X.F.; Huang, J.; Zhong, M.Z.; Lotze, M.T.; Zeh, H.J., 3rd; Kang, R.; Tang, D.L. Identification of baicalein as a ferroptosis inhibitor by natural product library screening. Biochem. Biophys. Res. Commun. 2016, 473, 775–780. [Google Scholar] [CrossRef]
  43. Liu, Y.; Song, Z.; Liu, Y.J.; Ma, X.B.; Wang, W.; Ke, Y.; Xu, Y.C.; Yu, D.Q.; Liu, H.M. Identification of ferroptosis as a novel mechanism for antitumor activity of natural product derivative a2 in gastric cancer. Acta Pharm. Sin. B 2021, 11, 1513–1525. [Google Scholar] [CrossRef]
  44. Batbold, U.; Liu, J.J. Novel Insights of Herbal Remedy into NSCLC Suppression through Inducing Diverse Cell Death Pathways via Affecting Multiple Mediators. Appl. Sci. 2022, 12, 4868. [Google Scholar] [CrossRef]
  45. He, C.Y.; Wang, C.Z.; Liu, H.S.; Shan, B.E. Kayadiol exerted anticancer effects through p53-mediated ferroptosis in NKTCL cells. BMC Cancer 2022, 22, 724. [Google Scholar] [CrossRef]
  46. Zhang, M.Y.; Zhang, T.; Song, C.L.; Qu, J.; Gu, Y.P.; Liu, S.J.; Li, H.B.; Xiao, W.; Kong, L.D.; Sun, Y. Guizhi Fuling Capsule ameliorates endometrial hyperplasia through promoting p62-Keap1-NRF2-mediated ferroptosis. J. Ethnopharmacol. 2021, 274, 114064. [Google Scholar] [CrossRef]
  47. Greco, G.; Schnekenburger, M.; Catanzaro, E.; Turrini, E.; Ferrini, F.; Sestili, P.; Diederich, M.; Fimognari, C. Discovery of sulforaphane as an inducer of ferroptosis in u-937 leukemia cells: Expanding its anticancer potential. Cancers 2021, 14, 76. [Google Scholar] [CrossRef] [PubMed]
  48. Zhang, X.; Jiang, L.; Chen, H.; Wei, S.; Yao, K.; Sun, X.; Yang, G.; Jiang, L.; Zhang, C.; Wang, N. Resveratrol protected acrolein-induced ferroptosis and insulin secretion dysfunction via ER-stress-related PERK pathway in MIN6 cells. Toxicology 2022, 465, 153048. [Google Scholar] [CrossRef]
  49. Zhang, S.; Cao, S.; Zhou, H.; Li, L.; Hu, Q.; Mao, X.; Ji, S. Realgar-induced nephrotoxicity via ferroptosis in mice. J. Appl. Toxicol. 2022, 42, 1843–1853. [Google Scholar] [CrossRef]
  50. Wang, Y.; Wang, Z.; Wu, Z.; Chen, M.; Dong, D.; Yu, P.; Lu, D.; Wu, B. Involvement of REV-ERBα dysregulation and ferroptosis in aristolochic acid I-induced renal injury. Biochem. Pharmacol. 2021, 193, 114807. [Google Scholar] [CrossRef] [PubMed]
  51. Chang, W.T.; Bow, Y.D.; Fu, P.J.; Li, C.Y.; Wu, C.Y.; Chang, Y.H.; Teng, Y.N.; Li, R.N.; Lu, M.C.; Liu, Y.C.; et al. A Marine Terpenoid, Heteronemin, Induces Both the Apoptosis and Ferroptosis of Hepatocellular Carcinoma Cells and Involves the ROS and MAPK Pathways. Oxid. Med. Cell. Longev. 2021, 2021, 7689045. [Google Scholar] [CrossRef] [PubMed]
  52. Wen, Y.; Chen, H.; Zhang, L.; Wu, M.; Zhang, F.; Yang, D.; Shen, J.; Chen, J. Glycyrrhetinic acid induces oxidative/nitrative stress and drives ferroptosis through activating NADPH oxidases and iNOS, and depriving glutathione in triple-negative breast cancer cells. Free Radic. Biol. Med. 2021, 173, 41–51. [Google Scholar] [CrossRef]
  53. Wang, B.; Ma, W.; Di, Y.L. Activation of the Nrf2/GPX4 Signaling by Pratensein From Trifolium pretense Mitigates Ferroptosis in OGD/R-Insulted H9c2 Cardiomyocytes. Nat. Prod. Commun. 2022, 17, 1934578X221115313. [Google Scholar] [CrossRef]
  54. Li, X.P.; Li, W.; Yang, P.; Zhou, H.G.; Zhang, W.D.; Ma, L. Anticancer effects of Cryptotanshinone against lung cancer cells through ferroptosis. Arab. J. Chem. 2021, 14, 103177. [Google Scholar] [CrossRef]
  55. Tian, L.; Ji, H.; Wang, W.; Han, X.; Zhang, X.; Li, X.; Guo, L.; Huang, L.; Gao, W. Mitochondria-targeted pentacyclic triterpenoid carbon dots for selective cancer cell destruction via inducing autophagy, apoptosis, as well as ferroptosis. Bioorg. Chem. 2023, 130, 106259. [Google Scholar] [CrossRef]
  56. Wang, X.; Wang, Y.; Huang, D.; Shi, S.; Pei, C.; Wu, Y.; Shen, Z.; Wang, F.; Wang, Z. Astragaloside IV regulates the ferroptosis signaling pathway via the Nrf2/SLC7A11/GPX4 axis to inhibit PM2.5-mediated lung injury in mice. Int. Immunopharmacol. 2022, 112, 109186. [Google Scholar] [CrossRef]
  57. Li, J.; Tian, X.; Liu, J.; Mo, Y.; Guo, X.; Qiu, Y.; Liu, Y.; Ma, X.; Wang, Y.; Xiong, Y. Therapeutic material basis and underling mechanisms of Shaoyao Decoction-exerted alleviation effects of colitis based on GPX4-regulated ferroptosis in epithelial cells. Chin. Med. 2022, 17, 96. [Google Scholar] [CrossRef]
  58. Xu, X.; Wei, Y.; Hua, H.; Jing, X.; Zhu, H.; Xiao, K.; Zhao, J.; Liu, Y. Polyphenols Sourced from Ilex latifolia Thunb. Relieve Intestinal Injury via Modulating Ferroptosis in Weanling Piglets under Oxidative Stress. Antioxidants 2022, 11, 966. [Google Scholar] [CrossRef]
  59. Wang, C.; Su, Z.; Xu, J.H.; Ko, C.Y. Danshensu attenuated lipopolysaccharide-induced LX-2 and T6 cells activation through regulation of ferroptosis. Food Sci. Nutr. 2023, 11, 344–349. [Google Scholar] [CrossRef]
  60. Lan, B.; Ge, J.W.; Cheng, S.W.; Zheng, X.L.; Liao, J.; He, C.; Rao, Z.Q.; Wang, G.Z. Extract of Naotaifang, a compound Chinese herbal medicine, protects neuron ferroptosis induced by acute cerebral ischemia in rats. J. Integr. Med. 2020, 18, 344–350. [Google Scholar] [CrossRef]
  61. Huang, S.; Cao, B.; Zhang, J.; Feng, Y.; Wang, L.; Chen, X.; Su, H.; Liao, S.; Liu, J.; Yan, J.; et al. Induction of ferroptosis in human nasopharyngeal cancer cells by cucurbitacin B: Molecular mechanism and therapeutic potential. Cell Death Dis. 2021, 12, 237. [Google Scholar] [CrossRef] [PubMed]
  62. Han, N.; Li, L.G.; Peng, X.C.; Ma, Q.L.; Yang, Z.Y.; Wang, X.Y.; Li, J.; Li, Q.R.; Yu, T.T.; Xu, H.Z.; et al. Ferroptosis triggered by dihydroartemisinin facilitates chlorin e6 induced photodynamic therapy against lung cancerthrough inhibiting GPX4 and enhancing ROS. Eur. J. Pharmacol. 2022, 919, 174797. [Google Scholar] [CrossRef] [PubMed]
  63. Catanzaro, E.; Turrini, E.; Kerre, T.; Sioen, S.; Baeyens, A.; Guerrini, A.; Bellau, M.L.A.; Sacchetti, G.; Paganetto, G.; Krysko, D.V.; et al. Perillaldehyde is a new ferroptosis inducer with a relevant clinical potential for acute myeloid leukemia therapy. Biomed. Pharmacother. 2022, 154, 113662. [Google Scholar] [CrossRef] [PubMed]
  64. Du, X.; Zhang, J.; Liu, L.; Xu, B.; Han, H.; Dai, W.; Pei, X.; Fu, X.; Hou, S. A novel anticancer property of Lycium barbarum polysaccharide in triggering ferroptosis of breast cancer cells. J. Zhejiang Univ. Sci. B 2022, 23, 286–299. [Google Scholar] [CrossRef] [PubMed]
  65. Wang, J.B.; Tang, P.L.; Cai, Q.; Xie, S.F.; Duan, X.Y.; Pan, Y.Z. Matrine can inhibit the growth of colorectal cancer cells by inducing ferroptosis. Nat. Prod. Commun. 2020, 15, 1934578X20982779. [Google Scholar] [CrossRef]
  66. Liu, G.; Wei, C.; Yuan, S.; Zhang, Z.; Li, J.; Zhang, L.; Wang, G.; Fang, L. Wogonoside attenuates liver fibrosis by triggering hepatic stellate cell ferroptosis through SOCS1/P53/SLC7A11 pathway. Phytother. Res. 2022, 36, 4230–4243. [Google Scholar] [CrossRef] [PubMed]
  67. Riccio, G.; Nuzzo, G.; Zazo, G.; Coppola, D.; Senese, G.; Romano, L.; Costantini, M.; Ruocco, N.; Bertolino, M.; Fontana, A.; et al. Bioactivity Screening of Antarctic Sponges Reveals Anticancer Activity and Potential Cell Death via Ferroptosis by Mycalols. Mar. Drugs 2021, 19, 459. [Google Scholar] [CrossRef]
  68. Huang, Q.; Li, J.; Ma, M.; Lv, M.; Hu, R.; Sun, J.; Zhong, X.; Sun, X.; Feng, W.; Ma, W.; et al. High-throughput screening identification of a small-molecule compound that induces ferroptosis and attenuates the invasion and migration of hepatocellular carcinoma cells by targeting the STAT3/GPX4 axis. Int. J. Oncol. 2023, 62, 42. [Google Scholar] [CrossRef]
  69. Tsai, Y.; Xia, C.; Sun, Z. The Inhibitory Effect of 6-Gingerol on Ubiquitin-Specific Peptidase 14 Enhances Autophagy-Dependent Ferroptosis and Anti-Tumor in vivo and in vitro. Front. Pharmacol. 2020, 11, 598555. [Google Scholar] [CrossRef]
  70. Yang, R.; Chen, F.; Xu, H.; Guo, Z.; Cao, C.; Zhang, H.; Zhang, C. Exploring the Mechanism of Realgar against Esophageal Cancer Based on Ferroptosis Induced by ROS-ASK1-p38 MAPK Signaling Pathway. Evid. Based Complement. Alternat. Med. 2022, 2022, 3698772. [Google Scholar] [CrossRef]
  71. Li, L.; Li, W.J.; Zheng, X.R.; Liu, Q.L.; Du, Q.; Lai, Y.J.; Liu, S.Q. Eriodictyol ameliorates cognitive dysfunction in APP/PS1 mice by inhibiting ferroptosis via vitamin D receptor-mediated Nrf2 activation. Mol. Med. 2022, 28, 11. [Google Scholar] [CrossRef] [PubMed]
  72. Chen, P.; Li, X.J.; Zhang, R.N.; Liu, S.P.; Xiang, Y.; Zhang, M.M.; Chen, X.Y.; Pan, T.; Yan, L.L.; Feng, J. Combinative treatment of β-elemene and cetuximab is sensitive to KRAS mutant colorectal cancer cells by inducing ferroptosis and inhibiting epithelial-mesenchymal transformation. Theranostics 2020, 10, 5107. [Google Scholar] [CrossRef] [PubMed]
  73. Song, C.Y.; Feng, M.X.; Li, L.; Wang, P.; Lu, X.; Lu, Y.Q. Tripterygium wilfordii Hook.f. ameliorates paraquat-induced lung injury by reducing oxidative stress and ferroptosis via Nrf2/HO-1 pathway. Ecotoxicol. Environ. Saf. 2023, 252, 114575. [Google Scholar] [CrossRef]
  74. Dai, W.; Pang, X.; Peng, W.; Zhan, X.; Chen, C.; Zhao, W.; Zeng, C.; Mei, Q.; Chen, Q.; Kuang, W.; et al. Liver Protection of a Low-Polarity Fraction from Ficus pandurata Hance, Prepared by Supercritical CO(2) Fluid Extraction, on CCl(4)-Induced Acute Liver Injury in Mice via Inhibiting Apoptosis and Ferroptosis Mediated by Strengthened Antioxidation. Molecules 2023, 28, 2078. [Google Scholar] [CrossRef] [PubMed]
  75. He, Q.; Yang, J.; Pan, Z.; Zhang, G.; Chen, B.; Li, S.; Xiao, J.; Tan, F.; Wang, Z.; Chen, P.; et al. Biochanin A protects against iron overload associated knee osteoarthritis via regulating iron levels and NRF2/System xc-/GPX4 axis. Biomed. Pharmacother. 2023, 157, 113915. [Google Scholar] [CrossRef]
  76. Liu, X.J.; Lv, Y.F.; Cui, W.Z.; Li, Y.; Liu, Y.; Xue, Y.T.; Dong, F. Icariin inhibits hypoxia/reoxygenation-induced ferroptosis of cardiomyocytes via regulation of the Nrf2/HO-1 signaling pathway. FEBS Open Bio 2021, 11, 2966–2976. [Google Scholar] [CrossRef]
  77. Feng, S.; Zhou, Y.; Huang, H.; Lin, Y.; Zeng, Y.; Han, S.; Huang, K.; Liu, Q.; Zhu, W.; Yuan, Z.; et al. Nobiletin Induces Ferroptosis in Human Skin Melanoma Cells Through the GSK3β-Mediated Keap1/Nrf2/HO-1 Signalling Pathway. Front. Genet. 2022, 13, 865073. [Google Scholar] [CrossRef]
  78. Yang, J.H.; Nguyen, C.D.; Lee, G.; Na, C.S. Insamgobonhwan Protects Neuronal Cells from Lipid ROS and Improves Deficient Cognitive Function. Antioxidants 2022, 11, 295. [Google Scholar] [CrossRef]
  79. Shen, Y.; Shen, X.; Wang, S.; Zhang, Y.; Wang, Y.; Ding, Y.; Shen, J.; Zhao, J.; Qin, H.; Xu, Y.; et al. Protective effects of Salvianolic acid B on rat ferroptosis in myocardial infarction through upregulating the Nrf2 signaling pathway. Int. Immunopharmacol. 2022, 112, 109257. [Google Scholar] [CrossRef]
  80. Zhang, J.; Wang, N.; Zhou, Y.; Wang, K.; Sun, Y.; Yan, H.; Han, W.; Wang, X.; Wei, B.; Ke, Y.; et al. Oridonin induces ferroptosis by inhibiting gamma-glutamyl cycle in TE1 cells. Phytother. Res. 2021, 35, 494–503. [Google Scholar] [CrossRef]
  81. Li, J.; Yang, J.; Zhu, B.; Fan, J.; Hu, Q.; Wang, L. Tectorigenin protects against unilateral ureteral obstruction by inhibiting Smad3-mediated ferroptosis and fibrosis. Phytother. Res. 2022, 36, 475–487. [Google Scholar] [CrossRef] [PubMed]
  82. Li, P.; Zhang, L.; Guo, Z.J.; Kang, Q.J.; Chen, C.; Liu, X.Y.; Ma, Q.T.; Zhang, J.X.; Hu, Y.J.; Wang, T. Epimedium koreanum Nakai–Induced Liver Injury—A Mechanistic Study Using Untargeted Metabolomics. Front. Pharmacol. 2022, 13, 934057. [Google Scholar] [CrossRef] [PubMed]
  83. Wang, D.; Liu, J.; Chen, X.; Chen, J.; Zhao, T.; Du, J.; Wang, C.; Meng, Q.; Sun, H.; Wang, F.; et al. Renal transporter OAT1 and PPAR-α pathway co-contribute to icaritin-induced nephrotoxicity. Phytother. Res. 2023, 37, 549–562. [Google Scholar] [CrossRef] [PubMed]
  84. Lin, J.H.; Yang, K.T.; Ting, P.C.; Luo, Y.P.; Lin, D.J.; Wang, Y.S.; Chang, J.C. Gossypol Acetic Acid Attenuates Cardiac Ischemia/Reperfusion Injury in Rats via an Antiferroptotic Mechanism. Biomolecules 2021, 11, 1667. [Google Scholar] [CrossRef]
  85. Jiao, H.; Yang, H.; Yan, Z.; Chen, J.; Xu, M.; Jiang, Y.; Liu, Y.; Xue, Z.; Ma, Q.; Li, X.; et al. Traditional Chinese Formula Xiaoyaosan Alleviates Depressive-Like Behavior in CUMS Mice by Regulating PEBP1-GPX4-Mediated Ferroptosis in the Hippocampus. Neuropsychiatr. Dis. Treat 2021, 17, 1001–1019. [Google Scholar] [CrossRef]
  86. Tang, X.; Li, X.; Zhang, D.; Han, W. Astragaloside-IV alleviates high glucose-induced ferroptosis in retinal pigment epithelial cells by disrupting the expression of miR-138-5p/Sirt1/Nrf2. Bioengineered 2022, 13, 8240–8254. [Google Scholar] [CrossRef]
  87. Li, J.; Lu, K.; Sun, F.; Tan, S.; Zhang, X.; Sheng, W.; Hao, W.; Liu, M.; Lv, W.; Han, W. Panaxydol attenuates ferroptosis against LPS-induced acute lung injury in mice by Keap1-Nrf2/HO-1 pathway. J. Transl. Med. 2021, 19, 96. [Google Scholar] [CrossRef]
  88. Hao, X.; Fan, H.; Yang, J.; Tang, J.; Zhou, J.; Zhao, Y.; Huang, L.; Xia, Y. Network Pharmacology Research and Dual-omic Analyses Reveal the Molecular Mechanism of Natural Product Nodosin Inhibiting Muscle-Invasive Bladder Cancer in Vitro and in Vivo. J. Nat. Prod. 2022, 85, 2006–2017. [Google Scholar] [CrossRef]
  89. Liu, X.; Tian, Y.; Yang, A.; Zhang, C.; Miao, X.; Yang, W. Antitumor Effects of Poplar Propolis on DLBCL SU-DHL-2 Cells. Foods 2023, 12, 283. [Google Scholar] [CrossRef]
  90. Sun, Y.; He, L.; Wang, W.; Xie, Z.; Zhang, X.; Wang, P.; Wang, L.; Yan, C.; Liu, Z.; Zhao, J.; et al. Activation of Atg7-dependent autophagy by a novel inhibitor of the Keap1-Nrf2 protein-protein interaction from Penthorum chinense Pursh. attenuates 6-hydroxydopamine-induced ferroptosis in zebrafish and dopaminergic neurons. Food Funct. 2022, 13, 7885–7900. [Google Scholar] [CrossRef]
  91. Shao, Y.; Sun, L.; Yang, G.; Wang, W.; Liu, X.; Du, T.; Chen, F.; Jing, X.; Cui, X. Icariin protects vertebral endplate chondrocytes against apoptosis and degeneration via activating Nrf-2/HO-1 pathway. Front. Pharmacol. 2022, 13, 937502. [Google Scholar] [CrossRef] [PubMed]
  92. Liu, Z.; Ma, H.; Lai, Z. Revealing the potential mechanism of Astragalus membranaceus improving prognosis of hepatocellular carcinoma by combining transcriptomics and network pharmacology. BMC Complement Med. Ther. 2021, 21, 263. [Google Scholar] [CrossRef] [PubMed]
  93. Ma, R.; Shimura, T.; Yin, C.; Okugawa, Y.; Kitajima, T.; Koike, Y.; Okita, Y.; Ohi, M.; Uchida, K.; Goel, A.; et al. Antitumor effects of Andrographis via ferroptosis-associated genes in gastric cancer. Oncol. Lett. 2021, 22, 523. [Google Scholar] [CrossRef] [PubMed]
  94. Sun, G.; Su, W.; Bao, J.; Teng, T.; Song, X.; Wang, J.; Shi, B. Dietary full-fat rice bran prevents the risk of heart ferroptosis and imbalance of energy metabolism induced by prolonged cold stimulation. Food Funct. 2023, 14, 1530–1544. [Google Scholar] [CrossRef]
  95. Shimura, T.; Sharma, P.; Sharma, G.G.; Banwait, J.K.; Goel, A. Enhanced anti-cancer activity of andrographis with oligomeric proanthocyanidins through activation of metabolic and ferroptosis pathways in colorectal cancer. Sci. Rep. 2021, 11, 7548. [Google Scholar] [CrossRef]
  96. Sharma, P.; Shimura, T.; Banwait, J.K.; Goel, A. Andrographis-mediated chemosensitization through activation of ferroptosis and suppression of β-catenin/Wnt-signaling pathways in colorectal cancer. Carcinogenesis 2020, 41, 1385–1394. [Google Scholar] [CrossRef] [PubMed]
  97. Xia, Y.; Yang, J.; Li, C.; Hao, X.; Fan, H.; Zhao, Y.; Tang, J.; Wan, X.; Lian, S.; Yang, J. TMT-Based Quantitative Proteomics Analysis Reveals the Panoramic Pharmacological Molecular Mechanism of β-Elemonic Acid Inhibition of Colorectal Cancer. Front. Pharmacol. 2022, 13, 830328. [Google Scholar] [CrossRef]
  98. Yan, W.; Wang, D.; Wan, N.; Wang, S.; Shao, C.; Zhang, H.; Zhao, Z.; Lu, W.; Tian, Y.; Ye, H.; et al. Living Cell-Target Responsive Accessibility Profiling Reveals Silibinin Targeting ACSL4 for Combating Ferroptosis. Anal. Chem. 2022, 94, 14820–14826. [Google Scholar] [CrossRef]
  99. Soriano-Castell, D.; Liang, Z.; Maher, P.; Currais, A. Profiling the chemical nature of anti-oxytotic/ferroptotic compounds with phenotypic screening. Free Radic. Biol. Med. 2021, 177, 313–325. [Google Scholar] [CrossRef]
  100. Loboda, A.; Damulewicz, M.; Pyza, E.; Jozkowicz, A.; Dulak, J. Role of Nrf2/HO-1 system in development, oxidative stress response and diseases: An evolutionarily conserved mechanism. Cell. Mol. Life Sci. 2016, 73, 3221–3247. [Google Scholar] [CrossRef]
  101. Chiang, S.K.; Chen, S.E.; Chang, L.C. A Dual Role of Heme Oxygenase-1 in Cancer Cells. Int. J. Mol. Sci. 2019, 20, 39. [Google Scholar] [CrossRef]
  102. Pan, F.; Lin, X.R.; Hao, L.P.; Wang, T.; Song, H.Z.; Wang, R. The critical role of ferroptosis in hepatocellular carcinoma. Front. Cell Dev. Biol. 2022, 10, 882571. [Google Scholar] [CrossRef]
  103. Pey, A.L.; Megarity, C.F.; Timson, D.J. NAD(P)H quinone oxidoreductase (NQO1): An enzyme which needs just enough mobility, in just the right places. Biosci. Rep. 2019, 39, BSR20180459. [Google Scholar] [CrossRef] [PubMed]
  104. Ross, D.; Siegel, D. NAD(P)H:Quinone Oxidoreductase 1 (NQO1, DT-Diaphorase), Functions and Pharmacogenetics. Methods Enzymol. 2004, 382, 115–144. [Google Scholar] [PubMed]
  105. Zhan, S.; Lu, L.; Pan, S.S.; Wei, X.Q.; Miao, R.Q.; Liu, X.H.; Xue, M.; Lin, X.K.; Xu, H.L. Targeting NQO1/GPX4-mediated ferroptosis by plumbagin suppresses in vitro and in vivo glioma growth. Br. J. Cancer 2022, 127, 364–376. [Google Scholar] [CrossRef] [PubMed]
  106. Yu, J.; Zhong, B.l.; Zhao, L.; Hou, Y.; Ai, N.n.; Lu, J.J.; Ge, W.; Chen, X.p. Fighting drug-resistant lung cancer by induction of NAD (P) H: Quinone oxidoreductase 1 (NQO1)-mediated ferroptosis. Drug Resist. Updates 2023, 70, 100977. [Google Scholar] [CrossRef] [PubMed]
  107. Wang, T.X.; Duan, K.L.; Huang, Z.X.; Xue, Z.A.; Liang, J.Y.; Dang, Y.J.; Zhang, A.; Xiong, Y.; Ding, C.Y.; Guan, K.L. Tanshinone functions as a coenzyme that confers gain of function of NQO1 to suppress ferroptosis. Life Sci. Alliance 2023, 6, e202201667. [Google Scholar] [CrossRef] [PubMed]
  108. Lin, L.; Li, X.N.; Zhu, S.D.; Long, Q.S.; Hu, Y.Z.; Zhang, L.Y.; Liu, Z.X.; Li, B.; Li, X.S. Ferroptosis-related NFE2L2 and NOX4 Genes are Potential Risk Prognostic Biomarkers and Correlated with Immunogenic Features in Glioma. Cell Biochem. Biophys. 2023, 81, 7–17. [Google Scholar] [CrossRef]
  109. Yang, X.P.; Yu, Y.; Wang, Z.R.; Wu, P.F.; Su, X.L.; Wu, Z.P.; Gan, J.X.; Zhang, D.K. NOX4 has the potential to be a biomarker associated with colon cancer ferroptosis and immune infiltration based on bioinformatics analysis. Front. Oncol. 2022, 12, 968043. [Google Scholar] [CrossRef] [PubMed]
  110. Li, D.S.; Li, Y.S. The interaction between ferroptosis and lipid metabolism in cancer. Signal Transduct. Target. Ther. 2020, 5, 108. [Google Scholar] [CrossRef]
  111. Liberati, A.; Altman, D.G.; Tetzlaff, J.; Mulrow, C.; Gøtzsche, P.C.; Ioannidis, J.P.; Clarke, M.; Devereaux, P.J.; Kleijnen, J.; Moher, D. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: Explanation and elaboration. J. Clin. Epidemiol. 2009, 62, e1–e34. [Google Scholar] [CrossRef] [PubMed]
  112. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. Int. J. Surg. 2021, 88, 105906. [Google Scholar] [CrossRef] [PubMed]
Figure 1. PRISMA flow diagram detailing the number of papers included at each stage and the reasons for removal.
Figure 1. PRISMA flow diagram detailing the number of papers included at each stage and the reasons for removal.
Molecules 28 07929 g001
Figure 2. Publications reporting the activity of natural products on enzymes involving ferroptosis.
Figure 2. Publications reporting the activity of natural products on enzymes involving ferroptosis.
Molecules 28 07929 g002
Table 1. The enzymes involved in ferroptosis.
Table 1. The enzymes involved in ferroptosis.
PathwayEffect on FerroptosisGene NameUniprot IDProtein NameRef.
Redox homeostasisInhibitGPX4P36969Glutathione peroxidase 4[15,16]
NQO1P15559NAD(P)H Quinone Dehydrogenase 1[17]
PromoteNOX1Q9Y5S8NADPH oxidase 1[18]
NOX4Q9NPH5NADPH oxidase 4[19]
ALOX5P09917Polyunsaturated fatty acid 5-lipoxygenase[20]
DPP4Q9NPH5Dipeptidyl peptidase 4[21]
Iron homeostasis/redox homeostasisHeme metabolismHMOX1P09601Heme oxygenase 1[22,23]
GSH homeostasisInhibitGCLCP48506Glutamate-cysteine ligase catalytic subunit[24]
GCLMP48507Glutamate-cysteine ligase regulatory subunit[24]
PromoteCARSP49589Cysteine-tRNA ligase, cytoplasmic[25]
CHAC1Q9BUX1Glutathione-specific gamma-glutamylcyclotransferase 1[26]
Lipid metabolismInhibitHELLSQ9NRZ9Lymphoid-specific helicase[27]
SCDO00767Stearoyl-CoA desaturase[20]
FADS2O95864Acyl-CoA 6-desaturase[28]
PromoteACSL4O60488Acyl-CoA Synthetase long chain family member 4 [29]
ACSL3O95573Acyl-CoA Synthetase long chain family member 3[30]
LPCAT3Q6P1A2Lysophosphatidylcholine acyltransferase 3[31]
CSO75390Citrate synthase[32]
Glucose metabolismInhibitPHGDHO43175D-3-phosphoglycerate dehydrogenase[33]
G6PDP11413Glucose-6-phosphate 1-dehydrogenase[34]
PromotePHKG2P15735Phosphorylase kinase subunit gamma-2[35]
Mevalonate pathwayPromoteSQLEQ14534Squalene monooxygenase[36]
FDFT1P37268Squalene synthase[37]
Table 2. Natural products’/phytochemicals’ regulation on enzymes involving ferroptosis.
Table 2. Natural products’/phytochemicals’ regulation on enzymes involving ferroptosis.
EnzymeNatural Product
(Compound)
Natural Product
(Herb
/Chinese Medicine)
Cell LineAnimalActivity/MechanismRelevant DiseaseRef.
GPX4Baicalein (5,6,7-trihydroxyflavone)Solanum nigrum L.PANC1, BxPc-3— —At the protein level, baicalein suppresses erastin-mediated degradation of GPX4.Ferroptosis-associated tissue injury[42]
GPX4Jiyuan oridonin A derivative (a2)Jiyuan Rabdosia rubescensHGC-27, MGC-803, BGC-823, AGS, GES1Gastric cancer cell line-derived xenograftmicemodelsCompound a2 decreased GPX4 expression Gastric cancer[43]
GPX4Artemisia santolinifolia ethanol extract (ASE)Artemisia santolinifoliaA549 and H23— —ASE decreased the GPX4 level at a significantly higher rate in ASE concentrations of 200 µg/mL.Non-small cell lung cancer (NSCLC)[44]
GPX4KayadiolTorreya nuciferaNKTCL— —Kayadiol decreased the expression of GPX4. Cancer[45]
GPX4DMOCPTLA derivative of natural product parthenolideMDAMB-231, SUM159, BT574,4T1, Hs578T, MDA-MB-468MouseDMOCPTL directly binding to GPX4 protein, inducing GPX4 ubiquitination.Triple-negative breast cancer (TNBC)[16]
GPX4— —Guizhi Fuling Capsule (GFC)— —C57BL/6 miceGFC could inhibit the expression of GPX4 at protein level.Gynecological diseases[46]
GPX4Isothiocyanate sulforaphaneCruciferous vegetablesU-937, MV4-11— —Isothiocyanate sulforaphane decreased GPX4 protein expression level. Acute myeloid leukemia[47]
GPX4Resveratrol (RSV)— —MIN6— —RSV could increase the expression of GPX4, and decrease the expression of ACSL4.Insulin secretion dysfunction[48]
GPX4ArsenicRealgarHK2MouseRealgar reduced expression of SLC7A11 and GPX4.Nephrotoxicity[49]
GPX4Aristolactam I (ALI)Aristolochia and Asarum plantsmRTECsWild-type C57BL/6 mice and kidney-specific Rev-erbα knockout mice with AAI-induced nephropathyALI treatment led to decreased GPX4 in mRNA and protein level. Nephropathy[50]
GPX4HeteroneminHippospongia sp.HA22T/VGH, HA59T— —Heteronemin treatment reduced the expression of GPX4.Hepatocellular carcinoma (HCC)[51]
GPX4Glycyrrhetinic acid (GA)Licorice (Glycyrrhiza glabra)MDA-MB-231, BT-549, and MCF-10A— —GA down-regulated the expression of SLC7A11 at protein level; GA treatment (40 μM) significantly inhibited the activity of GPX4 but no influence on GPX4 expression.Triple-negative breast cancer (TNBC)[52]
GPX4Pratensein (PTS)Trifolium pretense L.H9c2— —PTS treatment up-regulated Nrf2 expression and GPX4 expression at protein level in OGD/R-treated H9c2 cells. Myocardial infarction (MI), myocardial ischemia-reperfusion (I/R) injury[53]
GPX4Cryptotanshinone (CTN)Salvia miltiorrhiza Bunge (Danshen)A549, NCI-H520, and BAES-2B— —CTN can inhibit the activity of GPX4. Lung cancer[54]
GPX4Pentacyclic triterpenoids (PTs), including glycyrrhetinic acid (GA), ursolic acid (UA) and oleanolic acid (OA)PTs are natural products that can be found in various plants, such as licorice, rosemary, olive, and loquat.HCT116, HeLa, A549, L02, AT-II, and HEK293Nude mice with HCT116 tumor xenograftsGA treatment led the ferroptosis negative regulatory protein expression (GPX4, SLC40A1, and SLC7A11) decreased considerably.Colorectal cancer (CRC)[55]
GPX4Astragaloside IV (Ast-IV)Astragalus membranaceus Bunge, a traditional Chinese herb— —C57BL/6J male mice, lung injury model induced by PM2.5The Ast-IV intervention increased the expression of GPX4, which was decreased in PM2.5 group.Lung injury caused by PM2.5 exposure[56]
GPX4Wogonoside, wogonin, palmatine, paeoniforin and liquiritinShaoyao Decoction (SYD)Caco-2Sprague–Dawley male rats with TNBS-induced colitis modelSYD induced activation of GPX4 transcription and increase of protein expression.Inflammatory bowel disease (IBD), especially ulcerative colitis (UC)[57]
GPX4Polyphenols sourced from Ilex latifolia Thunb. (PIT)Ilex latifolia Thunb.Porcine intestinal epithelial cells (IPEC-J2)Weanling piglets under oxidative stress induced by diquat injectionSupplementation with PIT significantly reduced jejunal TFR1 gene expressions and improved SLC7A11 and GPX4 gene expressions in the piglets under oxidative stress.Intestinal injury caused by oxidative stress[58]
GPX4Danshensu (Dan)Dan is a pure molecule derived from the root of the Salvia miltiorrhiza herb, Danshen. LX-2 human and T6 rat hepatic stellate cells (HSCs) — —Dan up-regulated GPX4, and SLC7A11.Liver fibrosis and cirrhosis[59]
GPX4— —Naotaifang extract (NTE): a compound traditional Chinese herbal medicine composed of four herbs: Radix Astragali (Huangqi), Rhizoma chuanxiong (Chuangxiong), Pheretima (Dilong), and Bombyx batryticatus (Jiangcan)— —Sprague-Dawley (SD) rats with middle cerebral artery occlusion (MCAO) model of acute cerebral ischemiaNTE increased the expression levels of SLC7A11, GPX4.Ischemic stroke[60]
GPX4Cucurbitacin B (CuB)Trichosanthes kirilowii MaximowiczHuman nasopharyngeal carcinoma CNE1 cellsBALB/c nude mice, human nasopharyngeal carcinoma murine xenograft modelCuB induced ferroptosis by increasing lipid peroxidation and reducing the expression of GPX4 in mRNA and protein level.Nasopharyngeal carcinoma[61]
GPX4Dihydroartemisinin (DHA) and Chlorin e6 (Ce6)DHA derived from the natural plant Artemisia annua, is a sesquiterpene lactone compoundLewis cells (LLC), a lung cancer cell model— —DHA could reduce GPX4 at mRNA and protein level.Lung cancer[62]
GPX4PerillaldehydeAmmodaucus leucotrichus Coss. and Dur (A. leucotrichus), commonly known as hairy cuminHL-60, Jurkat, DLD-1, SHSY5Y— —Perillaldehyde decreased GPX4 protein expression.Acute myeloid leukemia[63]
GPX4Lycium barbarum polysaccharide (LBP)The fruits of the traditional Chinese herb L. barbarum.MCF-7, MDA-MB-231— —LBP down-regulated the protein level of xCT (SLC7A11) and GPX4.Breast cancer[64]
GPX4MatrineSophora flavescensHCT116— —Matrine down-regulated the protein level of GPX4.Colorectal cancer (CRC)[65]
GPX4Wogonoside (WG)Scutellaria baicalensis Georgi, a perennial herb of the Labiatae familyHSC-T6, AML-12, RAW264.7C57BL/6 mice, induced by CCl4 for liver fibrosis modelWG down-regulated the protein levels of GPX4 and SLC7A11.Liver fibrosis[66]
GPX4Suberitenones A and B, mycalolsHemimycale topsenti, Haliclona (Rhizoniera) dancoiHepG2, A549, A2058, MRC5— —Mycalols could reduce the level of GPX4 protein in HepG2 cells. Hepatocellular carcinoma, lung carcinoma, melanoma, anaplastic thyroid carcinoma[67]
GPX4Polyphyllin VI (PPVI)Paris polyphyllaHCCLM3, Huh7 and THLE-2Male BALB/c nude mice with subcutaneous tumor model of Huh7 cellsThe GPX4 protein expression levels in the HCCLM3 and Huh7 cells were down-regulated, and negatively associated with the concentration of PPVI.Hepatocellular carcinoma (HCC)[68]
GPX46-GingerolGinger (Zingiber officinale Roscoe)A549BALB/cNude mice, A549 tumor xenografts6-Gingerol down-regulate the expressions of GPX4 at protein level.Lung cancer[69]
GPX4Realgar (REA)— —Eca109, KYSE150— —REA IC50 and 2IC50 caused significant down-regulation of GPX4.Esophageal cancer[70]
GPX4, HO-1EriodictyolCitrus fruits and some Chinese herbal medicinesHT-22APPswe/PS1E9 transgenic mice, a mouse model of Alzheimer’s diseaseEriodictyol treatment could increase the GPX4, and HO-1 at protein level in both the cortex and hippocampus of APP/PS1 mice. Alzheimer’s disease (AD)[71]
GPX4, HO-1β-elemeneCurcumae rhizomaHCT116, LoVo, CaCO2 β-elemene in combination with cetuximab was shown to upregulate the HO-1 and down-regulate GPX4.Colorectal cancer[72]
GPX4, HO-1— —Tripterygium wilfordii Hook.f. (TwHF)— —APPswe/PS1E9 transgenic mice, a mouse model of Alzheimer’s diseaseTwHF treatment reduced the PQ up-regulated HO-1 level, and it up-regulated the expression of GPX4 compared to the PQ group.Alzheimer’s disease (AD)[73]
GPX4, HO-1FPHLP (low-polarity fraction from Ficus pandurata Hance)Ficus pandurata Hance HepG2C57BL6/J mice, CCl4-induced acute liver injury modelFPHLP significantly reduced the level of Fe2+ and expression of TfR1, xCT/SLC7A11, and Bcl2, while increasing the expression of GPX4.Acute liver injury[74]
GPX4, HO-1Biochanin A (BCA)Huangqi— —C57BL/6 miceThe tendency of HO-1, and GPX4 expression was downregulated when treated with ferric ammonium citrate and upregulated after incubation with BCA.knee osteoarthritis[75]
GPX4, HO-1, ACSL4Icariin (ICA)EpimediumH9C2— —The mRNA expressions and protein levels of Nrf2 and HO-1 were reduced notably after H/R stimulation but were elevated with the increase of ICA concentration. The expression of ACSL4 were decreased with the increase of ICA concentration, while the expression of GPX4 was increased with the increase of ICA concentration.Myocardial ischemia /reperfusion injury[76]
GPX4, HO-1NobiletinCitrus peelSK-MEL-28— —The mRNA and protein level of GPX4 and HO-1 was decreased in nobiletin-treated melanoma cells.Melanoma[77]
GPX4, HO-1— —GBH (Insamgobonhwan)HT22ICR mouseGBH can reverse the RSL3 induced GPX4 reduction and HO-1 increase.Alzheimer’s disease (AD)[78]
GPX4, HO-1Salvianolic acid B(Sal B)Salvia miltiorrhiza (Danshen in Chinese)H9C2Sprague-Dawley rats, myocardial infarction modelNrf2 was strongly activated in MI rats pretreated with Sal B, and resulted in the upregulation of its target genes including HO-1, xCT, Gpx4, Fth1, and Fpn1.Myocardial infarction (MI)[79]
GPX4, GCLCOridonin (Ori)Rabdosia rubescens (Hemsl.)TE1— —Ori can decrease the enzymatic activity of GCLC and GPX4.Esophageal cancer[80]
GPX4, NOX4TectorigeninBelamcanda chinensisPrimary renal tubular epithelial cells— —Tectorigenin treatment greatly inhibited Smad3 phosphorylation, and the transcription and protein level of Nox4; while it indirectly restored the expression of GPX4.Chronic kidney disease[81]
GPX4, ACSL4— —Epimedium koreanum Nakai— —RatEpimedium koreanum Nakai ethanol extract (EEE) downregulated GPX4, and up-regulated ACSL4 significantly.Toxicity-natural products-induced liver injury[82]
GPX4, ACSL3, ACSL4IcaritinEpimedium, a traditional Chinese medicineHK-2 cells, hOAT1-HEK293 cells, and mock-HEK293 cellsC57BL/6 mice, with icaritin-induced nephrotoxicity modelIcaritin significantly downregulated the protein expression of ACSL3, and ACSL4, and it can reduce the activity of GPX4.Icaritin-induced nephrotoxicity[83]
GPX4, ACSL4Gossypol acetic acid (GAA)The seeds of cotton plantsH9C2 cardiomyoblast cells and neonatal rat cardiomyocytesSprague-DawleyGAA significantly decreased the mRNA levels of Ptgs2 and Acsl4, decreased the protein levels of ACSL4 and NRF2, and increased the protein levels of GPX4 in I/R-induced ex vivo rat hearts. Cardiac Ischmia/Reperfusion Injury[84]
GPX4, ACSL4— —Xiaoyaosan: Radix Angelicae sinensis, Radix Paeoniae alba, Poria, Radix bupleuri, Rhizoma Atractylodis macrocephalae, Radix glycyrrhizae, Herba menthae, Rhizoma Zingiberis recens— —C57BL/6The Xiaoyaosan treatment could upregulate the stress-induced down-regulation of GPX4; and down-regulate the stress-induced up-regulation of ACSL4.Depressive-like behavior[85]
GPX4, GCLC, GCLMAstragaloside-IV (AS-IV)Astragalus membranaceusARPE-19 cells— —AS-IV could restore the down-regulated expression of GPX4, GCLM, and GCLC under high glucose conditions.Diabetic retinopathy (DR)[86]
HO-1Panaxydol (PX)Panax ginsengBEAS-2BSpecific pathogen-free (SPF) male C57BL/6 mice, murine model of LPS-induced ALI. PX increased Nrf2 and HO-1 expression in comparison with LPS group. Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) [87]
HO-1Tagitinin CTithonia diversifoliaHCT116— —Tagitinin C increased mRNA expression level of HO-1Colorectal cancer[22]
HO-1NodosinIsodon serra (Maxim.) KudoT24, UMUC3, SW780Xenograft tumor modelNodosin up-regulated the expression of genes, including HMOX1, G0S2, SQSTM1, FTL and AIFM2Muscle-invasive bladder cancer (MIBC)[50,88]
HO-1— —PropolisSU-DHL-2— —The expressions of HO-1 were up-regulated at mRNA level when treated by propolis.Diffuse large B-cell lymphoma (DLBCL)[89]
HO-1Thonningianin A (Th A)Penthorum chinense Pursh. SH-SY5YZebrafish (AB strain of wild-type zebrafish); 6-OHDA-induced zebrafish model of Parkinson’s diseaseTh A treatment significantly facilitated the Nrf2 nuclear translocation and subsequently increased the HO-1 protein level.Parkinson’s disease (PD)[90]
HO-1Icariin (ICA)Herba epimediiEndplate chondrocytes, nucleus pulposus cells, annulus fibrosus cellsC57BL/6JICA treatment led to notably increased protein levels of Nrf-2 and its downstream HO-1.Intervertebral disc degeneration[91]
HO-1Astragalus membranaceus, Astragaloside IV, Astragalus polysaccharide, Swainsonine, DaidzeinAstragalus membranaceus, a kind of traditional Chinese medicineHepG2— —miRNA level of HMOX1 can be up-regulated by Astragalus membranaceus.Hepatocellular carcinoma (HCC)[92]
HO-1, GCLC, GCLM AndrographolideAndrographis paniculata, a traditional Chinese herbal medicineMKN74 and NUGC4— —Andrographis treatment upregulated the expression of HMOX1, GCLC, and GCLMGastric cancer[93]
HO-1, NQO1Full-fat rice branA by-product of rice processing.— —Yorkshire pigs were used as experimental animals to establish a prolonged cold stimulation model.Full-fat rice bran promoted the mRNA expression of Nrf2 and NQO1, as well as the protein content of Nrf2 and HO-1.Cardiac injury and energy metabolism disturbance caused by prolonged cold stimulation.[94]
HO-1, GCLC, GCLM AndrographolideAndrographis paniculata, a traditional herbHCT116, HT29, NCM460athymic nude mice, subcutaneous xenograft modelHMOX1, GCLC, and GCLM were significantly up-regulated in the andrographis and the combination treatment group vs. the untreated group.Colorectal cancer (CRC)[95]
HO-1, GCLC, GCLM Andrographis extract (standardized to 20% andrographolide content)Andrographis paniculataHCT116 and SW480MouseAndrographis could upregulate the
HMOX1, GCLC, and GCLM, in mRNA and protein level individually or in combination with 5FU.
Colorectal cancer (CRC)[96]
ACSL4β-Elemonic acid (EA)EA is isolated from Boswellia papyrifera, a plant used in traditional medicine.CRC lines SW480, HCT116 and HT29; the control colorectal cell line NCM460Female BALB/c nude mice; subcutaneous xenograft model of CRCEA at high concentration induces ferroptosis by downregulating FTL and upregulating TF, CP, and ACSL4.Colorectal cancer (CRC)[97]
ACSL4Silibinin (SIL)Silybum marianumHepG2, HEK293T, Hep1-6— —The enzymatic assays showed SIL inhibited ACSL4 enzymatic activity, thereby mitigating the ACSL4-mediated ferroptosis. Liver diseases[98]
NQO1Quinones: Cryptotanshinone, idebenone, menaquinone 4, dopamine, lipoic acid, ascorbic acid, baicalein, 3,3′-diindolylmethane, fisetin, triptophenolide, cynaroside (luteoloside), icaritin, gossypol, carnosol, honokiol, β-lapachone, dihydroisotanshinone I (DHIT I), dihydrotanshinone I (DHT I), tanshinone I and cryptotanshinone.— —HT22, MC65— —All quinones that were substrates of these proteins as identified by the NADH decay assay displayed an increase in anti-lipid peroxidation activity in the liposomes in the presence of NQO1 or FSP1.Alzheimer’s disease (AD)[99]
Table 3. Eligibility criteria of selected articles.
Table 3. Eligibility criteria of selected articles.
No.Eligibility Criteria
1Not included article types. e.g., review, proceedings, feature, editorial material.
2Not in the life Sciences
3Irrelevant object/topic
4Full text not available
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zuo, H.-L.; Huang, H.-Y.; Lin, Y.-C.-D.; Liu, K.-M.; Lin, T.-S.; Wang, Y.-B.; Huang, H.-D. Effects of Natural Products on Enzymes Involved in Ferroptosis: Regulation and Implications. Molecules 2023, 28, 7929. https://doi.org/10.3390/molecules28237929

AMA Style

Zuo H-L, Huang H-Y, Lin Y-C-D, Liu K-M, Lin T-S, Wang Y-B, Huang H-D. Effects of Natural Products on Enzymes Involved in Ferroptosis: Regulation and Implications. Molecules. 2023; 28(23):7929. https://doi.org/10.3390/molecules28237929

Chicago/Turabian Style

Zuo, Hua-Li, Hsi-Yuan Huang, Yang-Chi-Dung Lin, Kun-Meng Liu, Ting-Syuan Lin, Yi-Bing Wang, and Hsien-Da Huang. 2023. "Effects of Natural Products on Enzymes Involved in Ferroptosis: Regulation and Implications" Molecules 28, no. 23: 7929. https://doi.org/10.3390/molecules28237929

Article Metrics

Back to TopTop