International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
The maintenance of redox homeostasis is dependent upon the levels of pro-oxidant active molecules and antioxidants (1). The disruption of this equilibrium plays a crucial role in the development and progression of numerous diseases (2). The accumulation of pro-oxidative reactive factors, including reactive oxygen species (ROS), reactive nitrogen species (RNS) and reactive lipids (RLS), leads to the oxidation of DNA, proteins and lipids, altering their structure, activity and physical properties, and resulting in oxidative stress and cellular dysfunction (3). ROS are among the most ubiquitous oxidants in cells and are capable of reacting with numerous substances to form superoxide (4). The reaction between ROS and polyunsaturated fatty acids (PUFAs) in plasma, cell membranes and organelle membranes is critical, leading to the production of lipid peroxides (5). RLS produced by lipid peroxidation, such as 4-hydroxynonenal (4-HNE) and malondialdehyde (MDA), are closely associated with the onset and progression of numerous diseases, including cancer, diabetes, neurodegenerative disorders, cardiovascular diseases and ischemia/reperfusion (I/R) injury. Redox homeostasis therefore plays a critical role in the pathogenesis of multiple diseases.
The aryl hydrocarbon receptor (AhR) is a transcription factor intricately linked to redox homeostasis in a variety of inflammatory diseases (6). AhR is activated by a diverse array of exogenous and endogenous ligands and transcriptional regulates downstream target genes. This process consequently modulates the oxidative and antioxidant balance and a variety of cellular functions. AhR target genes, including the NAD(P)H dehydrogenase [quinone] 1 (NQO1), glutathione S-transferase (GST), UDP-glucuronosyltransferase 1-1 (UGT1A1), CYP1A1 and CYP1B1 genes, play a crucial role in regulating cellular redox homeostasis (7). Moreover, AhR plays a crucial role in the metabolism of exogenous substances and drugs, iron and heme metabolism, glutathione synthesis, and lipid metabolism (8,9). The dysregulation of AhR is observed in numerous disease processes, highlighting its essential role in cell survival, particularly under conditions of redox homeostasis imbalance.
AhR plays a multifaceted role in maintaining cell survival, and both AhR agonists and inhibitors have been shown to modulate cell survival or death, particularly in the context of tumor therapy (10-14). In 2003, Dixon et al (15) identified a novel form of cell death termed ferroptosis, characterized as an iron-dependent process triggered by lipid peroxidation. This pathway was initially discovered through the high-throughput screening of anti-neoplastic drugs (15). The ferroptosis-inducing drug, erastin, induces ferroptosis through the inhibition of the cystine/glutamate antiporter system xC-/xCT, which is essential for the synthesis of reduced glutathione, ultimately leading to the accumulation of toxic lipid peroxides (16). Transcriptome analysis conducted by Kwon et al (17) on pan-cancer cell lines revealed that the activity of transcription factors, including Nuclear factor erythroid 2-related factor 2 (NFE2L2/NRF2) and AhR, serves as a critical marker of erastin resistance. AhR plays a context-dependent role in modulating ferroptosis sensitivity across tumor cell lines (18,19). For instance, the siRNA-mediated knockdown of AhR increases erastin sensitivity in erastin-insensitive A549 cells, while reducing sensitivity in erastin-sensitive Calu1 cells, highlighting its dual role in ferroptosis resistance (17). Supporting this, Kou et al (19) and Peng et al (20) demonstrated that AhR expression levels were closely associated with erastin sensitivity in both A549 and BEAS-2B (human normal bronchial epithelial cells). Their research further identified the solute carrier family 7 member 11 (SLC7A11) antioxidant system as a key downstream effector of AhR (19,20). Despite these advancements, the precise molecular mechanisms underlying the involvement of AhR in ferroptosis remain to be fully elucidated.
AhR plays a pivotal role in modulating disease progression through the regulation of lipid peroxidation and ferroptosis across diverse pathological contexts (17,18). Beyond its established functions in tumors, AhR activation has been implicated in other diseases. For example, in I/R injury (liver, myocardium and kidneys), it exacerbates tissue damage by promoting lipid peroxidation and ferroptosis (21-24); AhR-mediated lipid peroxidation and ferroptosis have been implicated in neurodegenerative disorders, such as Alzheimer's disease (25); in intestinal inflammation, AhR induces ferroptosis in intestinal intraepithelial lymphocytes (natural TCRαβ+ CD8αα+ T-cells and TCRγδ+ CD8αα+ T-cells, as well as induced TCRαβ+ CD4+ and TCRαβ+ CD8αβ+ T-cells) (26); and in asthma pathogenesis, AhR modulates ferroptosis in lung epithelial cells (BEAS-2B) (27). Notably, AhR exhibits context-dependent roles in these processes. In I/R injury, AhR activation uniformly aggravates tissue damage. By contrast, its role in tumors is more intricate, displaying dual effects that vary by cell type and ligand specificity. While the AhR-ferroptosis axis has been extensively studied in tumors and I/R injury, its implications in inflammatory bowel disease, neurodegenerative diseases and lung diseases remain underexplored, representing critical gaps in current research.
Early studies have identified several key mechanisms underlying the context-dependent roles of AhR in response to diverse ligands or cellular environments (28). The differential effects of AhR activation may be attributed to the following factors: i) Ligand-specific receptor conformation. Distinct ligands, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and the tryptophan metabolite, 6-formylindolo[3,2-b]carbazole (FICZ), induce unique conformational changes in AhR, affecting its interaction with the exenobiotic response element (XRE), the core DNA sequence regulating gene transcription in the AhR signaling pathway. ii) Ligand affinity and chemical properties: The binding stability and nuclear retention of AhR are determined by ligand characteristics (e.g., hydrophobicity, molecular size). High-affinity ligands prolong nuclear localization and sustained signaling, while low-affinity ligands (e.g., plant-derived metabolites) typically trigger transient activation. iii) Cofactor recruitment specificity: The AhR-AhR nuclear translocator (ARNT) complex recruits ligand-dependent cofactors, such as inflammation-related cofactors (e.g., NF-κB and STAT), metabolic regulators (e.g., glypican 1α and sterol regulatory element-binding protein 1), to direct the downstream responses. iv) Cell type and microenvironment dependency: Tissue-specific expression of cofactors results in the activation of different genes by the same ligand in different cells. For example, AhR may preferentially bind to IL-6 or IL-22 promoters in immune cells, while promoting CYP450 transcriptione in hepatocytes. Furthermore, ligands may remodel the chromatin accessibility of AhR, and at the same time, change the epigenetic state through metabolites. v) Species/tissues-specific AhR isoforms: AhR subtype expression (e.g., AhR1 and AhR2) induces differential responses across tissues or species.
Given that AhR target genes critically regulate redox homeostasis, its role in lipid peroxidation and ferroptosis varies by ligand context and disease setting, suggesting that targeted AhR modulation may have potential for use in various pathological conditions (Table I). Ferroptosis has emerged as a crucial pathological mechanism in diverse diseases, particularly in cancer and I/R injury, where its initiation is intricately linked to disruptions in redox homeostasis and excessive lipid peroxidation (29). This review specifically examines the therapeutic implications of targeting AhR-mediated lipid peroxidation and ferroptosis in these two clinically significant conditions. The present review focuses on the fundamental association between lipid peroxidation and ferroptosis, summarizing and critically analyzing the role and potential mechanisms of AhR in these processes. The present review aimed to enhance the understanding of AhR signaling in diseases pathogenesis and to explore novel therapeutic avenues targeting this pathway for disease intervention.
Table ISummary of AhR ligands and antagonists regulating lipid peroxidation and ferroptosis pathways. |
Lipid peroxidation is an ubiquitous phenomenon observed in a wide array of disease conditions and serves as a critical triggering factor for ferroptosis. This process typically manifests as a free radical chain reaction, characterized by an overabundance of reactive species, such as ROS, RNS and RLS, along with the excessive accumulation of labile iron pools in cells (30). The primary event in lipid peroxidation involves the generation of labile hydroperoxides (LOOH) through the interaction of ROS with membrane PUFAs. LOOH compounds are highly susceptible to decomposition, leading to the formation of metabolically toxic by-products, MDA and 4-HNE, which are closely associated with iron-induced cell death (31). Furthermore, ferrous iron plays a crucial role as an initiator of ferroptosis (32). Ferrous iron can catalyze the conversion of hydrogen peroxide into hydroxyl radicals, which subsequently react with PUFAs to produce lipid peroxides, leading to cellular membrane damage and the onset of ferroptosis.
Lipid peroxides and their degradation products are controlled by intracellular antioxidant mechanisms (33). It has been found that the defense systems against ferroptosis primarily consist of glutathione (GSH)-dependent and GSH-independent antioxidant pathways. Reduced glutathione serves as a substrate for glutathione peroxidase 4 (GPX4), which catalyzes the reduction of lipid peroxides to non-toxic alcohol forms, thereby detoxifying these harmful compounds. Ferroptosis suppressor protein 1 (FSP1) utilizes NADPH to generate reduced coenzyme Q10, thereby shielding the cell membrane from damage induced by lipid peroxides. These two independent antioxidant defense systems exert stringent control over lipid peroxidation levels. Erastin and icFSP1 are specific inhibitors of both ferroptosis defense systems and have been used in the treatment of various diseases (34,35).
The induction of lipid peroxidation-mediated ferroptosis is critically influenced by three pivotal factors: The generation of reactive species, the concentration of the intracellular labile iron pool and the modulation of the antioxidant defense system (Fig. 1). These pathways are governed by multiple factors, many of which are transcriptionally regulated by AhR. For instance, stearoyl-CoA desaturase 1 regulates the metabolism of unsaturated fatty acids and reduces the oxidative vulnerability of PUFAs, thereby promoting lipid peroxide formation (36). Heme oxygenase 1 (HMOX1) regulates heme catabolism, releasing free iron that upregulates the levels of labile iron pools within cells (37). Cyclooxygenase-2 (COX-2) facilitates ROS production via multiple metabolic pathways, inducing lipid peroxidation and ferroptosis (38). The SLC7A1/GPX4 system, a GSH-dependent antioxidant mechanism, maintains intracellular redox homeostasis by promoting the synthesis of reduced GSH (39). FSP1, an antioxidant system independent of GSH, inhibits lipid peroxidation and ferroptosis by consuming NADPH and promoting the reduction of ubiquinone Q10 (34). NFE2L2/NRF2 is a master regulator of the cellular antioxidant response, promoting the transcription of numerous downstream antioxidant-related genes through binding to their antioxidant response elements (ARE) (40). CYP1B1, a member of the cytochrome P450 enzyme family, mediates metabolic processes that generate ROS, contributing to lipid peroxidation and ferroptosis (41). Other members of the cytochrome P450 enzyme family, including CYP1A1 and CYP1A2, have also been implicated in lipid peroxidation, although to the best of our knowledge, no studies to date have demonstrated a direct link between these enzymes and ferroptosis. Notably, the key factors regulating lipid peroxidation and ferroptosis are either target genes of AhR or are regulated by AhR target genes. This suggests that AhR may serve as a crucial regulatory node for lipid peroxidation and ferroptosis. Investigating the mechanisms by which AhR modulates ferroptosis and identifying the potential targets of AhR in this process holds significant promise for the development of novel therapeutic agents and the treatment of associated diseases.
AhR orchestrates lipid peroxidation and ferroptosis through diverse molecular mechanisms, thereby critically influencing the pathogenesis of multiple diseases. The AhR-mediated regulation of lipoferroptosis includes the following: The regulation of the gene expression levels of pro-oxidant and antioxidant enzymes; the regulation of iron, heme and arachidonic acid metabolism; and the regulation of the ferroptosis defense system and synthesis of antioxidant compounds. Notably, AhR activation plays a dual role in lipid peroxidation-ferroptosis axis, with distinct regulatory effects depending on the specific ligands (42). This mechanistic plasticity, evidenced by ChIP-seq and microarray analyses across different cell lineages and developmental stage (43-46), positions AhR as both a potential therapeutic target and a challenge for precise pharmacological intervention.
AhR is a ligand-dependent transcription factor that can mediate both protective and pathological responses. Initially identified due to its involvement in the toxicity of dioxins such as TCDD, AhR predominantly resides in the cytoplasm in an inactive state, complexed with chaperone proteins, including HSP90, AIP and p23. Upon binding to TCDD, AhR translocates to the nucleus, dimerizes with ARNT, and binds to XRE in target gene enhancer regions (47). This classical AhR/ARNT pathway upregulates the expression of CYP1 family members, such as CYP1A1, CYP1A2 and CYP1B1 (48). These enzymes represent the most extensively studied target genes of the AhR/ARNT complex of the activation of the classic pathway of AhR. CYP450 enzymes, including CYP1, CYP2 and CYP3, catalyze metabolic reactions that facilitate the production of ROS (49). The upregulation of CYP1A1 and CYP1B1 promote lipid peroxidation and trigger ferroptosis, and both TCDD and benzo(a)pyrene promote lipid peroxidation via this pathway (50), while the AhR antagonist, CH223191, reverses this effect (51). Similarly, decabromodiphenyl ether (BDE-209) induces lipid peroxidation via the AhR-CYP1A1-ROS pathway (52).
Notably, AhR activation by natural ligands, such as dietary indole and sulforaphane, as well as other low-affinity AhR ligands, supports intestinal homeostasis and immune regulation. However, in chronic inflammatory bowel disease, dysregulated AhR signaling, driven by gut microbiota imbalances, exacerbates oxidative stress, lipid peroxidation and iron deficiency in intestinal epithelial lymphocytes, and this can potentially be mitigated by specific AhR inhibitors (26). Moreover, in neurological and hepatic contexts, butylphthalide mitigates traumatic brain injury-induced ferroptosis by inhibiting the AhR-CYP1B1-ROS pathway (53), while CYP1B1 drives ferroptosis in hepatocellular and cholangiocarcinoma (41). Collectively, these findings underscore the pivotal role of AhR in modulating lipid peroxidation and ferroptosis through pro-oxidant enzyme regulation.
While pro-oxidative ligands, such as TCDD and benzo(a) pyrene induce lipid peroxidation via AhR activation, emerging evidence highlights the dual role of AhR in redox regulation. Natural exogenous AhR agonists, such as extracts from artichoke (Cynara scolymus), soybean, fig tree and Houttuynia cordata, mitigate ROS production, oxidative stress and lipid peroxidation by regulation of antioxidant enzyme target genes, such as NQO1, GST and UGT1A1 genes by AhR (54-56) (Fig. 2). Furthermore, the key antioxidant stress transcription factor NRF2 serves as a direct target for AhR signaling transduction. Notably, its promoter region containing multiple XRE (57), antioxidant enzymes such as NQO1, GST and UGT1A1 are also direct target genes of NRF2. This indicates that AhR regulates the NRF2 gene and thereby promotes the transcription of antioxidant enzymes. In summary, the activation or inhibition of AhR classic signaling using appropriate AhR ligands under various disease conditions can influence lipid peroxidation and ferroptosis levels by regulating the synthesis of pro-oxidant and antioxidant enzymes.
I/R injury represents a condition wherein tissue cells, following a period of ischemia, experience reperfusion, leading to an accelerated exacerbation of tissue damage. This pathological process, observed in multiple organs and diseases, results in massive ROS production in I/R injury (58). During I/R injury, various endogenous ligands of AhR, such as the tryptophan metabolite kynurenine (Kyn), are generated to sustain AhR activation, leading to CYP450-mediated ROS overproduction and exacerbating oxidative stress (59). The restoration of blood flow and subsequent reoxygenation during reperfusion frequently result in reperfusion injury, a condition increasingly recognized as being mediated by ferroptosis (60). Previous studies have indicated that ferroptosis is more likely to occur during the reperfusion phase rather than the ischemic phase in I/R injury (61). The endogenous mechanisms responsible for scavenging ROS are compromised following I/R, leading to ineffective ROS neutralization. I/R injury is characterized by a series of intracellular events, including excessive ROS production during reperfusion, lipid peroxidation and elevated intracellular iron levels (62). Lipid peroxidation and oxidative damage induced by ROS can result in cellular damage and death, which aligns with the hallmarks of ferroptosis. Ferroptosis inhibitors, such as Fer-1 and deferoxamine (DFO), can effectively prevent or mitigate the progression of I/R injury when administered prior to or during the I/R event. DFO is applied in coronary artery bypass grafting procedures. The intravenous administration of DFO has been shown to protect the myocardium from reperfusion injury and mitigate lipid peroxidation (63).
Emerging evidence highlights AhR as a crucial regulator in I/R injury affecting the liver, kidneys and myocardium (21-23,64). During the early phase of I/R injury, oxidative stress stimulates the production of oxyindole, a compound that has been demonstrated to be an effective AhR agonist and significantly activates AhR (65). Moreover, under the oxidative stress conditions associated with I/R injury, there is an increased presence of endogenous pro-oxidative ligands for AhR (66). These multiple pro-oxidative endogenous ligands may constitute a critical component in the pathophysiology of I/R injury. In a previous study, in two distinct mouse models of liver I/R injury, both the conventional ferroptosis inhibitor DFO and the AhR antagonist CH223191 were found to be effective in reversing ferroptosis and lipid peroxidation. Moreover, CH223191 demonstrated superior efficacy compared with DFO (67), suggesting that AhR activation is closely related to I/R injury. HMOX1 and COX-2 are pivotal enzymes in heme and arachidonic acid metabolism, respectively. Transcriptomic analysis revealed that the STAT3-HMOX1/COX-2 signaling pathway was inhibited following the administration of CH223191 (67). The activation of HMOX1 facilitates the release of free iron ions from heme, consequently elevating intracellular free iron levels (68). COX-2 also catalyzes the production of ROS, leading to oxidative stress and lipid peroxidation (38). Both HMOX1 and COX-2 genes play crucial roles in the induction of ferroptosis. Previous studies have demonstrated that AhR signaling exerts direct or indirect effects on the HMOX1/COX-2 signaling pathway via multiple mechanisms (69-72), and the AhR signaling pathway is intricately associated with the metabolism of arachidonic acid and heme (73).
A previous study demonstrated that targeting ferroptosis is a promising therapeutic approach to mitigate renal tubular cell injury in acute tubular necrosis induced by I/R (74). The administration of the AhR antagonist, CH223191, or the ferroptosis inhibitor, α-col, prior to the onset of reperfusion and reoxygenation injury has the potential to prevent the production of ROS, reduce lipid peroxidation and mitigate cellular iron toxicity. The underlying mechanism may involve the transcriptional regulation of AhR through indoleamine 2,3-dioxygenase (IDO)-mediated Kyn metabolism (21,66). The inhibition of AhR signaling has been shown to mitigate reperfusion injury in a rat model of cardiac I/R (23). Arachidonate 12/15-lipoxygenase-mediated arachidonic acid metabolism significantly contributes to myocardial I/R injury (75). Therefore, arachidonic acid metabolism mediated by AhR signaling may be a potential target for alleviating myocardial I/R injury.
These studies demonstrate that AhR plays a crucial role in I/R injury across various parenchymal organs. The underlying mechanisms may be associated with the metabolism of hemin and arachidonic acid. Oxidative stress induced by I/R injury results in the activation of AhR through endogenous pro-oxidative ligands, such as Kyn and oxyindole. IDO, as a key enzyme in tryptophan metabolism, can produce many endogenous ligands of AhR. IDO also serves as the target gene of AhR. Due to this particular cyclic activation characteristic, it will easily cause the continuous activation of AhR (76). Moreover, studies have shown that IDO plays a key role in I/R injury. The pharmacological inhibition of IDO by using 1-MT (IDO inhibitor) limits the production of Kyn, inhibits the activation of AhR, and subsequently the production of ROS. This effect can also be achieved by using the AhR antagonist, CH223191 (66,77). This indicates that it is of utmost importance to reduce the production of ROS induced by arachidonic acid and heme metabolism mediated by the continuous activation of AhR through targeting this circular pathway. The overproduction of these pro-oxidant ligands during I/R injury may contribute to continuous activation of AhR signaling. This activation subsequently mediates oxidative stress and lipid peroxidation, indicating that targeting AhR signaling may be a promising therapeutic strategy for mitigating reperfusion injury.
SLC7A11 is a critical component of the cystine/glutamate antiporter, which is crucial for glutathione synthesis, and it exerts a significant negative regulatory influence on ferroptosis (78). The SLC7A11-GPX4-GSH-cysteine axis serves as the pivotal hub in the ferroptosis cascade (79). In patients with CKD, the accumulation of IS, a metabolite of tryptophan, as a result of impaired renal excretion, can result in various health-related complications (80). In a previous study, in a mouse model of CKD, IS induced ferroptosis in MC3T3-E1 cells by activating the AhR signaling pathway and mediating the inhibition of the SLC7A11-GPX4 antioxidant signaling pathway. This process inhibited the osteogenic differentiation of MC3T3-E1 cells and contributed to alveolar bone resorption in mice with CKD via ferroptosis (81). The inhibition of AhR activity can mitigate the osteotoxic effects induced by IS. However, the exact mechanisms through which AhR influences SLC7A11-GPX4 signaling to trigger ferroptosis in the context of IS activation remain elusive. A plausible explanation for this phenomenon is the interaction between AhR signaling and HIF-1α signaling. In patients with CKD, HIF-1α signaling is upregulated as a result of tissue hypoxia, which is caused by diminished erythropoietin synthesis (82). Research has demonstrated that the activation of HIF-1α signaling leads to the increased expression of SLC7A11 (83,84). AhR competes with HIF-1α for binding to ARNT, thereby competitively inhibiting the HIF-1α signaling pathway. This inhibition of SLC7A11 expression, mediated by the HIF-1α signaling pathway, may promote lipid peroxidation and ferroptosis.
In contrast to the previously described induction of ferroptosis by IS through the activation of the AhR-mediated inhibition of the SLC7A11-GPX4 antioxidant signaling pathway, the knockdown of AhR influences lipid peroxidation levels in both BEAS-2B and NSCLC cells. AhR affects the transcriptional regulation of SLC7A11. Notably, the pharmacological inhibition and genetic knockdown of AhR in BEAS-2B cells markedly increases erastin-induced ferroptosis. In PC9 cells, the promoter region of SLC7A11 harbors an AhR binding site. Furthermore, a comprehensive database search and analysis using JASPAR indicated that the promoter region of SLC7A11 contains specific XREs. The study also highlighted the role of I3P, an endogenous AhR agonist ligand and a tryptophan analog, in the development of a novel AhR receptor. The AhR-SLC7A11-GSH-GPX4 axis protects cells against ferroptosis via an AhR-dependent mechanism (19,20). These findings demonstrate that the activation of AhR signaling directly modulates the transcriptional expression of the antioxidant defense system, SLC7A11, thereby conferring resistance to ferroptosis in both BEAS-2B and NSCLC cells.
In addition to its direct influence on the transcriptional expression of SLC7A11, AhR signaling also indirectly transcriptionally regulates SLC7A11 by detecting lysosomal cysteine levels and inducing the adaptive expression of ATF4. ATF4 is crucial for cells adapting to adverse conditions of amino acid deprivation and mediates a variety of cellular phenotypic changes (85). The study by Swanda et al (86) revealed that lysosomal cysteine deficiency, sensed through the Kyn pathway of AhR, leads to adaptive ATF4 expression, which subsequently enhances SLC7A11 transcription and renders cells less susceptible to ferroptosis. The synthetic mRNA reagent, CysRx, was designed to convert cytosolic cysteine into lysosomal cysteine, thereby sensitizing tumor cells to ferroptosis by downregulating ATF4 expression. The AhR inhibitor stemregenin (SR1) was used to induce ferroptosis in cysteine-deficient cells, while the AhR activator indirubin fully rescued these cells from ferroptosis (86). Notably, this pathway regulates the transcriptional activity of ATF4 in the nucleus independently of the conventional integrated stress response and oxidative stress effects. These findings demonstrate that, apart from modulating cellular sensitivity to ferroptosis through the regulation of lysosomal cysteine levels, manipulating the AhR signaling pathway to induce ferroptosis in cancer cells is a promising therapeutic target.
Gut commensal microbiota metabolites serve as a significant source of AhR agonists (87). FICZ, Kyn, indirubin, indole and I3P are endogenous ligands of AhR. Metabolites produced by the gut microbiota can be categorized into two main groups: Immunosuppressive metabolites, including short-chain fatty acids, tryptophan metabolites and bile acid metabolites; inflammatory metabolites, such as those activating the stimulator of interferon genes pathway and lipopolysaccharides (88). Immunosuppressive metabolites derived from the gut microbiota are inclined to inhibit ferroptosis (89), whereas inflammatory metabolites facilitate ferroptosis (90). Studies have shown that the anaerobic gut bacterium Streptococcus gastro metabolizes tryptophan to produce trans-3-indoleacrylic acid (IDA), which, upon activating AhR, regulates the expression of aldehyde dehydrogenase 1 family, member A3 (ALDH1A3) by binding with a high affinity region approximately 100 base pairs upstream of the ALDH1A3 transcription start site. This regulation occurs through ARNT-mediated nuclear translocation, indicating that when AhR is activated by IDA, it is recruited to the ALDH1A3 promoter (91). ALDH1A3, an aldehyde dehydrogenase which uses retinaldehyde as its substrate, is predominantly overexpressed in tumor cells (92), potentially due to the elevated levels of redox metabolism observed in tumor cells. ALDH1A3 plays a crucial role in the production of NADPH, which serves as the primary electron donor essential for reduction reactions and activates the classical FSP1, thereby inhibiting ferroptosis (34). This indicates that the intestinal microbiota metabolite IDA can enhance FSP1-mediated synthesis of reduced coenzyme Q10 via the AhR-ALDH1A3 pathway, leading to NADPH production, ferroptosis inhibition, and ultimately promoting the progression of colorectal cancer (91).
Previously, transcriptomic analysis revealed a significant association between AhR levels and erastin sensitivity (17). Given the influence of AhR activity on the sensitivity of various tumor cell lines to erastin, and the role of erastin as a specific inhibitor of SLC7A11, it was hypothesized that the bidirectional effect of AhR signaling on SLC7A11 may be attributed to context-dependent variations in AhR function. Investigating the association between AhR and erastin across diverse tumor cell lines and precisely modulating AhR activity to influence erastin-induced ferroptosis sensitivity is an area worth further exploration. Pharmacologically regulating AhR activity in conjunction with erastin to eradicate tumor cells and delay tumor progression represents a promising therapeutic strategy.
Pharmacological modulation by inducing or inhibiting ferroptosis is a key potential approach for addressing drug-resistant cancers, ischemic organ damage, and a range of other diseases characterized by extensive lipid peroxidation (29). Previous studies have indicated that drug-resistant cancer cells, particularly those exhibiting a mesenchymal phenotype and propensity for metastasis, are highly vulnerable to ferroptosis (17,93). AhR modulates ferroptosis via multiple pathways, functioning as a promoter in specific conditions such as I/R injury and certain tumors (Fig. 3), while serving as a suppressor in bronchial epithelial cells and other tumor types (Fig. 4). This dual regulatory role of AhR in ferroptosis reflects its complex function. However, the role of AhR as either a promoter or inhibitor of ferroptosis in numerous diseases remains underexplored. A more comprehensive understanding of the regulatory mechanisms of AhR in ferroptosis could pave the way for novel therapeutic approaches and may provide profound insight into the fundamental mechanisms underlying ferroptosis.
In models of I/R injury, ferroptosis is predominantly observed during the reperfusion phase, rather than the ischemic phase (94). Consistent with these observations, AhR is reactivated during the reperfusion phase of I/R injury, leading to an increase in intracellular ROS production and lipid peroxidation (21). Following I/R, the antioxidant system is significantly impaired, leading to the inefficient scavenging of ROS. The reperfusion of ischemic tissue results in an overproduction of ROS, which mediates I/R injury. Evidence suggests that I/R injury is associated with multiple cellular events, including excessive ROS production (62), lipid peroxidation, and increased intracellular iron concentrations (95). Lipid peroxidation and the generation of ROS contribute to cellular damage and mortality (21). These phenomena align with the characteristics of iron-dependent ferroptosis, and studies in both experimental models and clinical settings have demonstrated that iron chelators can mitigate I/R injury by inhibiting the initiation and progression of ferroptosis (21,63,66).
DFO, a widely used non-toxic iron chelator, has been shown to suppress lipid peroxidation-induced ferroptosis across diverse conditions. Beyond its role in inhibiting AhR, as detailed in the preceding section, the AhR antagonist, CH223191, has proven effective in reducing I/R injury in multiple organ systems, including the liver, kidneys and myocardium, as evidenced in various animal models (21,66,77). Previous studies have demonstrated that AhR is closely related to arachidonic acid metabolism, and various AhR ligands can regulate arachidonic acid metabolism in endothelial cells, hepatocytes, and cardiomyocytes (96-98). Moreover, the COX-2 pathway, lipoxygenase (LOX) pathway and CYP450 pathway in arachidonic acid metabolism play key roles in I/R injury (99) and are potential targets for the AhR-mediated relief of I/R injury. Additionally, heme metabolism is closely related to AhR, such as in I/R injury, atherosclerosis, and liver poisoning, and AhR can affect their occurrence and development through iron and heme metabolism (72,100). As a transcriptional regulator, AhR may also modulate ferroptosis via multiple potential pathways. AhR may alleviate I/R injury through ROS production mediated by CYP450 and COX-2 arachidonic acid metabolism, iron/heme metabolism mediated by STAT3/6 and HMOX1 and Kyn metabolism mediated by AhR. Notably, some studies have demonstrated that the application of CH223191 in hepatic I/R injury is more effective than DFO in alleviating hepatocyte I/R injury and ferroptosis (21,66,67). These findings suggest the feasibility of targeting the AhR signaling pathway with specific inhibitors to ameliorate I/R injury in parenchymal organs.
Ferroptosis exerts a dual effect on tumor biology; targeting ferroptosis is a promising direct strategy for cancer cell eradication, while ferroptosis simultaneously contributes to tumor progression and immune response suppression (101). Similarly, AhR exhibits intricate roles in tumorigenesis and cancer therapy, exhibiting both pro-tumor and anti-tumor effects (102). The activation of AhR is also closely associated with tumor immunity (103), and these effects are contingent upon the specific tumor type and the involved ligand (18,91). In ferroptosis in cancer cells, AhR exhibits various effects, potentially associated with varying activation levels of the downstream transcription of AhR in distinct cell lines. For example, it was demonstrated that the activation level of CYP1A1 in the H358 lung cancer cell line was markedly higher compared with that in the Calu1 cell line. The disparity in the downstream transcriptional response of AhR to the same ligand across different tumor cell lines is likely a critical factor of the differential roles of AhR in ferroptosis within these cells (17). The mechanisms by which AhR inhibits tumor ferroptosis include the modulation of the SLC7A11/GPX4 axis and the enhancement of reduced NADPH production.
It has been shown that AhR-mediated CYP450 influences the extent of ferroptosis in neurons following traumatic brain injury (53). The stabilization of CYP1B1 protein may also play a crucial role in regulating ferroptosis in hepatocellular carcinoma and cholangiocarcinoma (41). However, to the best of our knowledge, there is currently no direct evidence demonstrating that AhR promotes ferroptosis in tumor cells via the transcriptional regulation of CYP450. Moreover, AhR activation in certain tumor cells has been hypothesized to be associated with a metastatic mesenchymal state, characterized by both metastatic and immunosuppressive properties (104). Mesenchyma-like tumor cells exhibit heightened sensitivity to ferroptosis inducers, which aligns with the dual role of ferroptosis in tumor biology. The AhR signaling pathway is intricately linked to heme and iron metabolism mediated by HMOX1. Previous research has demonstrated that tumor cells overexpressing AhR exhibit elevated basal levels of HMOX1 compared with wild-type cells (59), and a high HMOX1-mediated increase in the free iron pool level is a key mediator for the triggering of ferroptosis. A reasonable explanation for the close association between AhR and heme metabolism is that AhR signaling may indirectly mediate heme metabolism through the NRF2 signaling pathway. CYP450, EMT and heme metabolism may serve as key mechanisms underlying AhR-mediated ferroptosis in tumor cells.
The majority of drugs known to induce ferroptosis, such as sorafenib, sulfadiazine, FIN and 3-phenylquinazolinone, are systemic inhibitors of xCT, GPX4 and FSP1, which are regulated by AhR (19,81,88). Therefore, the modulation of AhR levels is anticipated to substantially enhance the efficacy of ferroptosis-inducing drugs. Both AhR gene silencing and stimulation with the exogenous ligand TCDD can induce ferroptosis and increase the sensitivity of tumor cells to erastin (17,18). Consequently, the strategic use of efficient AhR ligands, either alone or in combination with ferroptosis inducers, is a viable strategy for triggering ferroptosis in tumor cells.
The AhR agonists and inhibitors currently used in in cancer therapy include aminoflavone, ITE and SR1 (10,11,14,105). These compounds exhibit notable tumor-suppressive effects. The antitumor mechanism of ITE involves restoring tumor cell sensitivity to sorafenib. The antitumor effects of SR1 and aminoflavonone are partially mediated by ROS. These mechanisms may be facilitated through various potential pathways, such as those involving AhR-mediated CYP450, SLC7A11 and EMT. The application of these highly potent AhR ligands to modulate AhR signaling in conjunction with ferroptosis-inducing drugs is anticipated to yield enhanced antitumor efficacy.
AhR-mediated lipid peroxidation-ferroptosis plays a critical role in various diseases, particularly in tumors and I/R injury. Silencing the AhR gene in NSCLC A549 cells was previously shown to enhance the antitumor efficacy of erastin (17). Another study elucidated the underlying mechanism, revealing that the activation of AhR in A549 cells mediates the transcription of SLC7A11, a well-established target of the antitumor activity of erastin (20). It is notable that the lung cancer cell line, A549, with the silencing of the AhR gene, promotes the antitumor effect of erastin (19). Conversely, in the lung cancer cell line, Calu1, the silencing of the AhR gene inhibited the antitumor effect of erastin (17). The underlying mechanisms for these discrepancies may resemble those observed in immune cells, where AhR preferentially binds to the IL-6 or IL-22 promoter. In hepatocytes, AhR is more inclined to promote the transcription of the CYP450 enzyme system. These divergent outcomes may be attributed to variations in AhR reactivity towards target genes across different cell lines and within different states of the same cell line. Such differences may involve multiple factors, including unsaturated fatty acid anabolism, EMT, heme/iron metabolism, transcriptional regulation of oxidative antioxidant enzymes and GSH-dependent/independent ferroptosis defense mechanisms. Moreover, the application of specific AhR agonists and inhibitors, including aminoflavone, ITE, SR1 and CH223191, in conjunction with ferroptosis inducers, is anticipated to augment the antitumor efficacy against targeted tumor cells.
While AhR plays a dual role in mediating ferroptosis in cancer cells, it mainly acts as a pro-oxidant to promote ferroptosis induced by lipid peroxidation in disease models, such as I/R injury, CKD and colitis. This phenomenon may be attributed to the pro-oxidative nature of the predominant AhR ligands in these contexts, including oxyindole, IS, TCDD and dioxins. Conversely, the antioxidant AhR ligand I3P maintains redox homeostasis by activating the AhR-mediated GSH-dependent ferroptosis defense system in BEAS-2B cells (20). I3P also exhibits antioxidant properties across various disease types (106). Similarly, ILA, another antioxidant AhR agonist ligand, has also been shown to exert an inhibitory effect on ferroptosis (107). These observations suggest that AhR-mediated lipid peroxidation-ferroptosis in non-neoplastic conditions may primarily depend on the specific ligand involved. During I/R injury, oxidative stress activates the tryptophan metabolic pathway, generating numerous endogenous AhR ligands, such as Kyn and FICZ. Kyn has been shown to promote tumor progression by inhibiting ferroptosis in an AhR-dependent manner in glioblastoma multiforme and lung cancer A549 cell lines (20,108). However, Kyn also demonstrates context-dependent effects: It induces ferroptosis in natural killer cells within the gastric cancer microenvironment independently of AhR, while inhibiting ferroptosis in Hela cells also via an AhR-independent mechanism (89,109). Notably, the key enzyme IDO produced by Kyn is the target gene of AhR, indicating that AhR is a central molecule mediating oxidative stress and affecting ferroptosis pathways (76). This highlights the complex and multifaceted association between AhR and ferroptosis, requiring the consideration of multiple factors. Targeting AhR signaling with specific ligands presents a potential therapeutic strategy across various diseases. A comprehensive ligand-AhR-disease database could guide this approach.
Numerous ligands have demonstrated antitumor efficacy in cellular experiments or therapeutic effects in animal models. However, well-known AhR ligands, such as TCDD and benzo[a]pyrene may induce adverse effects. At present, a number of low-toxicity or non-toxic AhR ligands are under investigation and development, including indoles and flavonoids derived from natural plant metabolites, which are characterized by their favorable safety profiles. For instance, indole-3-carbinol exhibits both low toxicity and anti-cancer properties. As previously mentioned, a number of tryptophan derivatives/metabolites serve as endogenous AhR ligands (87). Recent findings indicate their potential in treating various inflammatory bowel diseases, although excessive levels may concurrently promote intestinal tumor progression (91).
Given the potent toxicity of traditional AhR ligands such as TCDD and the potential limitations of low-affinity natural ligands, synthesis AhR ligands represent a promising avenue. Compounds such as 6,2′,4′-trimethoxyflavone (TMF) retain AhR activation capabilities, but eliminate the chlorine atoms present in TCDD, reducing abnormal signaling. TMF also mimics structural features of endogenous ligands, such as the indole/carbazole scaffold of tryptophan metabolites, to enhance compatibility and incorporates formyl groups to facilitate metabolism and prevent long-term cellular accumulation. TMF has been shown to exert antitumor effects in liver cancer models (110). Similarly, YH439, a synthetic low-toxicity AhR ligand, demonstrated efficacy in alleviating hepatic fibrosis (111). 3,3′-diindolylmethane, a low-toxicity phytochemical from cruciferous vegetables, exhibits anti-hepatocellular carcinoma properties and can induce ferroptosis in hepatocytes (112). These findings underscore the substantial potential of both synthetic AhR ligands and natural plant-derived alternatives (113). Systematic investigations to discover or synthesize clinically applicable AhR ligands are highly promising. Furthermore, in diseases such as intestinal disorders, modulating the gut microbiota to regulate the production of endogenous AhR ligands provides an alternative therapeutic strategy worthy of attention.
In conclusion, the present review underscores the pivotal role of AhR in mediating disease onset and progression through ferroptosis across various diseases. Pharmacological inhibition of AhR signaling, particularly in I/R injury, can mitigate tissue damage. The pharmacological modulation of AhR signaling in specific tumor cell lines, either through activation or inhibition, can induce lipid peroxidation-ferroptosis directly or in combination with ferroptosis inducers, which provides a biological foundation for a promising therapeutic strategy against tumor progression (Fig. 5). Currently, research on AhR-regulated lipid peroxidation and ferroptosis in diseases is limited, with the majority of studies focusing on I/R injury in tumors or various organs. Exploring the role of AhR in lipid peroxidation and ferroptosis across different diseases and tissues, and implementing targeted regulation using natural low-toxicity or synthetic AhR ligands, holds substantial clinical potential.
Current research demonstrates that AhR target genes regulate numerous key genes and mediators involved in ferroptosis, encompassing cellular antioxidant systems, oxidative/antioxidant enzymes, heme/iron metabolism and the arachidonic acid pathway. Notably, AhR can regulate the SLC7A11-GPX4 axis, a pivotal suppressor of ferroptosis. Moreover, AhR modulates ferroptosis through the GSH-independent FSP1 pathway. Consequently, targeting AhR represents a promising therapeutic strategy for diseases driven by lipid peroxidation and ferroptosis, such as cancer and I/R injury.
Not applicable.
ZL, WL and XS designed the study. ZL completed the first draft of this manuscript. ZL, MH, SH and YZ collected the data from the literature and revised the manuscript. XS, WL and ZL designed the figures. YZ, WL and XS obtained funding. All authors have read and approved the final version of the manuscript. Data authentication is not applicable.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
Not applicable.
The present study was supported by the high-level talents scientific research start-up funds of the Affliated Hospital of Guangdong Medical University (grant no. GCC2022023) and the Zhanjiang Science and Technology Project (grant no. 2023A20709).
|
Ferreira MJ, Rodrigues TA, Pedrosa AG, Gales L, Salvador A, Francisco T and Azevedo JE: The mammalian peroxisomal membrane is permeable to both GSH and GSSG-Implications for intraperoxisomal redox homeostasis. Redox Biol. 63:1027642023. View Article : Google Scholar | |
|
Fujiki Y, Abe Y, Imoto Y, Tanaka AJ, Okumoto K, Honsho M, Tamura S, Miyata N, Yamashita T, Chung WK and Kuroiwa T: Recent insights into peroxisome biogenesis and associated diseases. J Cell Sci. 133:jcs2369432020. View Article : Google Scholar : PubMed/NCBI | |
|
Agmon E, Solon J, Bassereau P and Stockwell BR: Modeling the effects of lipid peroxidation during ferroptosis on membrane properties. Sci Rep. 8:51552018. View Article : Google Scholar : PubMed/NCBI | |
|
Jambunathan N: Determination and detection of reactive oxygen species (ROS), lipid peroxidation, and electrolyte leakage in plants. Methods Mol Biol. 639:292–298. 2010.PubMed/NCBI | |
|
Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS and Stockwell BR: Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc Natl Acad Sci USA. 113:E4966–E4975. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Grishanova AY and Perepechaeva ML: Aryl hydrocarbon receptor in oxidative stress as a double agent and its biological and therapeutic significance. Int J Mol Sci. 23:67192022. View Article : Google Scholar : PubMed/NCBI | |
|
Sondermann NC, Faßbender S, Hartung F, Hätälä AM, Rolfes KM, Vogel CFA and Haarmann-Stemmann T: Functions of the aryl hydrocarbon receptor (AHR) beyond the canonical AHR/ARNT signaling pathway. Biochem Pharmacol. 208:1153712023. View Article : Google Scholar : | |
|
Hu DG, Marri S, McKinnon RA, Mackenzie PI and Meech R: Deregulation of the genes that are involved in drug absorption, distribution, metabolism, and excretion in hepatocellular carcinoma. J Pharmacol Exp Ther. 368:363–381. 2019. View Article : Google Scholar | |
|
Martins M, Santos JM, Diniz MS, Ferreira AM, Costa MH and Costa PM: Effects of carcinogenic versus non-carcinogenic AHR-active PAHs and their mixtures: Lessons from ecological relevance. Environ Res. 138:101–111. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Brantley E, Callero MA, Berardi DE, Campbell P, Rowland L, Zylstra D, Amis L, Yee M, Simian M, Todaro L, et al: AhR ligand Aminoflavone inhibits α6-integrin expression and breast cancer sphere-initiating capacity. Cancer Lett. 376:53–61. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
McLean L, Soto U, Agama K, Francis J, Jimenez R, Sowers L and Brantley E: Aminoflavone induces oxidative DNA damage and reactive oxidative species-mediated apoptosis in breast cancer cells. Int J Cancer. 122:1665–1674. 2008. View Article : Google Scholar | |
|
Elson DJ, Nguyen BD, Korjeff NA, Wilferd SF, Sanvicens VP, Jang HS, Bernales S, Chakravarty S, Belmar S, Ureta G, et al: Suppression of Ah Receptor (AhR) increases the aggressiveness of TNBC cells and 11-Cl-BBQ-activated AhR inhibits their growth. Biochem Pharmacol. 215:1157062023. View Article : Google Scholar : PubMed/NCBI | |
|
Kober C, Roewe J, Schmees N, Roese L, Roehn U, Bader B, Stoeckigt D, Prinz F, Gorjánácz M, Roider HG, et al: Targeting the aryl hydrocarbon receptor (AhR) with BAY 2416964: A selective small molecule inhibitor for cancer immunotherapy. J Immunother Cancer. 11:e0074952023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang X, He B, Chen E, Lu J, Wang J, Cao H and Li L: The aryl hydrocarbon receptor ligand ITE inhibits cell proliferation and migration and enhances sensitivity to drug-resistance in hepatocellular carcinoma. J Cell Physiol. 236:178–192. 2021. View Article : Google Scholar | |
|
Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al: Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell. 149:1060–1072. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Wang L, Liu Y, Du T, Yang H, Lei L, Guo M, Ding HF, Zhang J, Wang H, Chen X and Yan C: ATF3 promotes erastin-induced ferroptosis by suppressing system Xc. Cell Death Differ. 27:662–675. 2020. View Article : Google Scholar | |
|
Kwon OS, Kwon EJ, Kong HJ, Choi JY, Kim YJ, Lee EW, Kim W, Lee H and Cha HJ: Systematic identification of a nuclear receptor-enriched predictive signature for erastin-induced ferroptosis. Redox Biol. 37:1017192020. View Article : Google Scholar : PubMed/NCBI | |
|
Chen X, Yao Y, Gong G, He T, Ma C and Yu J: The potential role of AhR/NR4A1 in androgen-dependent prostate cancer: Focus on TCDD-induced ferroptosis. Biochem Cell Biol. 103:1–11. 2025. View Article : Google Scholar | |
|
Kou Z, Tran F, Colon T, Shteynfeld Y, Noh S, Chen F, Choi BH and Dai W: AhR signaling modulates ferroptosis by regulating SLC7A11 expression. Toxicol Appl Pharmacol. 486:1169362024. View Article : Google Scholar : PubMed/NCBI | |
|
Peng Y, Ouyang L, Zhou Y, Lai W, Chen Y, Wang Z, Yan B, Zhang Z, Zhou Y, Peng X, et al: AhR promotes the development of non-small cell lung cancer by inducing SLC7A11-dependent antioxidant function. J Cancer. 14:821–834. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Eleftheriadis T, Pissas G, Filippidis G, Liakopoulos V and Stefanidis I: Reoxygenation induces reactive oxygen species production and ferroptosis in renal tubular epithelial ; cells by activating aryl hydrocarbon receptor. Mol Med Rep. 23:412021. | |
|
Kwon JI, Heo H, Chae YJ, Min J, Lee DW, Kim ST, Choi MY, Sung YS, Kim KW, Choi Y, et al: Is aryl hydrocarbon receptor antagonism after ischemia effective in alleviating acute hepatic ischemia-reperfusion injury in rats? Heliyon. 9:e155962023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang J, Wang B, Yuan S, Xue K, Zhang J and Xu A: Blocking the Aryl hydrocarbon receptor alleviates myocardial ischemia/reperfusion injury in rats. Curr Med Sci. 42:966–973. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Ma C, Han L, Zhao W, Chen F, Huang R, Pang CH, Zhu Z and Pan G: Targeting AhR suppresses hepatocyte ferroptosis in MASH by regulating the Pten/Akt/β catenin axis. Biochem Pharmacol. 232:1167112025. View Article : Google Scholar | |
|
Qian C, Yang C, Lu M, Bao J, Shen H, Deng B, Li S, Li W, Zhang M and Cao C: Activating AhR alleviates cognitive deficits of Alzheimer's disease model mice by upregulating endogenous Aβ catabolic enzyme Neprilysin. Theranostics. 11:8797–8812. 2021. View Article : Google Scholar : | |
|
Panda SK, Peng V, Sudan R, Ulezko Antonova A, Di Luccia B, Ohara TE, Fachi JL, Grajales-Reyes GE, Jaeger N, Trsan T, et al: Repression of the aryl-hydrocarbon receptor prevents oxidative stress and ferroptosis of intestinal intraepithelial lymphocytes. Immunity. 56:797–812.e4. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Yu H, Zhang C, Pan H, Gao X, Wang X, Xiao W, Yan S, Gao Y, Fu J and Zhou Y: Indeno[1,2,3-cd]pyrene enhances the sensitivity of airway epithelial cells to ferroptosis and aggravates asthma. Chemosphere. 363:1428852024. View Article : Google Scholar : PubMed/NCBI | |
|
Avilla MN, Malecki KMC, Hahn ME, Wilson RH and Bradfield CA: The Ah receptor: Adaptive metabolism, ligand diversity, and the xenokine model. Chem Res Toxicol. 33:860–879. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Yan HF, Zou T, Tuo QZ, Xu S, Li H, Belaidi AA and Lei P: Ferroptosis: Mechanisms and links with diseases. Signal Transduct Target Ther. 6:492021. View Article : Google Scholar : PubMed/NCBI | |
|
Rochette L, Dogon G, Rigal E, Zeller M, Cottin Y and Vergely C: Lipid peroxidation and iron metabolism: Two corner stones in the homeostasis control of ferroptosis. Int J Mol Sci. 24:4492022. View Article : Google Scholar | |
|
Ramana KV, Srivastava S and Singhal SS: Lipid peroxidation products in human health and disease 2016. Oxid Med Cell Longev. 2017:21632852017. View Article : Google Scholar : PubMed/NCBI | |
|
Wu Y, Ran L, Yang Y, Gao X, Peng M, Liu S, Sun L, Wan J, Wang Y, Yang K, et al: Deferasirox alleviates DSS-induced ulcerative colitis in mice by inhibiting ferroptosis and improving intestinal microbiota. Life Sci. 314:1213122023. View Article : Google Scholar | |
|
Liu J, Kang R and Tang D: Signaling pathways and defense mechanisms of ferroptosis. FEBS J. 289:7038–7050. 2022. View Article : Google Scholar | |
|
Nakamura T, Hipp C, Santos Dias Mourão A, Borggräfe J, Aldrovandi M, Henkelmann B, Wanninger J, Mishima E, Lytton E, Emler D, et al: Phase separation of FSP1 promotes ferroptosis. Nature. 619:371–377. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Tan H, Daniels JD, Zandkarimi F, Liu H, Brown LM, Uchida K, O'Connor OA and Stockwell BR: Imidazole ketone erastin induces ferroptosis and slows tumor growth in a mouse lymphoma model. Cell Chem Biol. 26:623–633.e9. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Xuan Y, Wang H, Yung MM, Chen F, Chan WS, Chan YS, Tsui SK, Ngan HY, Chan KK and Chan DW: SCD1/FADS2 fatty acid desaturases equipoise lipid metabolic activity and redox-driven ferroptosis in ascites-derived ovarian cancer cells. Theranostics. 12:3534–3552. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Wu D, Hu Q, Wang Y, Jin M, Tao Z and Wan J: Identification of HMOX1 as a critical ferroptosis-related gene in atherosclerosis. Front Cardiovasc Med. 9:8336422022. View Article : Google Scholar : PubMed/NCBI | |
|
Adham AN, Abdelfatah S, Naqishbandi AM, Mahmoud N and Efferth T: Cytotoxicity of apigenin toward multiple myeloma cell lines and suppression of iNOS and COX-2 expression in STAT1-transfected HEK293 cells. Phytomedicine. 80:1533712021. View Article : Google Scholar | |
|
He F, Zhang P, Liu J, Wang R, Kaufman RJ, Yaden BC and Karin M: ATF4 suppresses hepatocarcinogenesis by inducing SLC7A11 (xCT) to block stress-related ferroptosis. J Hepatol. 79:362–377. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Dodson M, Castro-Portuguez R and Zhang DD: NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biol. 23:1011072019. View Article : Google Scholar : PubMed/NCBI | |
|
Liu C, Zhang C, Wu H, Zhao Z, Wang Z, Zhang X, Yang J, Yu W, Lian Z, Gao M and Zhou L: The AKR1C1-CYP1B1-cAMP signaling axis controls tumorigenicity and ferroptosis susceptibility of extrahepatic cholangiocarcinoma. Cell Death Differ. 32:506–520. 2025. View Article : Google Scholar | |
|
Apetoh L, Quintana FJ, Pot C, Joller N, Xiao S, Kumar D, Burns EJ, Sherr DH and Weiner HL: The aryl hydrocarbon receptor interacts with c-Maf to promote the differentiation of type 1 regulatory T cells induced by IL-27. Nat Immunol. 11:854–861. 2011. View Article : Google Scholar | |
|
Adachi J, Mori Y, Matsui S and Matsuda T: Comparison of gene expression patterns between 2,3,7,8-tetrachlorodibenzo-p-dioxin and a natural arylhydrocarbon receptor ligand, indirubin. Toxicol Sci. 80:161–169. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Murray IA, Morales JL, Flaveny CA, DiNatale BC, Chiaro C, Gowdahalli K, Amin S and Perdew GH: Evidence for ligand-mediated selective modulation of Aryl hydrocarbon receptor activity. Mol Pharmacol. 77:247–254. 2010. View Article : Google Scholar : | |
|
Lo R and Matthews J: High-resolution genome-wide mapping of AHR and ARNT binding sites by ChIP-Seq. Toxicol Sci. 130:349–361. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Hao N, Lee KL, Furness SGB, Bosdotter C, Poellinger L and Whitelaw ML: Xenobiotics and loss of cell adhesion drive distinct transcriptional outcomes by aryl hydrocarbon receptor signaling. Mol Pharmacol. 82:1082–1093. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Chen G and Shin JA: AhR/Arnt:XRE interaction: Turning false negatives into true positives in the modified yeast one-hybrid assay. Anal Biochem. 382:101–106. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Vogel CFA, Van Winkle LS, Esser C and Haarmann-Stemmann T: The aryl hydrocarbon receptor as a target of environmental stressors-implications for pollution mediated stress and inflammatory responses. Redox Biol. 34:1015302020. View Article : Google Scholar | |
|
Lim HJ, Jang WB, Rethineswaran VK, Choi J, Lee EJ, Park S, Jeong Y, Ha JS, Yun J, Choi YJ, et al: StemRegenin-1 attenuates endothelial progenitor cell senescence by regulating the AhR pathway-mediated CYP1A1 and ROS generation. Cells. 12:20052023. View Article : Google Scholar : | |
|
Shertzer HG, Clay CD, Genter MB, Chames MC, Schneider SN, Oakley GG, Nebert DW and Dalton TP: Uncoupling-mediated generation of reactive oxygen by halogenated aromatic hydrocarbons in mouse liver microsomes. Free Radic Biol Med. 36:618–631. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Mi P, Li N, Ai K, Li L and Yuan D: AhR-mediated lipid peroxidation contributes to TCDD-induced cardiac defects in zebrafish. Chemosphere. 317:1379422023. View Article : Google Scholar : PubMed/NCBI | |
|
Yuan J, Sun X, Che S, Zhang L, Ruan Z, Li X and Yang J: AhR-mediated CYP1A1 and ROS overexpression are involved in hepatotoxicity of decabromodiphenyl ether (BDE-209). Toxicol Lett. 352:26–33. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Yan L, Gu L, Lv X, Ni Z, Qian W, Chen Z, Yang S, Zhuge Q, Yuan L and Ni H: Butylphthalide mitigates traumatic brain injury by activating anti-ferroptotic AHR-CYP1B1 pathway. J Ethnopharmacol. 337:1187582025. View Article : Google Scholar | |
|
Nakahara T, Mitoma C, Hashimoto-Hachiya A, Takahara M, Tsuji G, Uchi H, Yan X, Hachisuka J, Chiba T, Esaki H, et al: Antioxidant opuntia ficus-indica extract activates AHR-NRF2 signaling and upregulates filaggrin and loricrin expression in human keratinocytes. J Med Food. 18:1143–1149. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Takei K, Mitoma C, Hashimoto-Hachiya A, Uchi H, Takahara M, Tsuji G, Kido-Nakahara M, Nakahara T and Furue M: Antioxidant soybean tar Glyteer rescues T-helper-mediated downregulation of filaggrin expression via aryl hydrocarbon receptor. J Dermatol. 42:171–180. 2015. View Article : Google Scholar : | |
|
Takei K, Hashimoto-Hachiya A, Takahara M, Tsuji G, Nakahara T and Furue M: Cynaropicrin attenuates UVB-induced oxidative stress via the AhR-Nrf2-Nqo1 pathway. Toxicol Lett. 234:74–80. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Miao W, Hu L, Scrivens PJ and Batist G: Transcriptional regulation of NF-E2 p45-related factor (NRF2) expression by the aryl hydrocarbon receptor-xenobiotic response element signaling pathway: Direct cross-talk between phase I and II drug-metabolizing enzymes. J Biol Chem. 280:20340–20348. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang L, Yin X, Chen YH, Chen Y, Jiang W, Zheng H, Huang FQ, Liu B, Zhou W, Qi LW and Li J: Proteomic analysis reveals ginsenoside Rb1 attenuates myocardial ischemia/reperfusion injury through inhibiting ROS production from mitochondrial complex I. Theranostics. 11:1703–1720. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Kou Z, Tran F and Dai W: Heavy metals, oxidative stress, and the role of AhR signaling. Toxicol Appl Pharmacol. 482:1167692024. View Article : Google Scholar : | |
|
Li X, Ma N, Xu J, Zhang Y, Yang P, Su X, Xing Y, An N, Yang F, Zhang G, et al: Targeting ferroptosis: pathological mechanism and treatment of ischemia-reperfusion injury. Oxid Med Cell Longev. 2021:15879222021. View Article : Google Scholar : PubMed/NCBI | |
|
Wang X, Shen T, Lian J, Deng K, Qu C, Li E, Li G, Ren Y, Wang Z, Jiang Z, et al: Resveratrol reduces ROS-induced ferroptosis by activating SIRT3 and compensating the GSH/GPX4 pathway. Mol Med. 29:1372023. View Article : Google Scholar : PubMed/NCBI | |
|
Yan HF, Tuo QZ, Yin QZ and Lei P: The pathological role of ferroptosis in ischemia/reperfusion-related injury. Zool Res. 41:220–230. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Paraskevaidis IA, Iliodromitis EK, Vlahakos D, Tsiapras DP, Nikolaidis A, Marathias A, Michalis A and Kremastinos DT: Deferoxamine infusion during coronary artery bypass grafting ameliorates lipid peroxidation and protects the myocardium against reperfusion injury: Immediate and long-term significance. Eur Heart J. 26:263–270. 2005. View Article : Google Scholar | |
|
Wang B and Xu A: Aryl hydrocarbon receptor pathway participates in myocardial ischemia reperfusion injury by regulating mitochondrial apoptosis. Med Hypotheses. 123:2–5. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Kou Z, Yang R, Lee E, Cuddapah S, Choi BH and Dai W: Oxidative stress modulates expression of immune checkpoint genes via activation of AhR signaling. Toxicol Appl Pharmacol. 457:1163142022. View Article : Google Scholar : PubMed/NCBI | |
|
Eleftheriadis T, Pissas G, Golfinopoulos S, Liakopoulos V and Stefanidis I: Role of indoleamine 2,3-dioxygenase in ischemia-reperfusion injury of renal tubular epithelial cells. Mol Med Rep. 23:4722021. View Article : Google Scholar : PubMed/NCBI | |
|
Han L, Ma C, Wu Z, Xu H, Li H and Pan G: AhR-STAT3-HO-1/COX-2 signalling pathway may restrict ferroptosis and improve hMSC accumulation and efficacy in mouse liver. Br J Pharmacol. 181:125–141. 2024. View Article : Google Scholar | |
|
Zheng C, Zhang B, Li Y, Liu K, Wei W, Liang S, Guo H, Ma K, Liu Y, Wang J and Liu L: Donafenib and GSK-J4 synergistically induce ferroptosis in liver cancer by upregulating HMOX1 expression. Adv Sci (Weinh). 10:e22067982023. View Article : Google Scholar : PubMed/NCBI | |
|
Wong PS, Li W, Vogel CF and Matsumura F: Characterization of MCF mammary epithelial cells overexpressing the Arylhydrocarbon receptor (AhR). BMC Cancer. 9:2342009. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu P, Zhou K, Lu S, Bai Y, Qi R and Zhang S: Modulation of aryl hydrocarbon receptor inhibits esophageal squamous cell carcinoma progression by repressing COX2/PGE2/STAT3 axis. J Cell Commun Signal. 14:175–192. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Lo R and Matthews J: The aryl hydrocarbon receptor and estrogen receptor alpha differentially modulate nuclear factor erythroid-2-related factor 2 transactivation in MCF-7 breast cancer cells. Toxicol Appl Pharmacol. 270:139–148. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Fader KA, Nault R, Kirby MP, Markous G, Matthews J and Zacharewski TR: Convergence of hepcidin deficiency, systemic iron overloading, heme accumulation, and REV-ERBα/β activation in aryl hydrocarbon receptor-elicited hepatotoxicity. Toxicol Appl Pharmacol. 321:1–17. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Davies R, Clothier B, Robinson SW, Edwards RE, Greaves P, Luo J, Gant TW, Chernova T and Smith AG: Essential role of the AH receptor in the dysfunction of heme metabolism induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin. Chem Res Toxicol. 21:330–340. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao Z, Wu J, Xu H, Zhou C, Han B, Zhu H, Hu Z, Ma Z, Ming Z, Yao Y, et al: XJB-5-131 inhibited ferroptosis in tubular epithelial cells after ischemia-reperfusion injury. Cell Death Dis. 11:6292020. View Article : Google Scholar : PubMed/NCBI | |
|
Cai W, Liu L, Shi X, Liu Y, Wang J, Fang X, Chen Z, Ai D, Zhu Y and Zhang X: Alox15/15-HpETE aggravates myocardial ischemia-reperfusion injury by promoting cardiomyocyte ferroptosis. Circulation. 147:1444–1460. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhan J, Chen Y, Liu Y, Chen Y, Li Z, Li X, He Z, Meng F, Qian X, Yang L and Yang Q: IDO1-mediated AhR activation up-regulates pentose phosphate pathway via NRF2 to inhibit ferroptosis in lung cancer. Biochem Pharmacol. 236:1169132025. View Article : Google Scholar : PubMed/NCBI | |
|
Xu J, Sun X, Qin F, Wang X, Chen Q and Yan R: Protective effects of salvianolic acid B on intestinal ischemia/reperfusion injury in rats by regulating the AhR/IL-22/STAT6 axis. J Recept Signal Transduct Res. 43:73–82. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Shen L, Zhang J, Zheng Z, Yang F, Liu S, Wu Y, Chen Y, Xu T, Mao S, Yan Y, et al: PHGDH inhibits ferroptosis and promotes malignant progression by upregulating SLC7A11 in bladder cancer. Int J Biol Sci. 18:5459–5474. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Conrad M and Pratt DA: The chemical basis of ferroptosis. Nat Chem Biol. 15:1137–1147. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Lin CJ, Liu HL, Pan CF, Chuang CK, Jayakumar T, Wang TJ, Chen HH and Wu CJ: Indoxyl sulfate predicts cardiovascular disease and renal function deterioration in advanced chronic kidney disease. Arch Med Res. 43:451–456. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Chen H, Zhou Y, Liu Y, Zhou W, Xu L, Shang D, Ni J and Song Z: Indoxyl sulfate exacerbates alveolar bone loss in chronic kidney disease through ferroptosis. Oral Dis. 31:264–277. 2025. View Article : Google Scholar | |
|
Curran CS and Kopp JB: The complexity of nicotinamide adenine dinucleotide (NAD), hypoxic, and aryl hydrocarbon receptor cell signaling in chronic kidney disease. J Transl Med. 21:7062023. View Article : Google Scholar : PubMed/NCBI | |
|
Yuan S, Wei C, Liu G, Zhang L, Li J, Li L, Cai S and Fang L: Sorafenib attenuates liver fibrosis by triggering hepatic stellate cell ferroptosis via HIF-1α/SLC7A11 pathway. Cell Prolif. 55:e131582022. View Article : Google Scholar | |
|
Fan Y, Ma L, Fang X, Du S, Mauck J, Loor JJ, Sun X, Jia H, Xu C and Xu Q: Role of hypoxia-inducible-factor-1α (HIF-1α) in ferroptosis of adipose tissue during ketosis. J Dairy Sci. 107:10611–10627. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Jin HO, Hong SE, Kim JY, Jang SK and Park IC: Amino acid deprivation induces AKT activation by inducing GCN2/ATF4/REDD1 axis. Cell Death Dis. 12:11272021. View Article : Google Scholar : PubMed/NCBI | |
|
Swanda RV, Ji Q, Wu X, Yan J, Dong L, Mao Y, Uematsu S, Dong Y and Qian SB: Lysosomal cystine governs ferroptosis sensitivity in cancer via cysteine stress response. Mol Cell. 83:3347–3359.e9. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao C, Bao L, Qiu M, Feng L, Chen L, Liu Z, Duan S, Zhao Y, Wu K, Zhang N, et al: Dietary tryptophan-mediated Aryl hydrocarbon receptor activation by the gut microbiota alleviates escherichia coli-induced endometritis in mice. Microbiol Spectr. 10:e00811222022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang J, Zhu N, Su X, Gao Y and Yang R: Gut-microbiota-derived metabolites maintain gut and systemic immune homeostasis. Cells. 12:7932023. View Article : Google Scholar : PubMed/NCBI | |
|
Fiore A, Zeitler L, Russier M, Groß A, Hiller MK, Parker JL, Stier L, Köcher T, Newstead S and Murray PJ: Kynurenine importation by SLC7A11 propagates anti-ferroptotic signaling. Mol Cell. 82:920–932.e7. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Cui Y, Zhang Z, Zhou X, Zhao Z, Zhao R, Xu X, Kong X, Ren J, Yao X, Wen Q, et al: Microglia and macrophage exhibit attenuated inflammatory response and ferroptosis resistance after RSL3 stimulation via increasing Nrf2 expression. J Neuroinflammation. 18:2492021. View Article : Google Scholar : PubMed/NCBI | |
|
Cui W, Guo M, Liu D, Xiao P, Yang C, Huang H, Liang C, Yang Y, Fu X, Zhang Y, et al: Gut microbial metabolite facilitates colorectal cancer development via ferroptosis inhibition. Nat Cell Biol. 26:124–137. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Marcato P, Dean CA, Liu RZ, Coyle KM, Bydoun M, Wallace M, Clements D, Turner C, Mathenge EG, Gujar SA, et al: Aldehyde dehydrogenase 1A3 influences breast cancer progression via differential retinoic acid signaling. Mol Oncol. 9:17–31. 2015. View Article : Google Scholar | |
|
Schwab A, Rao Z, Zhang J, Gollowitzer A, Siebenkäs K, Bindel N, D'Avanzo E, Van Roey R, Hajjaj Y, Özel E, et al: Zeb1 mediates EMT/plasticity-associated ferroptosis sensitivity in cancer cells by regulating lipogenic enzyme expression and phospholipid composition. Nat Cell Biol. 26:1470–1481. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Tang LJ, Luo XJ, Tu H, Chen H, Xiong XM, Li NS and Peng J: Ferroptosis occurs in phase of reperfusion but not ischemia in rat heart following ischemia or ischemia/reperfusion. Naunyn Schmiedebergs Arch Pharmacol. 394:401–410. 2021. View Article : Google Scholar | |
|
Scindia Y, Leeds J and Swaminathan S: Iron homeostasis in healthy kidney and its role in acute kidney injury. Semin Nephrol. 39:76–84. 2019. View Article : Google Scholar | |
|
Doskey CM, Fader KA, Nault R, Lydic T, Matthews J, Potter D, Sharratt B, Williams K and Zacharewski T: 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) alters hepatic polyunsaturated fatty acid metabolism and eicosanoid biosynthesis in female Sprague-Dawley rats. Toxicol Appl Pharmacol. 398:1150342020. View Article : Google Scholar : PubMed/NCBI | |
|
Aboutabl ME, Zordoky BNM and El-Kadi AOS: 3-Methylcholanthrene and benzo(a)pyrene modulate cardiac cytochrome P450 gene expression and arachidonic acid metabolism in male Sprague Dawley rats. Br J Pharmacol. 158:1808–1819. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Sroczyńska K, Totoń-Żurańska J, Czepiel J, Zając-Grabiec A, Jurczyszyn A, Wołkow P, Librowski T and Gdula-Argasińska J: Therapeutic role of eicosapentaenoic and arachidonic acid in benzo(a) pyrene-induced toxicity in HUVEC endothelial cells. Life Sci. 293:1203452022. View Article : Google Scholar | |
|
Zhang C, He M, Ni L, He K, Su K, Deng Y, Li Y and Xia H: The role of arachidonic acid metabolism in myocardial ischemia-reperfusion injury. Cell Biochem Biophys. 78:255–265. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Bock KW and Köhle C: Contributions of the Ah receptor to bilirubin homeostasis and its antioxidative and atheroprotective functions. Biol Chem. 391:645–653. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Kim R, Hashimoto A, Markosyan N, Tyurin VA, Tyurina YY, Kar G, Fu S, Sehgal M, Garcia-Gerique L, Kossenkov A, et al: Ferroptosis of tumor neutrophils causes immune suppression in cancer. Nature. 612:338–346. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Paris A, Tardif N, Galibert MD and Corre S: AhR and cancer: From gene profiling to targeted therapy. Int J Mol Sci. 22:7522021. View Article : Google Scholar : PubMed/NCBI | |
|
Hezaveh K, Shinde RS, Klötgen A, Halaby MJ, Lamorte S, Ciudad MT, Quevedo R, Neufeld L, Liu ZQ, Jin R, et al: Tryptophan-derived microbial metabolites activate the aryl hydrocarbon receptor in tumor-associated macrophages to suppress anti-tumor immunity. Immunity. 55:324–340.e8. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Duan Z, Li Y and Li L: Promoting epithelial-to-mesenchymal transition by D-kynurenine via activating aryl hydrocarbon receptor. Mol Cell Biochem. 448:165–173. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Li HM, Li X, Xia R, Zhang X, Jin TZ and Zhang HS: PHGDH knockdown increases sensitivity to SR1, an aryl hydrocarbon receptor antagonist, in colorectal cancer by activating the autophagy pathway. FEBS J. 291:1780–1794. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zeitler L, Fiore A, Meyer C, Russier M, Zanella G, Suppmann S, Gargaro M, Sidhu SS, Seshagiri S, Ohnmacht C, et al: Anti-ferroptotic mechanism of IL4i1-mediated amino acid metabolism. Elife. 10:e648062021. View Article : Google Scholar : PubMed/NCBI | |
|
Lian J, Lin H, Zhong Z, Song Y, Shao X, Zhou J, Xu L, Sun Z, Yang Y, Chi J, et al: Indole-3-lactic acid inhibits doxorubicin-induced ferroptosis through activating Aryl hydrocarbon receptor/Nrf2 signalling pathway. J Cell Mol Med. 29:e703582025. View Article : Google Scholar : PubMed/NCBI | |
|
Tian Q, Dan G, Wang X, Zhu J, Chen C, Tang D, Wang Z, Chen D, Lei S, Yang C, et al: IDO1 inhibits ferroptosis by regulating FTO-mediated m6A methylation and SLC7A11 mRNA stability during glioblastoma progression. Cell Death Discov. 11:222025. View Article : Google Scholar : PubMed/NCBI | |
|
Cui JX, Xu XH, He T, Liu JJ, Xie TY, Tian W and Liu JY: L-kynurenine induces NK cell loss in gastric cancer microenvironment via promoting ferroptosis. J Exp Clin Cancer Res. 42:522023. View Article : Google Scholar : PubMed/NCBI | |
|
Yu L, Shang D, Lang L, Liu Y, Yu M, Song D, Jia S, Han S, Li C, Liu J, et al: TMF, a natural dihydroflavonoid isolated from Scutellaria javanica Jungh, stimulates anticancer activity of s180 cancer-bearing mice, induces apoptosis, inhibits invasion and migration on HepG-2 cells. J Ethnopharmacol. 263:1130722020. View Article : Google Scholar | |
|
Liu S: Aryl hydrocarbon receptor alleviates hepatic fibrosis by inducing hepatic stellate cell ferroptosis. J Cell Mol Med. 28:e702782024. View Article : Google Scholar : PubMed/NCBI | |
|
Guo L, Zhang J and Li Y, Gao Y, Huang J, Liu M, Li J, Chai W and Li Y: 3,3′-Diindolylmethane induces ferroptosis and inhibits proliferation in non-small-cell lung cancer through the AHR/NRF2/GPX4 axis. Discov Oncol. 16:3442025. View Article : Google Scholar | |
|
Prasanth MI, Malar DS, Verma K, Prasansuklab A and Tencomnao T: Hibiscus sabdariffa calyx extract protects human keratinocyte cells from fluoranthene-induced ferroptosis via the repression of aryl hydrocarbon receptor. Ecotoxicol Environ Saf. 291:1178712025. View Article : Google Scholar |