1 The First School of Clinical Medicine, Southern Medical University, 510000 Guangzhou, Guangdong, China
2 Blood Transfusion Department, Shenzhen Nanshan People’s Hospital, 518052 Shenzhen, Guangdong, China
3 Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, 421001 Hengyang, Hunan, China
4 Laboratory Medicine Centre, Shenzhen Nanshan People’s Hospital, 518052 Shenzhen, Guangdong, China
5 First Clinical Medical College of Ningxia Medical University, 750000 Yinchuan, Ningxia, China
6 Medical Laboratory Center, the Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, 518001, Shenzhen, Guangdong, China
†These authors contributed equally.
Abstract
Aldo-keto reductase 1B10 (AKR1B10) is expressed in various malignant tissues. Several studies have highlighted the essential function of AKR1B10 in lipid metabolism and in the detoxification of lipid peroxides. The aim of this research was to explore the role of AKR1B10 in the susceptibility of MDA-MB-231 cells to ferroptosis. These cells serve as a model for triple-negative breast cancer (TNBC).
Lentiviral transfection was used to establish stable cell lines with high or low expression of AKR1B10. Our model of ferroptosis used the ferroptosis activator RSL3, and the specific ferroptosis inhibitor ferrostatin-1 (Fer-1) to rescue cell death. Stable cell lines were treated with the specific inhibitor OSU-T315 directed against phosphorylation of Ser473 in protein kinase B (AKT) and Ser9 in glycogen synthase kinase 3 beta (GSK3β), either alone or in combination with RSL3. A fatty acid stress model was established using palmitic acid (PA) or arachidonic acid (AA), either in the presence or absence of serum starvation and with or without co-treatment with RSL3. Cell viability was evaluated with the cell counting kit-8 (CCK8) assay and lipid peroxidation levels by flow cytometry after staining with C11 BODIPY 581/591. Exploration of the underlying mechanisms was conducted through RNA sequencing and bioinformatics analysis. Western blotting was performed to evaluate protein levels, and quantitative real-time polymerase chain reaction (qPCR) was used to evaluate transcript levels.
Western blot and qPCR analyses validated the successful establishment of stable MDA-MB-231 cell lines with and without AKR1B10 overexpression. Cell viability and lipid reactive oxygen species (ROS) assays showed that AKR1B10 suppressed ferroptosis in the RSL3-induced cell death model. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Set Enrichment Analysis (GSEA) analyses indicated the phosphatidylinositol 3-kinase (PI3K)-AKT pathway was likely to play a role in the underlying mechanisms. AKR1B10 increased the expression of glutathione peroxidase 4 (GPX4), thus potentially implicating the AKT/GSK3β/nuclear factor erythroid 2-related factor 2 (NRF2)/GPX4 pathway in the mechanism. These changes in protein levels were also observed by Western blot analysis after 6 h of RSL3 treatment. Under the influence of RSL3, the transcript levels of NRF2-related genes including GPX4, ferritin heavy chain 1 (FTH1), heme oxygenase 1 (HO-1), and NAD(P)H quinone dehydrogenase 1 (NQO-1) were significantly elevated in the AKR1B10 overexpression cell line, whereas that of prostaglandin-endoperoxide synthase 2 (PTGS2) was significantly reduced. Similar changes were observed after treatment with OSU-T315. AKR1B10 was found to suppress the sensitivity to ferroptosis induced by treatment with OSU-T315, PA, or AA. These phenomena were rescued by the ferroptosis inhibitor Fer-1.
AKR1B10 appears to be an important mechanism protecting MDA-MB-231 cells from ferroptosis, possibly through the AKT Ser473/GSK3β Ser9/NRF2/GPX4 pathway. AKR1B10 may be a key factor underlying the therapeutic effect of RSL3 under different exogenous fatty acid microenvironments.
Keywords
- aldo-keto reductase family 1 member B10
- ferroptosis
- triple-negative breast neoplasms
- NFE2L2
Ferroptosis is a regulated form of cell death characterized by iron dependence and in which the driving mechanism is lipid peroxidation [1, 2, 3]. It is intricately associated with various biological processes, including development, aging, immune response, and cancer. Numerous metabolic pathways contribute directly or indirectly to the regulation of ferroptosis by balancing iron accumulation or lipid peroxidation with the metabolic processing of iron, lipids, and amino acids, along with certain degradation processes such as the ubiquitin-proteasome system (UPS) and macroautophagy/autophagy [4]. RSL3 is a known inducer of ferroptosis and selectively exerts a lethal effect on cells harboring oncogenic rat sarcoma (RAS) mutations. This compound targets the selenium-rich enzyme Glutathione Peroxidase 4 (GPX4) [5], which is an inhibitor of ferroptosis. The activity of GPX4 relies on glutathione (GSH) to facilitate the conversion of phospholipid hydroperoxide (PLOOH) into a harmless form [6], thereby effectively shielding cells from lipid peroxidation-induced damage.
Glycerophospholipids are crucial components of the lipid bilayer and significantly influence the occurrence of ferroptosis through the formation of oxidized phosphatidylcholine (OxPC). Among these, 1-palmitoyl-2-(5′-oxo-valeroyl)-sn-glycero-3-phosphocholine (POVPC) and 1-palmitoyl-2-(9′-oxo-nonoyl)-sn-glycero-3-phosphocholine (PONPC) cause significant cardiac toxicity depending on their concentration and exposure time. The accumulation of POVPC and PONPC markedly decreases the cellular activity of GPX4 in cells, rendering them more vulnerable to ferroptosis [7]. Aldo-keto reductase 1B10 (AKR1B10) has a detoxification function for lipid peroxides. Members of the AKR1B family effectively reduce substances such as the phospholipid aldehyde POVPC [8]. Consequently, AKR1B10 may be vital for maintaining the integrity of cell membranes and protecting cells from oxidative stress-induced damage.
AKR1B10 is absent in normal breast tissue, but expressed at significant levels in some primary and metastatic breast cancer samples, with elevated expression being strongly linked to poor prognosis [9]. In a case-control study, our research team found that the serum AKR1B10 level was markedly higher in individuals with localized or metastatic breast cancer, and was positively correlated with the expression of AKR1B10 in cancerous tissues. Furthermore, the serum AKR1B10 level fell significantly within three days after the excision of the primary tumor [10], thus reinforcing the connection between its expression and disease status in breast cancer. The triple-negative breast cancer (TNBC) phenotype has a poor prognosis and a high tendency for metastasis. The occurrence, growth, and metastasis of this aggressive breast cancer subtype are known to be affected by dysregulated lipid metabolism. In the present study, we utilized RSL3 as an inducer of ferroptosis and introduced different exogenous fatty acids to simulate various lipid microenvironments. This allowed us to explore the mechanism by which ectopic AKR1B10 expression can affect the survival of TNBC cells.
The cells used in this study were the human embryonic kidney cell line HEK293T (CL-0005, Procell, Wuhan, Hubei, China) and the human triple-negative breast cancer cell line MDA-MB-231 (CL-0150, Procell, Wuhan, Hubei, China). Both were maintained in DMEM medium (11995065, Gibco, Shanghai, China) enriched with 10% fetal bovine serum (FBS, 164210, Procell, Wuhan, Hubei, China) and containing 100 U/mL penicillin and 100 U/mL streptomycin. Penicillin/streptomycin was obtained from NCM (C100C5, NCM, Suzhou, Jiangsu, China). Cells were grown at 37 °C in an atmosphere containing 5% carbon dioxide. The cell lines used in this study underwent short tandem repeat (STR) profiling and tested negative for mycoplasma contamination.
Lentiviral technology was used to construct MDA-MB-231 cell lines that were stably transfected with vector and AKR1B10 and then screened with puromycin. The cell model for induction of ferroptosis was established using RSL3 (HY-100218A, MCE, Shanghai, China). Initial verification of the ferroptosis cell model was conducted using a 96-well plate format. MDA-MB-231 cells with high or low expression of AKR1B10 were exposed to varying concentrations (0–10 µM) of RSL3, either alone or in combination with the specific inhibitor OSU-T315 (0–5 µM), for 24 h. For the experiments involving fatty acid pretreatment, cells were first incubated for 2 h with fatty acids [300 µM palmitic acid (PA) or 50 µM arachidonic acid (AA)] dissolved in basal or complete culture medium, then treated with RSL3 (0–10 µM) for 24 h. Cell viability was assessed using a cell counting kit-8 (CCK8) assay following the manufacturer’s protocol. For the experimental protocol, vector and AKR1B10 cells were divided into the following treatment groups: RSL3 at various concentrations (0–10 µM) for 24 h, OSU-T315 (T5485, Topscience, Shanghai, China) at various concentrations (0–5 µM) for 24 h, a combination of OSU-T315 (2.5 µM or 5 µM) and RSL3 (2.5 µM) for 24 h, a combination of PA (300 µM, KC001, Xi’an Kunchuang, Xi’an, Shaanxi, China) and RSL3 (0–10 µM) for 24 h, a combination of AA (50 µM, KC014, Xi’an Kunchuang, Xi’an, Shaanxi, China) and RSL3 (0–10 µM) for 24 h, and fatty acids at various concentrations (0–600 µM PA or 0–100 µM AA) for 24 h or 48 h. For cell rescue experiments, the ferroptosis inhibitor ferrostatin-1 (Fer-1, 5 µM, S7243, Selleck, Shanghai, China) was applied 2 h prior to single or combination therapies and maintained throughout treatment.
The AKR1B10 DNA sequence was retrieved from the NCBI website (https://www.ncbi.nlm.nih.gov). The detailed sequence can be found in the Supplementary Materials. Lentiviral transduction was performed using the pCDH plasmid to establish MDA-MB-231 cell lines with or without AKR1B10 overexpression. Plasmids were obtained from Shanghai Kelei Biotechnology (Shanghai, China) and transfected using Lipofectamine 2000 reagent (11668019, Invitrogen, Shanghai, China). A mixture (4:3:1) of the target plasmid, packaging plasmid (psPAX2), and envelope plasmid (pMD2.G) was introduced into HEK293T cells to generate lentivirus and subsequently harvested to infect MDA-MB-231 cells. After 48 h of infection, 2 µg/mL puromycin (ST551, Beyotime, Shanghai, China) was added to select for stably expressing cell lines.
A total of 3000 cells were seeded (100 µL per well) in a 96-well plate and incubated overnight at 37 °C. Complete or basic culture medium containing various concentrations of drugs was added and the cells incubated for the required time. The medium was subsequently substituted with complete medium containing 10% CCK8 reagent (C6005, NCM, Suzhou, Jiangsu, China), and the absorbance was recorded at 450 nm using a microplate reader (SpectraMax® iD3, Molecular Devices, Shanghai, China). Cell viability was calculated using the following formula: cell viability (%) = (Asample – Ablank)/(Acontrol – Ablank)
MDA-MB-231 cells were cultured in 6-well plates, and stable cell lines of vector and AKR1B10 were established via lentiviral infection. Total RNA was extracted and purified from these cells using TRIzol reagent (15596018, Thermo Fisher Scientific, Shanghai, China) as recommended by the manufacturer. Oligo (dT) magnetic beads (61005, Thermo Fisher Scientific, Shanghai, China) were used to specifically isolate polyA mRNA from the samples. mRNA fragmentation occurred at elevated temperatures using a magnesium ion disruption kit (E6150S, NEB, Beijing, China), followed by cDNA synthesis using a reverse transcriptase kit (1896649, Thermo Fisher Scientific, Shanghai, China). The cDNA was subsequently converted into dsDNA through a double-stranding reaction using E. coli DNA polymerase I (m0209, NEB, Beijing, China) and RNase H (m0297, NEB, Beijing, China), during which dUTP solution (R0133, Thermo Fisher Scientific, Shanghai, China) was incorporated. End repair was then performed, and the product was subsequently purified using magnetic bead technology. A cDNA library was constructed via PCR amplification, and paired-end sequencing was carried out on the Illumina NovaseqTM 6000 platform (LC Bio Technology Co., Ltd., Hangzhou, China) using standard operating procedures.
Data analysis and visualization were performed using the online platform https://www.omicstudio.cn/home. For the analysis of differentially expressed genes (DEGs) and associated Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, datasets were screened using predefined thresholds. Gene expression levels were compared between the AKR1B10 expression group and the control vector group using the following specific screening criteria: Fold-Change (FC)
After discarding the culture medium, cells were incubated with complete medium containing 10 µM C11 581/591 BODIPY (D3861, Invitrogen, Shanghai, China) for 1 h, then rinsed with PBS (G4202, Servicebio, Wuhan, Hubei, China), digested with trypsin, and resuspended in 500 µL of PBS. To quantify lipid reactive oxygen species (ROS) levels, fluorescence intensity values in the FITC channel were assessed using flow cytometry (NAVIOS, Beckman Coulter, Brea, CA, USA). FlowJo v10.8.1 software (FlowJo LLC, Ashland, OR, USA) was employed for data analysis. The fluorescence intensity of the control group was used to set the gating threshold, ensuring that signals from negative cells were confined to the designated negative region. The fluorescence intensities of all groups were then compared to determine the proportion of positive cells. Data from replicate wells (FITC measurements) were combined and presented as the mean
Following cell lysis, the total protein content was isolated using RIPA buffer (P0013B, Beyotime, Shanghai, China) containing protease inhibitor (G2008, Servicebio, Wuhan, Hubei, China) and phosphatase inhibitor (G2007, Servicebio, Wuhan, Hubei, China) to preserve protein integrity. The extracted proteins were then resolved via sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) (P0014C, Beyotime, Shanghai, China) and transferred onto a PVDF membrane (ISEQ00010, Merck, Tipperary, Ireland) using transfer buffer (E173-01, Genstar, Beijing, China). The membrane was then blocked with a 5% non-fat milk solution (232100, BD Difco, Franklin Lakes, NJ, USA) for 2 h to prevent nonspecific binding. Following this, the membrane was incubated overnight at 4 °C with the primary antibody and then washed three times for 10 minutes each before incubation with the secondary antibody. The primary antibodies used in this study were as follows: AKR1B10 (1:1000, 18252-1-AP, Proteintech, Wuhan, Hubei, China), protein kinase B (AKT, 1:5000, ab179463, Abcam, Shanghai, China), Phospho-AKT Ser473 (1:2000, 4060T, CST, Danvers, MA, USA), glycogen synthase kinase 3 beta (GSK-3β, 1:1000, 12456T, CST, Danvers, MA, USA), Phospho-GSK-3β Ser9 (1:1000, 5558T, CST, Danvers, MA, USA), erythroid 2-related factor 2 (NRF2, 1:500, 12721T, CST, Danvers, MA, USA), Kelch-like ECH-associated protein 1 (KEAP1, 1:1000, D6B12, CST, Danvers, MA, USA), and GPX4 (1:1000, 52455T, CST, Danvers, MA, USA). The secondary antibody was goat anti-rabbit IgG conjugated to horseradish peroxidase (1:10,000, 7074, CST, Danvers, MA, USA). The PVDF membrane was immersed in the secondary antibody solution and incubated at room temperature for 1 h. After three membrane washes, the protein bands were detected using an ECL substrate (BL520B, Biosharp, Tallinn, Estonia), and the protein levels were evaluated using a chemiluminescence imaging system (OI 600 MF, BIO-OI, Guangzhou, China). ImageJ 1.53t software (National Institutes of Health, Bethesda, MD, USA) was used to quantify the signal intensity.
The relative expression level of the target protein was calculated using the following formula: relative expression of target protein = (signal intensity of target protein)/(signal intensity of internal reference protein). The relative expression level of the target protein in the experimental groups was determined by normalizing to the control group, which is defined as 1.
RNA was extracted from adherent cells using the TransZol Regular RNA Extraction Kit (ET101-01-V2, TransGen, Beijing, China) as recommended by the manufacturer. Briefly, cells were lysed with guanidine isothiocyanate (G108673, Aladdin, Shanghai, China) before adding the RNA extraction reagent. The solution was then separated into a colorless aqueous phase and a pink organic phase. RNA was present in the aqueous phase and was precipitated and recovered using isopropanol. qPCR experiments were conducted using HiScript III RT SuperMix (R323, Vazyme, Nanjing, Jiangsu, China) and PerfectStart® Green qPCR SuperMix (AQ602, TransGen, Beijing, China). The extracted RNA was reverse transcribed into cDNA and then amplified with the following human-specific primers (Shenzhen Hechengyuan Biotechnology Co., Ltd.): AKR1B10, 5′-CAGCAACAGAGAGCAGGACG-3′ and 5′-TGCCAAGAGGAGACTTCCAA-3′; GPX4, 5′-AAGATCCAACCCAAGGGCAA-3′, and 5′-AGACGGTGTCCAAACTTGGTG-3′; heme oxygenase 1 (HO-1), 5′-ACTGCGTTCCTGCTCAACAT-3′ and 5′-GGGGGCAGAATCTTGCACT-3′; NAD(P)H quinone dehydrogenase 1 (NQO-1), 5′-GCTGGTTTGAGCGAGTGTTC-3′ and 5′-CTGCCTTCTTACTCCGGAAGG-3′; ferritin heavy chain 1 (FTH1), 5′-ACTACCACCAGGACTCAGAGG-3′ and 5′-CAGTCATCACAGTCTGGTTTCTTG-3′; prostaglandin-endoperoxide synthase 2 (PTGS2), 5′-GCAAATTGCTGGCAGGGTTG-3′ and 5′-GCTCTGGTCAATGGAAGCCT-3′. The Applied Biosystems 7300 Real-Time PCR System (Thermo Fisher Scientific, Foster City, CA, USA) was used to analyze gene expression.
The Ct value of the target gene was first normalized to the Ct value of an internal reference gene (
Each experimental procedure was carried out in triplicate. Statistical analysis was conducted using GraphPad Prism 10.3.0 software (GraphPad Software Inc., La Jolla, CA, USA). Data are presented as the mean
Lentiviral technology was used to construct stable cell lines with either AKR1B10 overexpression or the control vector. Ectopic expression of AKR1B10 was confirmed at both the protein and mRNA levels (Fig. 1A,B). To explore how AKR1B10 influences the therapeutic efficacy of RSL3, we performed an in vitro assessment of the effect of the drug treatment. Overexpression of AKR1B10 caused a marked shift in the cell survival curve, indicating a potential protective effect (Fig. 1C). Furthermore, the ferroptosis inhibitor Fer-1 mitigated the cell death induced by RSL3 (Fig. 1D). Additionally, the C11 BODIPY 581/591 probe was used to label cells in order to assess the level of lipid peroxidation by flow cytometry. AKR1B10 overexpression was associated with a significant decrease (p
Fig. 1. Overexpression of aldo-keto reductase 1B10 (AKR1B10) protects MDA-MB-231 cells from ferroptosis. (A) Ectopic expression of the AKR1B10 protein in triple-negative breast cancer MDA-MB-231 cells was confirmed by Western blot analysis (n = 3). (B) Quantitative real-time polymerase chain reaction (qPCR) was used to verify the establishment of the control vector and AKR1B10 stably transfected cell lines (n = 4). (C) Cell viability was evaluated using the cell counting kit-8 (CCK8) assay following treatment with varying concentrations of the ferroptosis activator RSL3 (n = 6). (D) Cells were pretreated with the ferroptosis-specific inhibitor ferrostatin-1 (Fer-1) (5 µM) before co-incubation with RSL3 (5 µM) for 24 h. Cell viability was then assessed with the CCK8 assay (n = 6). (E,F) Following the same treatment as in (D) for 8 h, cells were labeled with the C11 BODIPY581/591 probe to measure lipid peroxidation levels using flow cytometry (n = 3). The overall levels of lipid reactive oxygen species (ROS) were presented as the mean
We next conducted RNA-seq analysis to study the mechanism underlying the effect of RSL3 treatment in the presence of AKR1B10 overexpression. This identified a set of DEGs (Fig. 2A), as shown in the heat map in Fig. 2B. KEGG pathway analysis of these DEGs showed significant enrichment of the top 20 pathways across various biological processes, including metabolism, PI3K-AKT, RAS and other signaling pathways (Fig. 2C). Further statistical evaluation of the classical pathways indicated significant enrichment in the PI3K-AKT pathway (Fig. 2D). To investigate the potential molecular mechanisms in more detail, GSEA was performed on the complete RNA-seq gene set, as well as KEGG pathway analysis. The results revealed notable differences in ferroptosis-related pathways, including PI3K-AKT, p53, proteasome, and the cell cycle (Fig. 2E,F).
Fig. 2. RNA sequencing revealed that the protective role of AKR1B10 in ferroptosis may be related to the phosphatidylinositol 3-kinase/protein kinase B (PI3K-AKT) signaling pathway. Gene expression changes caused by AKR1B10 overexpression were detected by RNA-seq technology. The results are presented as differentially expressed genes (DEGs) (A) and as a heatmap (B). (C) The DEGs identified in (A) underwent Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, with the top 20 pathways showing significant enrichment. (D) Statistical analysis of the relative signaling pathways revealed that the PI3K-AKT pathway was the most significantly enriched. (E,F) KEGG pathway analysis using Gene Set Enrichment Analysis (GSEA) revealed significant differences in ferroptosis-related pathways, including PI3K-AKT, p53, the proteasome, and the cell cycle. This figure was generated using the online platform https://www.omicstudio.cn/home.
GPX4 facilitates the reduction in lipid peroxides within the complex environment of the cellular membrane. Under normal conditions, we observed that AKR1B10 overexpression resulted in elevated GPX4 (p
Fig. 3. AKR1B10 reduces sensitivity to ferroptosis via the AKT Ser473/Glycogen Synthase Kinase 3 Beta (GSK3
To further investigate the role of the AKT Ser473/GSK3
Given the pivotal role of AKR1B10 in fatty acid metabolism, we next investigated the impact of various fatty acid environments on the therapeutic efficacy of ferroptosis in tumor cells. Previous research found that mice injected with MDA-MB-231-Luc cells with high AKR1B10 expression showed a marked increase in lung tumor burden [15]. High-fat diets are known to exacerbate cancer progression. Palmitic acid (PA), a saturated fatty acid, was previously identified as a promoter of cancer metastasis [16], while arachidonic acid (AA) is a common polyunsaturated fatty acid (PUFA), that was recently studied in relation to ferroptosis. Our earlier work showed that AKR1B10 reduces the sensitivity of MDA-MB-231 cells to RSL3-induced ferroptosis. To explore the potential of combined fatty acid and RSL3 therapy, cells were pretreated with 300 µM PA or 50 µM AA for 2 h in either complete or serum-free medium. These conditions simulate the complex fatty acid environment that tumor cells encounter during lung metastasis and in other biological contexts. The cells were then treated with RSL3 for 24 h to simulate lipid peroxide-induced stress damage. The combination of these fatty acids with RSL3 resulted in greater cytotoxicity than single-agent treatments, regardless of the presence of serum during pretreatment. Moreover, the sensitizing effect of these therapies could be rescued by Fer-1. AKR1B10 remained the determining factor for differential cell viability induced by the combined treatments (Fig. 4A–H). To assess whether the fatty acid concentrations used were cytotoxic, the cells underwent prolonged culture with either PA or AA alone. After 24 h of exposure to PA at concentrations ranging from 0 to 600 µM, the overexpression of AKR1B10 showed no significant effect on cell viability (Fig. 4I). However, after 48 h of incubation, AKR1B10 conferred protection against lipid toxicity induced by PA at a concentration of 300 µM (p
Fig. 4. AKR1B10 influences the efficacy of RSL3 therapy in the presence of different exogenous fatty acids. (A) MDA-MB-231 cells with or without overexpression of AKR1B10 were pre-incubated with 300 µM palmitic acid (PA) in complete culture medium for 2 h, followed by treatment with varying concentrations of RSL3 for 24 h. (B) MDA-MB-231 cells with or without high expression of AKR1B10 were pre-incubated with 300 µM PA in complete culture medium for 2 h, followed by 5 µM RSL3 for 24 h. Fer-1 was used to rescue from cell death. (C) MDA-MB-231 cells with or without high expression of AKR1B10 were pre-incubated with 300 µM PA in basal culture medium for 2 h, followed by treatment with varying concentrations of RSL3 in complete medium for 24 h; (D) MDA-MB-231 cells with or without high expression of AKR1B10 were pre-incubated with 300 µM PA in basal culture medium for 2 h, followed by 5 µM RSL3 in complete medium for 24 h. Fer-1 was used to rescue from cell death. (E–H) Using a treatment regimen analogous to that employed in (A–D) pre-incubation was carried out with 50 µM arachidonic acid (AA). Extended culture with varying concentrations of PA (I,J). Extended culture with varying concentrations of AA (K,L). In all experiments, the CCK8 assay was utilized to measure cell viability (n = 6). The data were analyzed by two-way ANOVA corrected by Sidak’s test. ns, not significant; * p
In summary, our results suggest that AKR1B10 can activate the NRF2-mediated antioxidant pathway via phosphorylation of the AKT/GSK3
Fig. 5. Ectopic expression of AKR1B10 enables cells to survive in ferroptosis. This figure was created using the website www.biorender.com.
This study demonstrated that AKR1B10 regulates ferroptosis in MDA-MB-231 cells via the AKT/GSK3
In the resting state, the UPS continuously degrades NRF2. Its interaction with KEAP1 facilitates this process by linking NRF2 to the ubiquitin ligase complex. However, exposure to stressors such as ROS modifies KEAP1 cysteine residues, resulting in functional inactivation. This modification stabilizes NRF2, enabling its nuclear translocation and subsequent activation of protective gene transcription. Under conditions of p62 overproduction or impaired autophagy, binding of p62 to the KEAP1-NRF2 interaction site stabilizes NRF2 and promotes the transcription of its target genes [20]. Significant accumulation of NRF2 was observed in the nucleus of ATG7-deficient (autophagy-deficient) hepatocytes. In pancreatic ductal adenocarcinoma (PDA) with a high incidence of activating KRAS mutations and TP53 inactivation, NRF2 controls cellular fate by inducing the expression of antioxidant genes expression under conditions of oxidative stress [21].
In a mouse model of breast cancer metastasis, HK2 was found to bind to the protein kinase GSK3
The serine-threonine kinase GSK3 plays a central role in a variety of cellular processes. TNBC is a more aggressive subtype of breast cancer that exhibits gene expression profiles similar to those found in cells that have undergone epithelial-mesenchymal transition (EMT). GSK3
Significant therapeutic effects may be obtained by combining exogenous fatty acids with the process of ferroptosis. Exogenous fatty acids can modulate lipid metabolism in cells, alter membrane fluidity, and enhance their sensitivity to ferroptosis, thus improving the anti-cancer efficacy. Highly saturated membrane lipids help to protect cancer cells from the harmful effects of ROS. This is because saturated lipids are less likely to undergo oxidation compared to unsaturated lipids, making them more stable and better able to prevent damage. PUFAs are highly susceptible to oxidation during ferroptosis, especially AA and adrenic acid. Oxidation leads to disruption of the lipid bilayer, thereby compromising cell membrane function. Lipid peroxidation of PUFAs is a key factor in promoting cell lysis and death, and interactions among multiple complex lipid metabolism pathways strongly impact the vulnerability of cells to ferroptosis [26].
MDA-MB-231 cells are a widely studied cell line derived from TNBC. They appear to have greater sensitivity to the lipid microenvironment than MCF7 cells, which are a Luminal type A breast cancer cell line. PA and docosahexaenoic acid (DHA) can remodel the endoplasmic reticulum (ER) membrane of MDA-MB-231 cells, thereby affecting the activity of certain resident enzymes. This difference in sensitivity to the lipid environment may account for variations in chemoresistance between breast cancer cell lines [27].
The PI3K/AKT/mTOR signaling pathway is crucial for the regulation of a wide range of cellular functions, including lipid metabolism. A study on the effects of various free fatty acids (FFAs) found that PA could reduce basal PI3K activity in MDA-MB-231 cells [28]. Furthermore, increased activity of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, leading to the generation of ROS and triggering of ferroptosis, was observed in both in vitro and in vivo models of myocardial lipotoxicity following exposure to PA and to a long-term high-fat diet (HFD) [29]. Targeting of the AKT/GSK3
AA shows varied therapeutic effects in different tumor types, including a dose-dependent effect on multiple myeloma (MM) cells. Compared to vehicle controls, high doses of AA significantly reduced the proliferation and viability of MM cells, whereas low doses had the opposite effect. The active metabolites of AA support cellular function, but AA also serves as a precursor for the generation of oxygen radicals that damage cells via peroxidation. A 50 µM dose of AA was shown to increase lipid peroxidation in MM cells [31]. Interferon gamma (IFN-
Wang et al. [33] reported that knockdown of AKR1B10 in tumor cells resulted in a reduction of more than 50% in total cellular lipids, especially phospholipids, along with an increase in intracellular lipid peroxides. It was further revealed that AKR1B10 helps to maintain acetyl-CoA carboxylase
Despite many advances in diagnosis, surgery, radiotherapy, and chemotherapy, the management of breast cancer, and in particular TNBC remains challenging and patient survival rates are still quite poor. TNBC displays a heterogeneous phenotype, particularly in terms of ferroptosis-related metabolites and metabolic pathways [35]. It is therefore crucial to identify other potential forms of cell death in order to find new biomarkers or therapeutic targets and thus overcome tumor chemoresistance. The research presented here provides better understanding of the role of AKR1B10 in TNBC and offers new perspectives and potential targets for related therapeutic strategies.
This study provides new evidence showing that AKR1B10 can protect tumor cells from ferroptosis. The underlying mechanism may be related to the AKT Ser473/GSK3
The researchers affirm that all information underpinning the findings of this investigation are accessible. The source data can be obtained from the corresponding authors upon reasonable request.
SWU, QL and DL conceived and designed the experiments. SWU, GY, YL, SWANG and JW performed the experiments. SWU, GY, XW, QL and DL analyzed the data. SWU, QL and DL drafted and revised the manuscript. All authors approved the final version of the manuscript. All authors contributed to editorial changes in the manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.
Not applicable.
The authors express their gratitude to the Medical Research Centre of Shenzhen Nanshan People’s Hospital for granting access to the experimental facilities. We also extend our thanks to Dr. Wen Ai and Mr. Yongshen He for their invaluable support.
This research received financial support from the following funding sources: General Projects of the National Natural Science Foundation of China (82273236), Guangdong Basic and Applied Basic Research Foundation (2022A1515220042), the Basic and Applied Basic Research Foundation of Guangdong Province (2023A1515030182), Shenzhen Natural Science Foundation Project (JCYJ20220530141609021), Supported by Municipal Financia 1 Subsidy of Shenzhen Medical Key Discipline Construction (SZXK054) and the Project of Technological Research and Development and Creative Design Itemized Fund (NS2024030).
The authors declare no conflict of interest.
Supplementary material associated with this article can be found, in the online version, at https://doi.org/10.31083/FBL36615.
References
Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.





