
ABCG8‑mediated sterol efflux increases cancer cell progression via the LRP6/Wnt/β‑catenin signaling pathway in radiotherapy‑resistant MDA‑MB‑231 triple‑negative breast cancer cells
- Authors:
- Published online on: March 20, 2025 https://doi.org/10.3892/ijmm.2025.5521
- Article Number: 80
-
Copyright: © Ko et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Breast cancer is the leading cause of cancer death for women worldwide. In addition, the incidence rates of breast cancer have markedly increased over the past few years. The incident cases of BC increased from 876,990 in 1990 to 2,002,350 in 2019, and the estimated annual percentage change for incidence increased by an average of 0.33% per year worldwide (1). The cure rate is high when the cancer is diagnosed early and treated comprehensively with surgical resection, radiation and chemotherapy. However, an increase in mortality has been reported due to treatment failure caused by cancer recurrence after an apparent cure or metastasis to distant sites from the original tumor. Furthermore, 25-30% of all patients with breast cancer experience disease recurrence, which often leads to mortality (2). Expression of receptors such as estrogen receptor (ER), progesterone receptor (PR) and HER2 is vital for the evaluation of treatments. Triple-negative breast cancer (TNBC) is characterized by the absence of ER, PR and HER2, and has the worst prognosis among aggressive breast cancer subtypes due to the lack of treatment targets (3). General anticancer therapy and radiation therapy are used after surgery for patients with TNBC if there is no metastasis when TNBC is diagnosed. However, if treatment resistance is acquired, anticancer drugs and radiation do not exhibit therapeutic efficacy, ultimately increasing tumor recurrence and metastasis (4,5). Due to the acquisition of therapy resistance, patients with TNBC have a short survival period and a mortality rate of 40% within 5 years from diagnosis (6). Therefore, innovative treatment strategies for patients with TNBC with acquired treatment resistance are urgently needed.
In our previous study, radiotherapy-resistant (RT-R)-TNBC cells (RT-R-MDA-MB-231 cells) were established and their characteristics compared with MDA-MB-231 cells were clarified (7). RT-R-MDA-MB-231 cells exhibited increased migration, adhesion to endothelial cells (ECs), and invasion through ECs-Matrigel-coated membranes by upregulating adhesion molecules (such as intercellular adhesion molecule 1 and vascular cell adhesion molecule 1) and epithelial-mesenchymal transition (EMT)-associated proteins (such as MMP-9, Snail-1 and β-catenin) (7). Furthermore, RT-R-MDA-MB-231 cells may contribute to tumor progression by enhancing premetastatic niche formation through the hypoxia-inducible factor 1α (HIF-1α)-lysyl oxidase axis (8). ATP-binding cassette (ABC) transporters, the largest family of transmembrane proteins, are divided into seven subfamilies (ABCA-ABCG). The role of ABC transporters in multidrug resistance is well recognized because they are responsible for ATP-dependent export of various xenobiotics, including drugs (9,10). However, the drug efflux-independent roles of ABC transporters in cancer biology are not well known. ABC transporters are involved in the export of lipids and metabolic products across plasma and intracellular membranes (11). These substances can act as signaling substances for cells and surrounding other cancer cells. For example, ABCC4 can export prostaglandins; ABCC1 can export leukotriene C4, sphingosine-1-phosphate and lysophosphatidyl inositol; ABCB5 can export IL1β; and ABCG2 can export androgens (12). ABCB1/2/5, ABCC1/2/3/4/5/6/10 and ABCG2 are known to be directly related to resistance to chemotherapy by mediating the efflux of chemotherapy agents (13,14). However, ABCG5 and ABC subfamily G member 8 (ABCG8) are known to be partially involved in the outflow of sterols and there is little research on their function in cancer by mediating sterol efflux (15,16). Therefore, the present study aimed to identify the differences in gene expression levels of ABC transporters between RT-R-MDA-MB-231 cells and MDA-MB-231 cells. The present study also aimed to investigate the role of the ABC transporter subtype that shows the most significant change between these two cell lines.
Materials and methods
Cell culture and treatment
The MCF10A breast epithelial cell line was obtained from American Type Culture Collection, and the MCF7 (non-TNBC), and MDA-MB-231 and MDA-MB-453 (TNBC) human breast cancer cell lines were obtained from the Korean Cell Line Bank; Korean Cell Line Research Foundation. RT-R breast cancer cells (RT-R-MCF7 and RT-R-MDA-MB-231 cells) were generated by repeatedly applying 2 Gy X-ray irradiation until a final dose of 50 Gy (2 Gy; 25 times) was achieved, as described previously (7). MCF10A cells were cultured in DMEM/F-12 (cat. no. 11320-033; Gibco; Thermo Fisher Scientific, Inc.) supplemented with 5% horse serum (cat. no. 16050122; Gibco; Thermo Fisher Scientific, Inc.), 100 U/ml penicillin, 100 g/ml streptomycin, 0.5 g/ml hydrocortisone, 10 g/ml insulin, 20 ng/ml epidermal growth factor, 2 mM L-glutamine and 0.5 μg/ml amphotericin B. All cancer cell lines were cultured in RPMI 1640 medium (cat. no. SH30027.01; Cytiva) supplemented with 10% fetal bovine serum (cat. no. 160000-044; Gibco; Thermo Fisher Scientific, Inc.), 1% penicillin and streptomycin (cat. no. SV30010; Cytiva). The cells were incubated at 37°C in a humidified atmosphere containing 5% CO2. The RT-R breast cancer cells were used through five passages. MCF10A, MCF7, RT-R-MCF7, MDA-MB-231, RT-R-MDA-MB-231 and MDA-MB-453 cells were used for the detection of ABC transporter subtypes (ABCA9, ABCB4, ABCG5 and ABCG8) by western blot analysis. In other assays, MDA-MB-231 and RT-R-MDA-MB-231 cells were used. RT-R-MDA-MB-231 and MDA-MB-231 cells were treated with digoxin (cat. no. D6003; Supelco; Merck KGaA; dissolved in DMSO) at a dose of 400 nM for 24 h at 37°C or cholesterol (C4951; Sigma-Aldrich; Merck KGaA; dissolved in sterile water) at various concentrations (0, 10 and 20 μM) for 24 h at 37°C for the indicated experiments.
Cell viability assay
MDA-MB-231 and RT-R-MDA-MB-231 cells were seeded at a density of 104 cells/well in 24-well plates. The cells were treated with different concentrations (0.01, 0.05, 0.1, 0.5 and 1 μM) of doxorubicin (DOXO; cat. no. D1515; Sigma-Aldrich; Merck KGaA; dissolved in sterile water) or paclitaxel (PTX; cat. no. T7402; Sigma-Aldrich; Merck KGaA; dissolved in DMSO) for 24-72 h at 37°C. Cell viability was assessed using the D-Plus™ CCK cell viability assay kit (cat. no. CCK-3000; Donginbiotech Co., Ltd.). After treatment, 20 μl CCK-8 solution was added to 200 μl serum-free medium in each well, and the cells were incubated for 30 min at 37°C in the dark. The absorbance at 450 nm was measured using a microplate reader (Molecular Devices VersaMax; Molecular Devices, LLC).
Gene expression array analysis
Gene expression profiling was performed using the QuantiSeq 3′ mRNA-Seq Service (ebiogen, Inc.). Briefly, total RNA was extracted from MDA-MB-231 and RT-R-MDA-MB-231 cells using TRIzol® reagent (cat. no. 15596-026; Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. The quality of the RNA was evaluated using the Agilent 2100 Bioanalyzer System (Agilent Technologies, Inc.). Libraries were prepared from the total RNA (500 ng) using the Lexogen Quant-Seq 96 prep kit (cat. no. 015.2X96; Lexogen GmbH) according to the provided instructions. The library concentration was measured using an Agilent 2100 Bioanalyzer (Agilent Technologies, Inc.) and a Qubit™ Flex Fluorometer (Invitrogen; Thermo Fisher Scientific, Inc.), and the library concentration per sample was ~4 nM, which was diluted to a concentration of 20 pM. The final library concentration was adjusted to 1.5 pM after quantitative PCR-based quantification (StepOnePlusTM; Applied Biosystems; Thermo Fisher Scientific, Inc.) before sequencing. Reverse transcription was performed using an oligo-dT primer containing an Illumina-compatible sequence. The reaction conditions were 85°C for 3 min (primer hybridization) and 42°C for 15 min (cDNA synthesis). The Lexogen Quant-Seq 96 prep kit (cat. no. 015.2X96; Lexogen GmbH) contained reverse transcriptase and master mix, including buffer, dNTPs and oligo-dT primer. The RNA template was then degraded, and second strand synthesis was initiated using a random primer with an Illumina-compatible linker sequence at its 5′ end by incubation at 98°C for 1 min and 25°C for 30 min. Subsequently, the double-stranded cDNA library was amplified as follows: Initial denaturation at 98°C for 30 sec, 12 cycles of 98°C for 10 sec, 65°C for 20 sec and 72°C for 30 sec, and a final extension at 72°C for 1 min. This procedure was also performed using the Lexogen Quant-Seq 96 prep kit. Purified libraries were sequenced using an Illumina NextSeq500 system with Single-End 75 bp (cat. no. 20024906; Illumina, Inc.). The raw sequencing data underwent quality control checks using FastQC (version 0.11.5; https://www.bioinformatics.babraham.ac.uk/projects/fastqc/). Data mining was carried out using ExDEGA (v5.2.1; ebiogen, Inc.). The corresponding datasets have been submitted to a public database (Gene Expression Omnibus; www.ncbi.nlm.nih.gov/geo). ABC transporters were selected from the dataset (GSE287883) and analyzed further.
Gene silencing with small interfering (si)RNA
MDA-MB-231 cells and RT-R-MDA-MB-231 cells were transfected with 100 nM negative control siRNA (siCTRL; cat. no. SN-1003; Bioneer Corporation), ABCG8 siRNA (siABCG8; cat. no. 64241-1; Bioneer Corporation) and LDL receptor (LDLR)-related protein 6 (LRP6) siRNA (siLRP6; cat. nos. 4040-1; Bioneer Corporation) using Lipofectamine® 3000 (cat. no. L3000015; Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. The cells were incubated in complete medium containing transfection reagent for 24 h at 36°C, and the medium was then replaced with fresh medium and cells were used for experiments after 24 h of incubation. The siRNA sequences were as follows: siCTRL forward, 5′-CCUACGCCACCAAUUUCGU-3′ and reverse, 5′-ACGAAAUUGGUGGCGUAGG-3′; siABCG8 forward, 5′-CGUCAGAUUUCCAACGACU-3′ and reverse, 5′-AGUCGUUGGAAAUCUGACG-3′; siLRP6 1 forward, 5′-CUGCUUUGGAUUUUGAUGU-3′ and reverse, 5′-ACAUCAAAAUCCAAAGCAG-3′; aiLRP6 2 forward, 5′-UGAGAACACCUAUUCUACA-3′ and reverse, 5′-UGUAGAGGUGUUCUCA-3′; and siLRP6 3 forward, 5′-UGUUGACCAGUUAUCAGUA-3′ and reverse, 5′-UACUGAUAACUGGUCAACA-3′. The silencing efficiency was determined by western blot analysis.
Western blot analysis
MDA-MB-231 cells and RT-R-MDA-MB-231 cells were harvested, and lysed in RIPA buffer containing 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1% NP-40, 0.1% SDS, 0.5% sodium deoxycholate and protease inhibitors. The samples were centrifuged at 16,000 × g for 20 min at 4°C and the supernatants were collected. The protein concentration was determined using the Bradford method. Equal amounts of protein (20-50 μg/lane) were subjected to 8-10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis and transferred onto Hybond-P+ polyvinylidene fluoride membranes (Amersham; Cytiva). The membranes were blocked with 5% non-fat milk in Tris-buffered saline containing 0.05% Tween-20 for 1 h at room temperature, and then incubated with the following primary antibodies overnight at 4°C: Anti-ABCA9 (cat. no. PA5-75606; 1:1,000; Invitrogen; Thermo Fisher Scientific, Inc.), anti-ABCB4 (cat. no. PA5-78692; 1:1,000; Invitrogen; Thermo Fisher Scientific, Inc.), anti-ABCG5 (cat. no. bs-5013R; 1:1,000; BIOSS), anti-ABCG8 (cat. no. ab223056; 1:1,000; Abcam), anti-GAPDH (cat. no. MA5-15738; 1:5,000; Invitrogen; Thermo Fisher Scientific, Inc.), anti-Cyclin D1 (cat. no. ab16663; 1:1,000; Abcam), anti-Cyclin B1 (cat. no. ab32053; 1:1,000; Abcam), anti-OCT4 (cat. no. ab18976; 1:1,000; Abcam), anti-β-catenin (cat. no. sc-7963; 1:1,000; Santa Cruz Biotechnology, Inc.), anti-AKT (cat. no. sc-8312; 1:1,000; Santa Cruz Biotechnology, Inc.), anti-GSK3β (cat. no. sc-9166; 1:1,000; Santa Cruz Biotechnology, Inc.), anti-p-GSK3β (cat. no. sc-11358; 1:1,000; Santa Cruz Biotechnology, Inc.), anti-p-AKT (cat. no. 9271; 1:1,000; Cell Signaling Technology, Inc.), anti-fatty acid synthase (FASN; cat. no. 3180; 1:1,000; Cell Signaling Technology, Inc.), anti-STAT3 (cat. no. 4904; 1:1,000; Cell Signaling Technology, Inc.), anti-p-STAT3 (cat. no. 9131; 1:1,000; Cell Signaling Technology, Inc.), anti-N-cadherin (cat. no. 4061; 1:1,000; Cell Signaling Technology, Inc.), anti-HIF-1α (cat. no. ab179483; 1:1,000; Abcam), anti-inducible degrader of LDL (IDOL; cat. no. ab74562; 1:1,000; Abcam), anti-LDLR (cat. no. ab52818; 1:1,000; Abcam), anti-LRP6 (cat. no. 3395; 1:1,000; Cell Signaling Technology, Inc.), anti-phosphorylated-(p-)LRP6 (cat. no. 2568; 1:1,000; Cell Signaling Technology, Inc.), anti-liver X receptor (LXR) α/β (cat. no. sc-377260; 1:1,000; Santa Cruz Biotechnology, Inc.), anti-Polo-like kinase 1 (PLK1; cat. no. 4513; 1:1,000; Cell Signaling Technology, Inc.) and anti-sterol regulatory element binding protein 1 (SREBP1; cat. no. sc-13551; 1:1,000; Santa Cruz Biotechnology, Inc.) (precursor form 125 kDa, mature form 68 kDa). The expression of the proteins was detected using horseradish peroxidase-conjugated secondary antibodies for 1 h at room temperature, including anti-rabbit IgG (cat. no. 12-348; 1:5,000; Sigma-Aldrich; Merck KGaA) and anti-mouse IgG (cat. no. A90-116P; 1:5,000; BETHYL; Fortis Life Sciences, LLC), and western ECL substrates (cat. no. 170-5061; Bio-Rad Laboratories, Inc.) for western blotting detection. The protein levels were normalized to β-actin (cat. no. MA5-15739; 1:5,000; Thermo Fisher Scientific, Inc.). The density of protein was assessed using the ChemiDoc™ XRS+ system (Bio-Rad Laboratories, Inc.), and relative protein levels were semi-quantified using ImageJ software (version 1.53e; National Institutes of Health).
Total cholesterol quantification
For quantification of intracellular total cholesterol, 1×106 cells (MDA-MB-231 and RT-R-MDA-MB-231 cells) were collected in 100 μl PBS (Ph 7.2-7.4). Cells were frozen for a few seconds using liquid nitrogen, and thawed at room temperature, which was repeated five times to release intracellular components. Subsequently, the supernatant was collected after centrifuging for 20 min at 4°C at 400-800 × g. For quantification of total cholesterol in the cell media, cell culture media were collected into aseptic tubes and centrifuged for 1 min at 4°C at 400-800 × g, and the supernatant was transferred into new tubes. Total cholesterol was quantified using the Human Total Cholesterol ELISA Kit (cat. no. ab287836; Abcam) according to the manufacturer's instructions. The absorbance at 450 nm was measured with a microplate reader (Molecular Devices VersaMax; Molecular Devices, LLC).
Cell cycle analysis
MDA-MB-231 or RT-R-MDA-MB-231 cells, which were transfected with siCTRL or siABCG8, were fixed with 75% ethanol at 4°C overnight, and stained with PI solution (50 μg/ml PI; cat. no. 537059; Sigma-Aldrich; Merck KGaA; 0.7 μg/ml RNase A; cat. no. R6513; Sigma-Aldrich; Merck KGaA) at room temperature for 30 min in the dark. DNA content was analyzed using flow cytometry (FACSverse; BD Biosciences) and the data were analyzed using FlowJo software (version 10; Tree Star, Inc.).
Statistical analysis
All data were statistically analyzed using GraphPad Prism 8 software (Dotmatics). Statistical comparisons were performed using an unpaired Student's t-test for two-group comparisons or one-way ANOVA followed by Tukey's post hoc test for comparisons of three or more groups. The results of the cell viability assay were compared using two-way ANOVA followed by Sidak's post hoc test. The data are presented as the mean ± SD of three to five independent experiments. P<0.05 was considered to indicate a statistically significant difference.
Results
ABCG8 expression is increased in TNBC cells compared with in non-TNBC cells, and is significantly increased in RT-R-TNBC cells compared with in TNBC cells
Gene expression array analysis of MDA-MB-231 and RT-R-MDA-MB-231 cells revealed that ABCA9, ABCB4 and ABCG8 were upregulated by >2-fold in RT-R-MDA-MB-231 cells (RT-R-TNBC) compared with in MDA-MB-231 cells (TNBC) (Fig. 1A). The protein levels of ABCA9, ABCB4 and ABCG8 in MCF10A (normal epithelial cells), MCF7 (non-TNBC), RT-R-MCF-7, MDA-MB-231 (TNBC) and RT-R-MDA-MB-231 cells were subsequently determined by western blot analysis. Because ABCG5 can form a heterodimer with ABCG8, and this heterodimer of ABCG5/G8 serves as a major sterol transporter in liver and intestinal cholesterol efflux (16), the expression levels of ABCG5 were also detected. The results showed that ABCG5 and ABCG8 protein levels were higher in MDA-MB-231 cells than in MCF7 cells. Furthermore, the expression levels of ABCG8, which were upregulated in MDA-MB-231 cells, were significantly increased in RT-R-MDA-MB-231 cells (Fig. 1B and F), although the increase of ABCG8 expression at the protein level in RT-R-MDA-MB-231 cells (1.6-fold compared with MDA-MB-231 cells) was slightly lower than that at the RNA level (~2.88-fold compared with MDA-MB-231 cells), which may be due to methodological differences. In addition, it was confirmed that ABCG8 protein expression was increased in other TNBC cell lines, such as MDA-MB-453 cells compared with in MCF7 cells, a non-TNBC cell line (Fig. 1G and H). However, ABCG5 protein expression did not significantly differ between MDA-MB-231 and RT-R-MDA-MB-231 cells. Therefore, in subsequent experiments, the role of ABCG8 in cancer progression was examined in RT-R-MDA-MB-231 cells.
ABCG8 knockdown enhances chemotherapy-mediated cytotoxicity in TNBC and RT-R-TNBC cells
ABC transporters are known to mediate chemotherapy resistance by pumping out chemotherapeutic drugs (14,17). Therefore, the present study examined the effect of ABCG8 knockdown on chemotherapy-mediated cytotoxicity in TNBC and RT-R-TNBC cells. After confirming the effectiveness of ABCG8 knockdown using siABCG8 by western blot analysis (Fig. 2A and B), the effect of ABCG8 knockdown on the viability of MDA-MB-231 and RT-R-MDA-MB-231 cells treated with Doxo or PTX (0.01, 0.05, 0.1, 0.5 and 1 μM) for 24-72 h was determined (Table SI). As shown in Table SI and Fig. 2C and D, Doxo and PTX reduced the viability of MDA-MB-231 and RT-R-MDA-MB-231 cells in a dose-dependent manner over 24-72 h, and the reduction in cell viability caused by Doxo or PTX was even greater when cells were transfected with siABCG8. Specifically, for 1 μM Doxo treatment, knockdown of ABCG8 significantly decreased cell viability from 86, 67 and 51% to 65, 57 and 43% at 24, 48 and 72 h, respectively, in MDA-MB-231 cells, and from 96, 67 and 46% to 72, 61 and 36% at 24, 48 and 72 h, respectively, in RT-R-MDA-MB-231 cells (Fig. 2C). For 1 μM PTX, siABCG8 transfection led to a significant reduction from 60 to 46% at 72 h in MDA-MB-231 cells, and from 70 and 67% to 54 and 43% at 48 and 72 h in RT-R-MDA-MB-231 cells (Fig. 2D). Notably, both MDA-MB-231 and RT-R-MDA-MB-231 cells transfected with siABCG8 exhibited substantial reductions in viability after 48 and 72 h of incubation (Fig. 2E). These results indicated that the presence of ABCG8 affected cell viability, and thus, ABCG8 may have additional functions beyond its role as a drug pump.
ABCG8 knockdown is associated with accumulation of intracellular cholesterol but reduces the expression levels of β-catenin and EMT proteins
The ABCG5/ABCG8 protein is a major sterol transporter, and tumor cells need lipids for their rapid proliferation (18). The present study examined the cholesterol production in RT-R-TNBC and TNBC cells. Both intracellular and medium cholesterol levels were significantly higher in the RT-R-MDA-MB-231 group compared with those in the MDA-MB-231 group (Fig. 3A). Subsequently, to investigate the role of ABCG8 in the cholesterol efflux from the cytosol into the media, total cholesterol levels in the cell lysates and culture media were measured. The level of total cholesterol in the lysate of RT-R-MDA-MB-231 cells was higher compared with that in MDA-MB-231 cells, and this increase was further enhanced by siABCG8 transfection. By contrast, the increased cholesterol levels in the culture media from RT-R-MDA-MB-231 cells, compared with those from MDA-MB-213 cells, were significantly decreased by siABCG8 transfection (Fig. 3B). These findings suggested that siABCG8 transfection decreased cholesterol efflux from the cytosol into the media. To investigate whether ABCG8 may be involved in regulating EMT-related proteins by expelling cholesterol, the levels of EMT-related proteins, β-catenin, N-cadherin and OCT4, were measured in MDA-MB-231 and RT-R-MDA-MB-231 cells transfected with siABCG8. As shown in Fig. 3C and D, β-catenin, N-cadherin and OCT4 were more highly expressed in RT-R-MDA-MB-213 cells than in MDA-MB-231 cells. However, their expression levels were significantly reduced by siABCG8 transfection. These results suggested that cholesterol pumped out of cells by ABCG8, rather than cholesterol inside cells, may affect the induction of EMT-related proteins. However, the levels of induced p-AKT, p-STAT3 and p-GSK3β were not influenced by siABCG8 transfection (Fig. 3E and F), suggesting that ABCG8-mediated cholesterol efflux did not affect the induction of EMT-related proteins through the p-AKT, p-STAT3 or p-GSK3β pathways.
ABCG8 knockdown induces cell cycle arrest at the G2/M phase in RT-R-MDA-MB-231 cells by inhibiting PLK1 and Cyclin B1
Dysregulation of the cell cycle is a hallmark of cancer that enables limitless cell division (19). The Wnt/β-catenin pathway can modulate the cell cycle. Conversely, the cell cycle also influences the Wnt/β-catenin pathway (20). Therefore, the present study subsequently determined the role of ABCG8 in the cell cycle. Flow cytometry data showed that there were a higher proportion of RT-R-MDA-MB-231 cells in the S and G2/M phases compared with MDA-MB-231 cells (Fig. 4A). Furthermore, when ABCG8 was knocked down, a 20% increase in cells in the S phase and a 17% increase in cells in the G2/M phase in MDA-MB-231 cells, and a 20% increase in cells in the S phase and a 40% increase in cells in the G2/M phase in RT-R-MDA-MB-231 cells were observed (Fig. 4B). This result suggested that ABCG8 knockdown affected the G2/M phase. However, the changes observed in the G2/M phase resulting from ABCG8 knockdown were not significant in the MDA-MB-231 cells. The increase in the G2/M cell population due to ABCG8 knockdown was more pronounced in RT-R-MDA-MB-231 cells than in MDA-MB-231 cells. PLKs serve an essential role as key mitotic kinases and cell cycle regulators, and they also serve key roles in proliferation and cell growth (21). Among the PLKs, PLK1 has been shown to be upregulated in various types of cancer and to be associated with poor patient prognosis (22). PLK1 is a central actor during mitosis (23), which promotes entry into mitosis in the cell cycle by activating Cyclin B1 (24). Furthermore, β-catenin can regulate transcriptional induction of cell cycle regulators such as PLKs, particularly PLK1 (25,26). Therefore, the present study subsequently determined the effect of ABCG8 knockdown on PLK1 and Cyclin B1 expression. RT-R-MDA-MB-231 cells exhibited significantly increased protein expression levels of PLK1 and Cyclin B1 compared with those in MDA-MB-231 cells, which was suppressed by siABCG8 transfection. However, Cyclin D1 protein expression was significantly increased in RT-R-MDA-MB-231 cells, but was not affected by aiABCG8 transfection (Fig. 4C and D). These results indicated that ABCG8 knockdown led to downregulation of PLK1 and Cyclin B1, which resulted in cell cycle arrest in the G2/M phase by inhibiting cells from entering mitosis.
LRP6, the Wnt co-receptor, is activated more in RT-R-TNBC cells than in TNBC cells, and is involved in the induction of β-catenin, PLK1 and Cyclin B1
It has been reported that LDLR mediates the entry of cholesterol into cells (27), while LRP6, the Wnt co-receptor, activates the Wnt/β-catenin pathway upon cholesterol binding, resulting in the proliferation of cancer cells (28,29). If cholesterol imported into cells by LDLR activates Wnt/β-catenin signaling, increased intracellular cholesterol levels observed in siABCG8-transfected cells are expected to increase β-catenin. However, siABCG8-transfected cells exhibited increased intracellular cholesterol levels but reduced β-catenin (Fig. 3B-D). Therefore, the possibility that increased intracellular cholesterol via LDLR-mediated endocytosis could activate Wnt/β-catenin signaling in cells was excluded. Instead, the present study investigated whether the cholesterol-binding receptor LRP6, a Wnt co-receptor, could activate β-catenin and the associated EMT-related gene expression. RT-R-MDA-MB-231 cells exhibited increased levels of p-LRP6 compared with those in MDA-MB-231 cells (Fig. 5A and B). Knockdown of ABCG8 using siRNA decreased both LRP6 expression and LRP6 phosphorylation (Fig. 5C and D). Additionally, silencing of LRP6 resulted in reduced levels of β-catenin, PLK1 and Cyclin B1 compared with the siCTRL-transfected group (Fig. 5E-G). Furthermore, extracellular cholesterol treatment (10 and 20 μM; 24 h) did not significantly affect LRP6 phosphorylation and expression, and β-catenin signaling in RT-R-MDA-MB-231 cells at 10 μM, but did at 20 μM (Fig. S1A and B). When cholesterol was administered to MDA-MB-231 cells, which secreted less cholesterol than RT-R-TNBC cells, LRP6 phosphorylation and β-catenin induction were more prominent compared with those in RT-R-MDA-MB-231 cells from 10 μM (Fig. S1C and D). These findings indicated that increased LRP6 activation in RT-R-MDA-MB-231 cells was dependent on ABCG8 expression, and was associated with the induction of β-catenin, PLK1 and Cyclin B1 expression, suggesting that extracellular cholesterol exported by ABCG8 may affect LRP6 to activate the Wnt/β-catenin pathway.
RT-R-MDA-MB-231 cells exhibit increased SREBP1 (mature) and FASN expression and reduced LXRα/β and IDOL expression compared with MDA-MB-231 cells
SREBPs are well-known key transcription factors that control the expression of genes important for the uptake and synthesis of cholesterol, fatty acids and phospholipids (30,31), and it has been reported that FASN and cholesterol synthesis are linked under specific conditions (32,33). In particular, the SREBP1/FASN/cholesterol axis facilitates radioresistance in colorectal cancer (34); therefore, the present study investigated the SREBP1/FASN signaling pathways. RT-R-MDA-MB-231 cells exhibited a significant increase in the mature form of SREBP1 and FASN compared with MDA-MB-231 cells (Fig. 6A and B). Consistent with a report that hypoxia induces changes in lipid metabolism (35), RT-R-MDA-MB-231 cells exhibited increased HIF-1α levels (Fig. 6A and B), and treatment with digoxin, a HIF-1α inhibitor (at 400 nM for 24 h), led to a marked reduction in SREBP1 (mature form), and FASN protein expression in RT-R-MDA-MB-231 cells (Fig. 6C and D). However, ABCG8 expression was not affected by digoxin (Fig. 6C and D). In addition, RT-R-MDA-MB-231 cells exhibited increased levels of LDLR and decreased levels of LXRα/β and IDOL compared with those in MDA-MB-231 cells (Fig. 6E and F). These results suggested that RT-R-TNBC increased intracellular cholesterol levels by inducing synthesis of cholesterol and by dysregulating feedback mechanisms such as the LXRα/β and IDOL feedback mechanisms to reduce intracellular cholesterol by decreasing LDLR expression.
Discussion
ABC transporters are ATP-dependent transporters composed of 48 genes. They are subdivided into seven subfamilies (ABCA-ABCG) (36), and transport xenobiotics, metabolites and signaling molecules across intracellular and extracellular membranes (11). ABC transporters are well known for mediating multidrug resistance in cancer, which can lead to chemotherapy failure (37). Studies have suggested the important roles of ABC transporters in cancer beyond their established function as a multidrug efflux pump (11); they have been reported to influence tumor cells in various cancer types by regulating their malignant potential, including tumor cell proliferation, differentiation, migration and invasion (38,39). However, there is still a lack of understanding of how ABC transporters affect cancer progression.
In the present study, RT-R-MDA-MB-231 cells exhibited a significant increase in ABCG8 expression compared with MDA-MB-231 cells. Furthermore, knockdown of ABCG8 not only increased Doxo- and PTX-mediated cytotoxicity, but also significantly reduced cell viability on its own after 48 and 72 h of incubation, suggesting the additional function of ABCG8 beyond its role as a drug pump. Since ABCG8 is involved in sterol efflux (16), the present study investigated the levels of cholesterol inside cells and in the surrounding medium. The levels of cholesterol in cells and surrounding medium were significantly higher in the RT-R-MDA-MB-231 group than in the MDA-MB-231 group. Additionally, when ABCG8 was knocked down, there was a further increase in intracellular cholesterol levels but a reduction in medium cholesterol levels. This result demonstrated that RT-R-MDA-MB-231 cells exhibited higher levels of cholesterol, and that ABCG8 in RT-R-MDA-MB-231 cells increases cholesterol efflux.
Tumor cells have an increased need for lipids to grow and proliferate quickly. Lipid synthesis and uptake are elevated in various types of cancer, including breast cancer, and represent a novel characteristic of human cancer (18,40). Cholesterol is a major component of cell membranes; it controls cellular processes such as cell proliferation, differentiation, survival and apoptosis, and is also considered a risk factor and prognostic factor in cancer (41). Studies have shown that cholesterol serves a crucial role as a signaling molecule, regulating several pathways such as the PI3K, Hedgehog and Wnt/β-catenin pathways (42,43). Therefore, the present study investigated whether cholesterol exported by ABCG8 could function as a signaling molecule and whether it might be involved in cancer progression. The present results demonstrated that the expression levels of EMT-related proteins (β-catenin, N-cadherin and OCT4) were significantly higher in RT-R-MDA-MB-231 cells than in MDA-MB-231 cells. However, these increases were reduced by siABCG8 transfection. EMT-related proteins are known to be regulated by the PI3K/AKT signaling pathway in cancer (44,45). Additionally, the STAT3 pathway is a key regulator of breast cancer metastasis, promoting cancer progression, drug resistance, inhibition of apoptosis and EMT (46,47). However, although intracellular cholesterol levels were increased by siABCG8 transfection, the increased levels of p-AKT and p-STAT3 in RT-R-MDA-MB-231 cells were not affected by siABCG8 transfection. Therefore, this result suggested that cholesterol exported by ABCG8 affected the induction of EMT-related proteins but not via the p-AKT or p-STAT3 pathways.
It has been reported that cholesterol may serve a role in cancer biology by activating the Wnt pathway through specific interactions with Dishevelled and LRP6 (48). LRP6 is a crucial Wnt co-receptor in the Wnt/β-catenin signaling pathway. LRP6 expression is frequently upregulated in human breast cancer, and LRP6 is more frequently upregulated in TNBC (49). LRP6 promotes TNBC cell migration and invasion by interacting with frizzled (a transmembrane receptor family) and by activating the Wnt/β-catenin signaling pathway (50,51) involved in various physiological processes, such as proliferation, apoptosis, migration, invasion and homeostasis. Emerging reports have suggested that abnormal activation of the Wnt/β-catenin signaling pathway can promote cancer stem cell progression and cancer metastasis (52,53), and patients with TNBC with upregulated Wnt signaling often have a poor prognosis (54). Based on the significance of LRP6 in binding cholesterol and activating the Wnt/β-catenin pathway, which ultimately contributes to the maintenance and proliferation of cancer cells, the present study investigated the expression levels of LRP6 in RT-R-MDA-MB-231 cells, and revealed that the levels of p-LRP6 were increased in RT-R-MDA-MB-231 cells compared with MDA-MB-231 cells. siLRP6 transfection reduced the expression levels of β-catenin, PLK1 and Cyclin B1. Based on these results, it was hypothesized that increased cholesterol in RT-R-MDA-MB-231 cells may be exported by ABCG8 and that the elevated cholesterol could subsequently influence LRP6/Wnt/β-catenin signaling to activate the expression of β-catenin and other EMT-related genes. ABCG8 protein levels were higher in TNBC cells such as MDA-MB-231 and MDA-MB-453 cells than in MCF7 cells, a non-TNBC cell line. Furthermore, siABCG8 transfection also affected cholesterol efflux and the LRP6/Wnt/β-catenin signaling pathway in MDA-MB-231 cells. Therefore, ABCG8 may mediate cholesterol efflux and activate LRP6/Wnt/β-catenin signaling in TNBC. The present study emphasized that ABCG8 expression, cholesterol efflux from cytosol to media, and activation of the LRP6/Wnt/β-catenin signaling were enhanced in RT-R-MDA-MB-231 cells compared with MDA-MB-231 cells. In addition, knockdown of ABCG8 using siRNA reduced the levels of p-LRP6/LRP6, β-catenin and EMT-related proteins without reducing p-GSKβ or GSKβ. This result indicated that the levels of β-catenin may be controlled by factors other than GSKβ in the Wnt signaling pathway.
PLKs serve an essential role as key mitotic kinases and cell cycle regulators, and they also serve a role in proliferation and cell growth (21). PLKs are upregulated in various types of cancer, including ovarian cancer (22), breast cancer (55) and colorectal cancer (56), and promote entry into mitosis in the cell cycle by activating Cyclin B1 (23). Additionally, Wnt signaling-mediated β-catenin can activate cJUN, which results in increased expression levels of PLK1 (26). c-Jun is known as a transcriptional target and an interacting partner of β-catenin (57) and is also considered a possible transcriptional factor regulating PLK1 expression (58). Kim et al (26) demonstrated that TGFβ-activated PLK1 can phosphorylate β-catenin at Ser311, leading to increased stability, and promotes its nuclear translocation. Consequently, this process leads to elevated c-JUN expression, which, as part of the AP-1 complex, upregulates PLK1 expression, forming a positive feedback loop that facilitates EMT in metastatic non-small cell lung cancer. In the present study, ABCG8 knockdown significantly reduced the levels of PLK1 and Cyclin B1 in RT-R-MDA-MB-231 cells, resulting in cell cycle arrest in the G2/M phase. LRP6 siRNA transfection also reduced β-catenin, PLK1 and Cyclin B1 levels. These results suggested that cholesterol exported by ABCG8 could activate the LRP6/Wnt/β-catenin signaling pathway, which in turn could induce the expression of PLK1 and Cyclin B1, ultimately leading to cell cycle progression.
SREBP is a crucial transcription factor that controls the expression of enzymes involved in lipid metabolism and synthesis (30), and its expression is increased in various types of cancer, including colorectal cancer (59), breast cancer (60) and hepatocellular carcinoma (61). HIF-1α is a transcription factor that is induced under hypoxia, which is involved in tumor progression, including metastasis, drug resistance and cell cycle progression. HIF-1α is related to poor prognosis in various types of cancer (62-64), including colon cancer, prostate cancer and breast cancer (65). Under hypoxic conditions, tumor cells can activate HIF-1α. Activated HIF-1α then upregulates SREBP1 and activates FASN, which can increase hypoxia-induced chemotherapy resistance and promote tumor cell survival (66,67). Our previous study revealed that increased HIF-1α expression in RT-R-MDA-MB-231 cells contributed to tumor progression (8). In the present study, the expression levels of the mature form of SREBP1 and FASN were significantly increased in RT-R-MDA-MB-231 cells compared with in MDA-MB-231 cells, and increased SREBP1 and FASN levels in RT-R-MDA-MB-231 cells were significantly reduced by treatment with digoxin, an inhibitor of HIF-1α. These results suggested that the cholesterol levels in RT-R-MDA-MB-231 cells were increased via HIF-1α-mediated SREBP1-FASN induction. However, ABCG8 induction in RT-R-MDA-MB-231 cells was not HIF-1α-dependent, as digoxin, a HIF-1α inhibitor, failed to inhibit ABCG8 induction in RT-R-MDA-MB-231 cells. Further investigation is needed to understand the mechanism by which RT-R-MDA-MB-231 cells induce ABCG8. In normal cells, a high level of cholesterol triggers regulatory mechanisms that control the synthesis and uptake of cholesterol, leading to downregulation of SREBP and degradation of β-hydroxy β-methylglutaryl-CoA reductase. In addition, LXRα/β and IDOL are increased to reduce intracellular cholesterol by decreasing LDLR expression (68). However, cancer cells may be able to evade these feedback mechanisms in a cholesterol-rich environment. The present results showed that the expression of LXRα/β and IDOL, a feedback mechanism for the downregulation of intracellular cholesterol levels, was reduced in RT-R-MDA-MB-231 cells, resulting in increased LDLR expression. Dysregulation of feedback mechanisms as well as induction of cholesterol synthesis might maintain a high level of cholesterol within cells. The cholesterol was exported by ABCG8, which was upregulated in RT-R-MDA-MB-231 cells. The exported cholesterol in turn activated LRP6, the Wnt co-receptor, leading to the mediation of Wnt/β-catenin signaling. This activation induced the expression of PLK1, Cyclin B1 and EMT-related proteins, ultimately contributing to cancer progression (Fig. 7).
The increased cholesterol levels in highly metastatic TNBC that has acquired radiotherapy resistance may affect the maintenance of cell morphology because cholesterol is a component of cell membranes and vital for cell membrane integrity (41,69). However, to the best of our knowledge, the present study was the first to suggest the role of ABCG8 and that the cholesterol exported by ABCG8, not inside the cells, affects cancer progression through the LRP6/Wnt/β-catenin signaling pathway in RT-R-TNBC cells. The present results demonstrated that intracellular and medium cholesterol levels were significantly higher in the RT-R-MDA-MB-231 group compared with in the MDA-MB-231 group, and LRP6 was highly phosphorylated and β-catenin signaling was upregulated at baseline without any external stimulation in RT-R-MDA-MB-231 cells. This may explain why the effect of externally administered cholesterol was minimal in RT-R-MDA-MB-231 cells. Therefore, it may be hypothesized that high levels of intracellular cholesterol exported by ABCG8 may serve an important role in cancer progression by maintaining high levels of pericellular cholesterol and continuously stimulating cells. This needs to be further investigated. In conclusion, targeting the LRP6-Wnt/β-catenin signaling pathway may provide a potential therapeutic strategy for the treatment of RT-R-TNBC.
Supplementary Data
Availability of data and materials
The sequencing data generated in the present study may be found in the Gene Expression Omnibus database under accession number GSE287883 or at the following URL: https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE287883. The other data generated in the present study may be requested from the corresponding author.
Authors' contributions
YSK performed the experiments, analyzed the data and wrote the manuscript. JYW, HJ, NBN, YW and VN conducted the experiments and analyzed the data. SPY and SWP interpreted the results, developed the methodology and revised the manuscript. HJK conceived and designed the study, directed the project, and revised the manuscript. YSK, HJ and HJK confirmed the authenticity of all the raw data. All authors have read and approved the final version of the manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Abbreviations:
ABC |
ATP-binding cassette |
CTRL |
control |
EC |
endothelial cell |
EMT |
epithelial-mesenchymal transition |
ER |
estrogen receptor |
FASN |
fatty acid synthase |
HIF-1α |
hypoxia-inducible factor 1α |
IDOL |
inducible degrader of LDL |
LDL |
low-density lipoprotein |
LDLR |
LDL receptor |
LRP6 |
LDLR-related protein 6 |
LXRα/β |
liver X receptor α/β |
PLK1 |
Polo-like kinase 1 |
PR |
progesterone receptor |
RT-R |
radiotherapy-resistant |
siRNA |
small interfering RNA |
SREBP1 |
sterol regulatory element binding protein 1 |
TNBC |
triple-negative breast cancer |
Acknowledgements
Not applicable.
Funding
The present study was supported by the Basic Science Research Program through the National Research Foundation of Korea, funded by the Ministry of Education (grant nos. 2021R1A2B5B01001446 and RS-2024-003397886138211653 0101), and by the Development Fund Foundation, Gyeongsang National University, 2024.
References
Xu Y, Gong M, Wang Y, Yang Y, Liu S and Zeng Q: Global trends and forecasts of breast cancer incidence and deaths. Sci Data. 10:3342023. View Article : Google Scholar : PubMed/NCBI | |
Courtney D, Davey MG, Moloney BM, Barry MK, Sweeney K, McLaughlin RP, Malone CM, Lowery AJ and Kerin MJL: Breast cancer recurrence: Factors impacting occurrence and survival. Ir J Med Sci. 191:2501–2510. 2022. View Article : Google Scholar : PubMed/NCBI | |
de Ruijter TC, Veeck J, de Hoon JP, van Engeland M and Tjan-Heijnen VC: Characteristics of triple-negative breast cancer. J Cancer Res Clin Oncol. 137:1831922011. View Article : Google Scholar | |
Bai X, Ni J, Beretov J, Graham P and Li Y: Triple-negative breast cancer therapeutic resistance: Where is the Achilles' heel? Cancer Lett. 28(497): 100–111. 2021. View Article : Google Scholar | |
Zhang C, Wang S, Israel HP, Yan SX, Horowitz DP, Crockford S, Gidea-Addeo D, Chao KSC, Kalinsky K and Connolly EP: Higher locoregional recurrence rate for triple-negative breast cancer following neoadjuvant chemotherapy, surgery and radiotherapy. Springer Plus. 4:3862015. View Article : Google Scholar : PubMed/NCBI | |
Dent R, Trudeau M, Pritchard KI, Wedad MP, Harriet KH, Sawka CA, Lickley LA, Rawlinson E, Sun P and Narod SA: Triple-negative breast cancer: Clinical features and patterns of recurrence. Clin Cancer Res. 13:4429–4434. 2007. View Article : Google Scholar : PubMed/NCBI | |
Ko YS, Jin H, Lee JS, Park SW, Chang KC, Kang KM, Jeong BK and Kim HJ: Radioresistant breast cancer cells exhibit increased resistance to chemotherapy and enhanced invasive properties due to cancer stem cells. Oncol Rep. 40:3752–3762. 2018.PubMed/NCBI | |
Ko YS, Rugira T, Jin H, Joo YN and Kim HJ: Radiotherapy-resistant breast cancer cells enhance tumor progression by enhancing premetastatic niche formation through the HIF-1α-LOX axis. Int J Mol Sci. 21:80272020. View Article : Google Scholar | |
Modi A, Roy D, Sharma S, Vishnoi JR, Pareek P, Elhence P, Sharma P and Purohit P: ABC transporters in breast cancer: Their roles in multidrug resistance and beyond. J Drug Target. 9:927–947. 2022. View Article : Google Scholar | |
Beretta GL, Cassinelli G, Pennate M, Zuco V and Gatti L: Overcoming ABC transporter-mediated multidrug resistance: The dual role of tyrosine kinase inhibitors as multitargeting agents. Eur J Med Chem. 142:271–289. 2017. View Article : Google Scholar : PubMed/NCBI | |
Fletcher JI, Henderson MJ and Norris MD: ABC transporters in cancer: More than just drug efflux pumps. Nat Rev Cancer. 10:147–156. 2010. View Article : Google Scholar : PubMed/NCBI | |
Begicevic RR and Falasca M: ABC transporters in cancer stem cells: Beyond chemoresistance. Int J Mol Sci. 18:23622017. View Article : Google Scholar : PubMed/NCBI | |
Chen ZS and Tiwari AK: Multidrug resistance proteins (MRPs/ABCCs) in cancer chemotherapy and genetic diseases. FEBS J. 278:3266–3245. 2011. View Article : Google Scholar | |
Sun YL, Patel A, Kumar P and Chen ZS: Role of ABC transporters in cancer chemotherapy. Chin J Cancer. 31:51–57. 2012. View Article : Google Scholar : PubMed/NCBI | |
Sabeva NS, Liu J and Graf GA: The ABCG5ABCG8 sterol transporter and phytosterols: Implications for cardiometabolic disease. Curr Opin Endocrinol Diabetes Obes. 16:172–177. 2009. View Article : Google Scholar : PubMed/NCBI | |
Wang J, Mitsche MA, Lütjohann D, Cohen JC, Xie XS and Hobbs HH: Relative roles of ABCG5/ABCG8 in liver and intestine. J Lipid Res. 56:319–330. 2015. View Article : Google Scholar : | |
Xiao H, Zheng Y, Ma L, Tian L and Sun Q: Clinically-relevant ABC transporter for anti-Cancer drug resistance. Front Pharmacol. 19(12): 6484072021. View Article : Google Scholar | |
Cheng X, Li J and Guo D: SCAP/SREBPs are central players in lipid metabolism and novel metabolic targets in cancer therapy. Curr Top Med Chem. 18:484–493. 2018. View Article : Google Scholar : PubMed/NCBI | |
Thu KL, Soria-Bretones I, Mak TW and Cescona DW: Targeting the cell cycle in breast cancer: Towards the next phase. Cell Cycle. 17:1871–1885. 2018. View Article : Google Scholar : PubMed/NCBI | |
Lecarpentier Y, Schussler O, Hébert JL and Vallée A: Multiple targets of the canonical WNT/β-Catenin signaling in cancers. Front Oncol. 9:12482019. View Article : Google Scholar | |
Lee SY, Jang C and Lee KA: Polo-like kinases (plks), a key regulator of cell cycle and new potential target for cancer therapy. Dev Reprod. 18:65–71. 2014. View Article : Google Scholar : PubMed/NCBI | |
Kressin M, Fietz D, Becker S and Strebhardt K: Modelling the functions of Polo-Like Kinases in mice and their applications as cancer targets with a special focus on ovarian cancer. Cells. 10:11762021. View Article : Google Scholar : PubMed/NCBI | |
Solc P, Kitajima TS, Yoshida S, Brzakova A, Kaido M, Baran V, Mayer A, Samalova P, Motlik J and Ellenber J: Multiple requirements of PLK1 during mouse oocyte maturation. PLoS One. 10:e01167832015. View Article : Google Scholar : PubMed/NCBI | |
Kumar S, Sharma G, Chakraborty C, Sharma AR and Kim JB: Regulatory functional territory of PLK-1 and their substrates beyond mitosis. Oncotarget. 8:37942–37962. 2017. View Article : Google Scholar : PubMed/NCBI | |
Shah K and Kazi J: Phosphorylation-dependent regulation Wnt/beta-catenin signaling. Front oncol. 12:8587822022. View Article : Google Scholar | |
Kim DE, Shin SB, Kim CH, Kim YB, Oh HJ and Yim HS: PLK1-mediated phosphrylatioh of β-catenin enhances its stability and transcriptional activity for extracellular matrix remodeling in metastaic NSCLC. Theranostics. 13:1198–1216. 2023. View Article : Google Scholar : | |
Brown MS and Goldstein JL: A receptor-mediated pathway for cholesterol homeostasis. Science. 232:34–47. 1986. View Article : Google Scholar : PubMed/NCBI | |
Alrefaei AF and Abu-Elmagd M: LRP6 receptor plays essential functions in development and human diseases. Genes (Basel). 13:1202022. View Article : Google Scholar : PubMed/NCBI | |
Raisch J, Côté-Biron A and Rivard N: A role for the WNT Co-receptor LRP6 in pathogenesis and therapy of epithelial cancers. Cancers (Basel). 11:11622019. View Article : Google Scholar : PubMed/NCBI | |
Bengoechea-Alonso MT and Ericsson J: SREBP in signal transduction: Cholesterol metabolism and beyond. Curr Opin Cell Biol. 19:215–222. 2007. View Article : Google Scholar : PubMed/NCBI | |
Horton JD, Goldstein JL and Brown MS: SREBPs: Activators of the complete program of cholesterol and fatty acid synthesis in the liver. J Clin Invest. 109:1125–1131. 2002. View Article : Google Scholar : PubMed/NCBI | |
Eid W, Dauner K, Courtney KC, Gagnon A, Parks RJ, Sorisky A and Zha X: mTORC1 activates SREBP-2 by suppressing cholesterol trafficking to lysosomes in mammalian cells. Proc Natl Acad Sci USA. 114:7999–8004. 2017. View Article : Google Scholar : PubMed/NCBI | |
Carroll RG, Zasłona Z, Galvan-Pena S, Koppe EL, Sevin DC, Angiari S, Triantafilou M, Triantafilou K, Modis LK and O'Neill LA: An unexpected link between fatty acid synthase and cholesterol synthesis in proinflammatory macrophage activation. J Biol Chem. 293:5509–5521. 2018. View Article : Google Scholar : PubMed/NCBI | |
Jin Y, Chen Z, Dong J, Wang B, Fan S, Yang X and Cui M: SEBP1/FASN/cholesterol axis facilitates radioresistance in colorectal cancer. FEBS Open Bio. 11:1343–1352. 2021. View Article : Google Scholar : PubMed/NCBI | |
Mylonis I, Simos G and Paraskeva E: Hypoxia-inducible factors and the regulation of lipid metabolism. Cells. 8:2142019. View Article : Google Scholar : PubMed/NCBI | |
Dean M: The genetics of ATP-binding cassette transporters. Methods Enzymol. 400:409–429. 2005. View Article : Google Scholar | |
Fletcher JI, Williams RT, Henderson MJ, Norris MD and Haber M: ABC transporters as mediators of drug resistance and contributors to cancer cell biology. Drug Resist Updat. 26:1–9. 2016. View Article : Google Scholar : PubMed/NCBI | |
Copsel S, Garcia C, Diez F, Vermeulem M, Baldi A, Bianciotti LG, Russel FGM, Shayo C and Davio C: Multidrug resistance protein 4 (MRP4/ABCC4) regulates cAMP cellular levels and controls human leukemia cell proliferation and differentiation. J Biol Chem. 286:6979–6988. 2011. View Article : Google Scholar : PubMed/NCBI | |
Henderson MJ, Haber M, Porro A, Munoz MA, Iraci N, Xue C, Murray J, Flemming CL, Smith J and Fletcher JI: ABCC multidrug transporters in childhood neuroblastoma: Clinical and biological effects independent of cytotoxic drug efflux. J Natl Cancer Inst. 103:1236–1251. 2011. View Article : Google Scholar : PubMed/NCBI | |
Beloribi-Djefaflia S, Vasseur S and Guillaumond F: Lipid metabolic reprogramming in cancer cells. Oncogenesis. 5:e1892016. View Article : Google Scholar : PubMed/NCBI | |
Yan A, Jia Z, Qiao C, Wang M and Ding X: Cholesterol metabolism in drug-resistant cancer. Int J Oncol. 57:1103–1115. 2020. | |
Sheng R, Chen Y, Gee HY, Stec E, Melowic HR, Blatner NR, Tun MP, Kim YJ, Källberg M and Fujiwara TK: Cholesterol modulates cell signaling and protein networking by specifically interacting with PDZ domain-containing scaffold proteins. Nat Commun. 3:12492012. View Article : Google Scholar : PubMed/NCBI | |
Halimi H and Farjadian S: Cholesterol: An important actor on the cancer immune scene. Front Immunol. 13:10575462022. View Article : Google Scholar : PubMed/NCBI | |
Song JW: Targeting Epithelial-mesenchymal transition pathway in hepatocellular carcinoma. Clin Mol Hepatol. 26:484–486. 2020. View Article : Google Scholar : PubMed/NCBI | |
Maharati A and Moghbeli M: PI3K/AKT signaling pathway as a critical regulator of Epithelial-mesenchymal transition in colorectal tumor cells. Cell Commun Signal. 21:2012023. View Article : Google Scholar : PubMed/NCBI | |
Manore SG, Doheny DL, Wong GL and Lo HW: IL-6/JAK/STAT3 signaling in breast cancer metastasis: Biology and Treatment. Front Oncol. 12:8660142022. View Article : Google Scholar : PubMed/NCBI | |
Zhang G, Hou S, Li S, Wang Y and Cui W: Role of STAT3 in cancer cell Epithelial-mesenchymal transition. Int J Oncol. 64:482024. View Article : Google Scholar | |
Sheng R, Kim HJ, Lee HY, Xin Y, Chen Y, Tian Y, Cui Y, Choi JC, Doh JS, Han JK and Cho WH: Cholesterol selectively activates canonical Wnt signalling over Non-canonical Wnt signaling. Nat Commun. 5:43932014. View Article : Google Scholar | |
Liu CC, Prior J, Piwnica-Worms D and Bu G: LRP6 overexpression defines a class of breast cancer subtype and is a target for therapy. Proc Natl Acad Sci USA. 107:5136–5141. 2010. View Article : Google Scholar : PubMed/NCBI | |
Yang L, Wu X, Wang Y, Zhang KW, Ju Y, Yuan YC, Deng X, Chen L, Kim CCH and Lau S: FZD7 has a critical role in cell proliferation in triple negative breast cancer. Oncogene. 30:4437–4446. 2011. View Article : Google Scholar : PubMed/NCBI | |
Ma J, Lu W, Chen D, Xu B and Li Y: Role of Wnt Co-receptor LRP6 in triple negative breast cancer cell migration and invasion. J Cell Biochem. 118:2968–2976. 2017. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y and Wang X: Targeting the Wnt/β-catenin signaling pathway in cancer. J Hematol Oncol. 13:1652020. View Article : Google Scholar | |
Paskeh MDA, Mirzaei S, Ashrafizadeh M, Zarrabi A and Sethi G: Wnt/β-Catenin signaling as a driver of hepatocellular carcinoma progression: An emphasis on molecular pathways. J Hepatocell Carcinoma. 8:1415–1444. 2021. View Article : Google Scholar | |
Bianchini G, Balko JM, Mayer IA, Sanders ME and Gianni L: Triple-negative breast cancer: Challenges and opportunities of a heterogeneous disease. Nat Rev Clin Oncol. 13:674–690. 2016. View Article : Google Scholar : PubMed/NCBI | |
Weichert W: Polo-like kinase isoforms in breast cancer: Expression patterns and prognostic implications. Virchows Arch. 446:442–450. 2005. View Article : Google Scholar : PubMed/NCBI | |
Takahashi T: Polo-like kinase 1 (PLK1) is overexpressed in primary colorectal cancers. Cancer Sci. 94:148–152. 2003. View Article : Google Scholar : PubMed/NCBI | |
Mann B, Gelos M, Siedow A, Hanski ML, Gratchev A and Ilyas M: Target genes of beta-catenin-T cell-factor/lymphoid-enhancer-factor signaling in human colorectal carcinomas. Proc Natl Acad Sci USA. 96:1603–1608. 1999. View Article : Google Scholar : PubMed/NCBI | |
Martin BT and Strebhardt K: Polo-like kinase 1: Target and regulator of transcriptional control. Cell Cycle. 5:2881–2085. 2006. View Article : Google Scholar : PubMed/NCBI | |
Gao Y, Nan X, Shi X, Mu X, Liu B and Zhu H: SREBP1 promotes the invasion of colorectal cancer accompanied upregulation of MMP7 expression and NF-Kappa b pathway activation. BMC Cancer. 19:6852019. View Article : Google Scholar | |
Zhu Z, Zhao X, Zhao L, Yang H, Liu L and Li J: P54(nrb)/NONO regulates lipid metabolism and breast cancer growth through SREBP-1A. Oncogene. 35:1399–1410. 2006. View Article : Google Scholar | |
Li C, Yang W, Zhang J, Zheng X, Yao Y and Tu K: SREBP-1 has a prognostic role and contributes to invasion and metastasis in human hepatocellular carcinoma. Int J Mol Sci. 15:7124–7138. 2014. View Article : Google Scholar : PubMed/NCBI | |
Yong L, Tang S, Yu H, Zhang H, Zhang Y, Wan Y and Cai F: The role of hypoxia-inducible factor-1 alpha in multidrug-resistant breast cancer. Front Oncol. 12:9649342022. View Article : Google Scholar : PubMed/NCBI | |
Jun JC, Rathore A, Younas H, Gilkes D and Polotsky YV: Hypoxia-inducible factors and cancer. Curr Sleep Med Rep. 3:1–10. 2017. View Article : Google Scholar : PubMed/NCBI | |
Zhong H, De Marzo AM, Laughner E, Lim M, Hilton DA, Zagzag D, Buechler P, Isaacs WB, Semenza GL and Simons JW: Overexpression of hypoxia-inducible factor 1alpha in common human cancers and their metastases. Cancer Res. 59:5830–5835. 1999.PubMed/NCBI | |
Generali D, Berruti A, Brizzi MP, Campo L, Bonardi S and Wigfield S: Hypoxia-inducible factor-1alpha expression predicts a poor response to primary chemoendocrine therapy and disease-free survival in primary human breast cancer. Clin Cancer Res. 12:4562–4568. 2006. View Article : Google Scholar : PubMed/NCBI | |
Ezzeddini R, Taghikhani M, Somi MH, Samadi N and Rasaee MJ: Clinical importance of FASN in relation to HIF-1α and SREBP-1c in gastric adenocarcinoma. Life Sci. 224:169–176. 2019. View Article : Google Scholar : PubMed/NCBI | |
Furuta E, Pai SK, Zhan R, Bandyopadhyay S, Watabe M, Mo YY, Hirota S, Hosobe S, Tsukada T and Miura K: Fatty Acid Synthase gene is up-regulated by hypoxia via activation of Akt and Sterol Regulatory Element Binding Protein-1. Cancer Res. 68:1003–1011. 2008. View Article : Google Scholar : PubMed/NCBI | |
Zhang L, Reue K, Fong LG, Young SG and Tontonoz P: Feedback regulation of cholesterol uptake by the LXR-IDOL-LDLR axis. Arterioscler Thromb Vasc Biol. 32:2541–2546. 2012. View Article : Google Scholar : PubMed/NCBI | |
Yoon HJ, Jillian L, Shaw JL, Haigis MC and Greka A: Lipid metabolism in sickness and in health: Emerging regulators of lipotoxicity. Mol Cell. 81:3708–3730. 2021. View Article : Google Scholar : PubMed/NCBI |