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.
Non-small cell lung carcinoma (NSCLC), responsible for >80% of all lung cancer cases, is the deadliest form of malignancy worldwide (1). Despite advancements in therapeutic strategies such as surgical resection, chemotherapy, radiotherapy, immunotherapy and combination therapies, each focusing primarily on symptom alleviation, the 5-year overall survival rate of NSCLC remains <20% (2). This is further complicated by the widespread occurrence of chemoresistance and the increased metastatic potential of the disease, which undermine the efficacy of conventional treatments (3). This challenge is, at least in part, attributable to the self-renewal properties of cancer stem cells (4), which are resistant to chemotherapy, rendering traditional treatments less effective. As such, there is an urgent need for innovative therapeutic approaches targeting NSCLC cancer stem cells. Research has intensified efforts to understand self-renewal and tumor growth mechanisms, aiming to identify novel therapeutic targets or develop innovative curative interventions for various cancer types, including NSCLC (4–6). Studies have identified a small subset of tumor cells that, derived from bulk tumors, possess self-renewal and tumor-initiating abilities (7–9). These cells are typically enriched using cell surface markers (CD133, CD44, Sox2 and Oct4) or sphere formation assays in suspension culture (4,5,8,10,11). Notably, the sphere culture assay has proven effective in enriching sphere-forming cells (SFCs) from NSCLC cell lines, and SFCs exhibit stronger self-renewal potential (12,13). There is a critical need to develop effective therapeutic strategies, particularly novel targeted drugs aimed at directly addressing cancer cell self-renewal and tumor growth in NSCLC.
Genistein, an isoflavone found abundantly in soybeans and related products, has exhibited anticancer properties across various malignancies such as retinoblastoma, laryngeal cancer and colorectal cancer (14–16). 7-difluoromethoxyl-5,4′-di-n-octylgenistein (DFOG), a new genistein analog synthesized independently by the Department of Pharmacy, Hunan Normal University (Changsha, China), has been shown to induce apoptosis in ovarian cancer cells, and to inhibit their self-renewal and carcinogenesis (17,18). Despite these promising findings, the exact mechanism through which DFOG suppresses self-renewal and tumor growth in NSCLC cells remains to be fully elucidated.
MicroRNAs (miRNAs/miRs) are evolutionarily conserved, endogenous small noncoding RNAs, typically 19–23 nucleotides in length (19). Despite lacking protein-coding potential, they regulate a wide array of genes post-transcriptionally through complementary binding to mRNAs (20). These molecules are critical in processes such as apoptosis, differentiation, proliferation and metabolism, making them central to cancer development and stemness in cancer cells (21). One such miRNA, miR-152, has been identified as a tumor suppressor and is linked to malignant phenotypes of various types of cancer such as colon cancer, breast cancer, prostate cancer and ovarian cancer (22–25). A recent study has demonstrated that miR-152-3p is involved in the self-renewal and tumor growth of non-small cell lung cancer (26). Additionally, miR-152-3p is associated with tumor invasion, metastasis, drug resistance and proliferation (27,28). However, whether DFOG can inhibit the self-renewal and tumor growth of NSCLC by regulating miR-152-3p remains to be determined.
Persistent activation of STAT3 impacts gene regulation, thereby influencing self-renewal, migration and invasion in cancer cells (29,30). The miR-152/STAT3 axis is associated with poor prognosis in epithelial ovarian cancer (31). A previous study using JSI-124, a specific STAT3 inhibitor, suggested that suppressing STAT3 activation can diminish stem cell-like properties in hepatocellular carcinoma cells (32). However, it remains unclear whether miR-152-3p-mediated STAT3 inactivation can effectively reduce the self-renewal and tumor growth of SFCs derived from NSCLC. Therefore, the present study aimed to examine the hypothesis that reinstating miR-152-3p expression to suppress STAT3 can synergistically enhance the inhibitory effects of DFOG on self-renewal and tumor growth in SFCs derived from NSCLC.
The NCI-H460 and NCI-A549 human NSCLC cell lines were obtained from Shanghai Zhong Qiao Xin Zhou Biotechnology Co., Ltd., and Procell Life Science & Technology Co., Ltd., respectively, while the BEP2D human bronchial epithelial cell line was sourced from Otwo Biotech. All cell lines were authenticated through short tandem repeat profiling and mycoplasma testing. The cells were cultured in DMEM (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% FBS (Gibco; Thermo Fisher Scientific, Inc.), penicillin (100 U/ml) and streptomycin (100 µg/ml). All cells were maintained in a 5% CO2 incubator at 37°C. The STAT3 inhibitor S3I 201 (97%; cat. no. ab146606; Abcam) was stored at room temperature, and the cells were treated with S3I 201 (10 µM) at 37°C for 24 h before subsequent experiments.
To study sphere formation, H460 and A549 cells were cultured in stem-cell culture medium at a density of 5,000 cells per well in ultra-low attachment 6-well plates until spheres containing >20 cells formed (12,13). Stem-cell culture medium (DMEM/F12; Gibco; Thermo Fisher Scientific, Inc.), supplemented with 2% B27, 1% penicillin/streptomycin, basic fibroblast growth factor (20 ng/ml), epidermal growth factor (20 ng/ml) and insulin (4 µg/ml), was used. The inhibitory effects of DFOG on sphere formation were assessed by incubating SFCs derived from H460 and A549 cells with varying concentrations of DFOG (1, 5 and 10 µM) at 37°C for 72 h. Subsequently, the cells were reseeded at a density of 1,000 cells per well in ultra-low attachment 24-well plates and cultured until spheres reformed in the absence of DFOG. Subsequently, the number and status of spheres were evaluated manually under a light microscope (Leica Microsystems GmbH). The sphere formation rate was calculated using the following formula: Number of spheres/number of cells seeded ×100%. Experiments were performed in triplicate.
Cell viability was assessed using the Cell Counting Kit-8 (CCK-8) assay (Dojindo Laboratories, Inc.). Single-cell suspensions were seeded at 1,000 cells per well in 96-well plates for 24 h and treated with varying concentrations of DFOG (1, 5 and 10 µM) at 37°C. After 72 h, the cells were incubated with 10 µl CCK-8 solution per well for 2 h, and the optical density (OD450) was measured using a microplate reader (BioTek; Agilent Technologies, Inc.).
The Superscript IV RT kit and SYBR Green fluorophore were purchased from Thermo Fisher Scientific, Inc. The RT reaction conditions were incubation at 37°C for 5 min, 50°C for 15 min and 75°C for 5 min. Total RNA was extracted from H460 cells, A549 cells or SFCs (1×105 cells) using TRIzol® reagent (Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. For RNA extraction from tissue samples, grinding using a tissue homogenizer (Beyotime Institute of Biotechnology) was first performed. cDNA synthesis was conducted according to the supplier's instructions (Thermo Fisher Scientific, Inc.). The 2−ΔΔCq method (33) was used for qPCR, with U6 as the internal control for miR-152 and GAPDH as the internal control for STAT3. To identify candidate miRNAs affected by DFOG treatment, H460 cells, A549 cells or SFCs were treated with DFOG (5 µM) at 37°C for 24 h, followed by total RNA extraction and cDNA synthesis. PCR amplification was performed using specific primers (Table I), with the following thermocycling conditions: 95°C for 10 min, followed by 40 cycles of 95°C for 30 sec, 55°C for 30 sec and 70°C for 30 sec. For miRNA quantification, 2 µg total miRNA was transcribed and amplified using the All-in-One™ miRNA qRT-PCR Detection Kit (Applied Biosystems; Thermo Fisher Scientific, Inc.) and the TaqMan MicroRNA Assay (GeneCopoeia, Inc.), with U6 (Sangon Biotech Co., Ltd.) as the reference gene. Data analysis was conducted using the 2−ΔΔCq method. All experiments were conducted in triplicate independently.
For the colony formation assay, a bottom agar layer was prepared by mixing 1.2% agarose (Invitrogen; Thermo Fisher Scientific, Inc.) with DMEM in equal proportions, and 500 µl of this mixture was added to each well of a 24-well plate. The top agar layer was prepared by mixing H460 cells, A549 cells or SFCs (1,000 cells) with 0.7% agarose and 500 µl of 20% FBS-supplemented DMEM. After 12 h of cell seeding, different concentrations of DFOG (1, 5 and 10 µM) were added according to the needs of each group. The drug was continuously administered at 37°C until the end of the experiment. Images of colony formation were captured under a light microscope (Leica Microsystems GmbH) and colonies were counted manually. More than 20 cells were defined as a colony. The cells were incubated for 14 days at 37°C, and colonies were counted to calculate the colony formation rate per 1,000 cells based on triplicate experiments.
The RIPA protein extraction kit was purchased from Thermo Fisher Scientific, Inc. BCA was used to quantitatively determine the protein concentration. Each lane was loaded with 20 µg of protein. The gel concentration used was 10%. After electrophoresis, protein was transferred to a PVDF membrane. Blocking was performed using 5% skimmed milk at 37°C for 1 h. Membranes were incubated with the primary antibody at 4°C for 6 h, and membranes were incubated with the horseradish peroxidase-conjugated IgG secondary antibody (1:1,000 dilution; cat. no. RGAR011; Proteintech Group, Inc.) at room temperature for 1 h. Antibodies against α-tubulin (1:1,000 dilution; cat. no. 2125; Cell Signaling Technology, Inc.), STAT3 (1:1,000 dilution; cat. no. 12640; Cell Signaling Technology, Inc.), phosphorylated-STAT3 (p-STAT3; 1:2,000 dilution; cat. no. 9145; Cell Signaling Technology, Inc.), CD133 (1:1,000 dilution; cat. no. 64326; Cell Signaling Technology, Inc.), CD44 (1:1,000 dilution; cat. no. 37259; Cell Signaling Technology, Inc.), Oct4 (1:1,000 dilution; cat. no. 2890; Cell Signaling Technology, Inc.) and Sox2 (1:1,000 dilution; cat. no. 3579; Cell Signaling Technology, Inc.) were used as previously described (12). Finally, the chemiluminescent substrate (ECL) was purchased from Beyotime Institute of Biotechnology, and ImageJ 1.37 (National Institutes of Health) was used for gray-scale analysis.
MicrON™ miR-152-3p mimic (5′-UCAGUGCAUGACAGAACUUGG-3′) and micrOFF™ miR-152-3p inhibitor (5′-CCAAGUUCUGUCAUGCACUGA-3′), miR-152-3p mimic negative control (5′-UUGUACUACACAAAAGUACUG-3′) and miR-152-3p inhibitor negative control (5′-GGAACUUAGCCACUGUGAAUU-3′), were purchased from Guangzhou RiboBio Co., Ltd., and transfected into SFCs using transfection reagent iboFECT™ CP (Guangzhou RiboBio Co., Ltd.) at a concentration of 50 nM, according to the manufacturer's instructions. The small RNA complexes were incubated with cells for 2 h before the medium was replaced, then the culture was continued at 37°C for 48 h, and cells were used for subsequent experiments.
The binding sites of miR-152-3p and STAT3 were predicted using RNAhybrid (http://bibiserv.techfak.uni-bielefeld.de/rnahybrid). For the luciferase reporter assays, SFCs were co-transfected with miR-152-3p or miR-control and the pGL3 luciferase vector (Guangzhou RiboBio Co., Ltd.) containing the firefly luciferase reporter, along with the wild-type (WT) or mutant (MUT) 3′-untranslated region (UTR) sequence of STAT3. After 48 h, luciferase activity was measured using a luciferase assay kit (Promega Corporation), and normalized to Renilla luciferase activity in triplicate experiments.
Transfection was performed using 10 µg nucleic acid with a concentration of 1 µg/µl at 37°C for 48 h, and subsequent experiments were performed 48 h after transfection. For STAT3 overexpression, cells were transfected with pcDNA3.1-Control or pcDNA3.1-STAT3 plasmids obtained from Invitrogen; Thermo Fisher Scientific, Inc., using Lipofectamine™ 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) in Opti-MEM (Gibco; Thermo Fisher Scientific, Inc.), according to the manufacturer's guidelines.
A total of 18 female pathogen-free nude BALB/c mice (aged 4–5 weeks; weight, 18–22 g) were sourced from GemPharmatech Co., Ltd., and housed in a specific pathogen-free facility [SYXK (Xiang) 2020-0012] under a standard 12-h light/12-h dark cycle, at 20–26°C, with an atmospheric pressure of 20–50 Pa and a relative humidity of 40–70%, and ad libitum access to regular mouse chow and water. The site of cell injection was the armpit of the upper limb. When the tumor grew to ~100 cm3, the mouse was treated with drug treatment for 21 days. The time interval between the injection of cells and the end of the experiment was 6 weeks, and tumor volume was detected every 2 days until the end of the experiment. The ethical approval (approval no. D2023045) was granted by the Ethics Committee of Hunan Normal University (Changsha, China).
To evaluate the effects of DFOG in the xenograft mouse model, 1×106 SFCs were suspended in PBS and mixed with 100% Matrigel at a 1:1 ratio (BD Biosciences). A 100 µl mixture was subcutaneously injected into each mouse. When the xenograft volume reached ~100 mm3, mice in the control group received 200 µl of 2% DMSO every 2 days, while those in the experimental groups were orally administered DFOG (10 and 50 mg/kg) for 3 weeks every 2 days. Each group consisted of 6 mice. Tumor volume was calculated using the following formula: V (mm3)=(L × W2)/2, where L is the longest diameter and W is the shortest diameter of the xenograft, measured using a Vernier caliper. At the end of the experiment, xenograft-bearing mice were euthanized using CO2 asphyxiation (CO2 replacement rate of 30%), and the xenografts were collected, weighed, and snap-frozen in liquid nitrogen, and tumor tissues were fixed in 4% paraformaldehyde for subsequent H&E staining and immunohistochemistry. Tumor tissues for qPCR analysis were preserved in RNAlater.
For H&E staining, tumor tissues were fixed in 4% paraformaldehyde at 4°C for 24 h, and the slice thickness was 4 µm. Hematoxylin staining was performed for 5 min, followed by eosin staining for 1 min, and these were performed at 25°C. Staining was observed under a light microscope (Leica Microsystems GmbH).
Tumor tissues were fixed in 4% paraformaldehyde at 4°C for 24 h. Tissue sections (thickness, 4 µm) from paraffin-embedded and fixed samples were subjected to deparaffinization in citrate buffer. Sections were heated in an oven at 90°C for 20 min, followed by a series of ethanol washes (anhydrous ethanol, 95, 85 and 75% ethanol). Subsequently, the slices underwent three consecutive washes with PBS. After blocking endogenous peroxidase activity with 3% H2O2 and nonspecific binding with 5% goat serum (Beyotime Institute of Biotechnology) for 15 min at 25°C, the sections were incubated overnight at 4°C with the primary antibody against p-STAT3 (1:200 dilution; cat. no. 9145; Cell Signaling Technology, Inc.). As a negative control, PBS was used in place of the primary antibody. The sections were then incubated with horseradish peroxidase-conjugated anti-rabbit IgG antibodies (1:500 dilution; cat. no. RGAR011; Proteintech Group, Inc.) for 20 min at 25°C. Staining was developed using the 3,3′diaminobenzidine substrate (Fuzhou Maixin Biotechnology Development Co., Ltd.). The results were observed and images were captured under a light microscope (Leica Microsystems GmbH). The signal intensity was evaluated as previously described (34), and semi-quantitatively analyzed using ImageJ 1.37.
All statistical analyses were performed using GraphPad Prism Software 9 (Dotmatics). Data are presented as the mean ± SD. Comparisons between groups were performed using one-way analysis of variance with Tukey's post hoc test or an unpaired Student's t-test. For in vitro analyses, experiments were performed in triplicate. P<0.05 was considered to indicate a statistically significant difference.
The effect of DFOG on cell viability was assessed in BEP2D, H460 and A549 cell lines using the CCK-8 assay, following treatment at various concentrations. A reduction in cell viability was observed in H460 and A549 cells compared with the control group (0 µM), yielding an IC50 of ~10 µM (Fig. 1A). Noncytotoxic concentrations of DFOG (1, 5 and 10 µM) were selected for subsequent experiments.
Natural phytochemicals have been reported to modulate miRNA-mediated suppression of stemness characteristics in hepatocellular carcinoma cells (34). Therefore, it was detected whether tumor suppressor miRNAs in NSCLC cells, including miR-671-5p (31), miR-148a-3p (35), miR-340-5p (36), miR-342-3p (37), miR-34a-5p (38) and miR-152-3p (39), are regulated by DFOG (5 µM). DFOG treatment (5 µM) led to significant upregulation of miR-152-3p in both H460 and A549 cell lines, with the most notable increase among the tested miRNAs (Fig. 1B and C).
To examine the role of miR-152-3p and STAT3 expression in self-renewal and tumor growth in NSCLC, miR-152-3p expression was compared between H460 cells and H460-derived SFCs. The results revealed lower miR-152-3p levels in SFCs compared with H460 cells (Fig. 1D). Additionally, SFCs exhibited elevated STAT3 mRNA expression and p-STAT3 levels (Fig. 1E and F).
The sphere formation and colony formation were significantly increased in H460-derived SFCs compared with H460 cells (Fig. 1G and H). Furthermore, the expression levels of stem cell-associated markers, including CD133, CD44, Oct4 and Sox2, were elevated in H460-derived SFCs compared with H460 cells (Fig. 1I and J).
DFOG treatment at noncytotoxic concentrations (1, 5 and 10 µM) induced a dose-dependent increase in miR-152-3p expression in H460-derived SFCs (Fig. 2A). miR-152-3p has been recognized for its role in suppressing carcinogenesis by inhibiting carcinogenic transcription factors and signaling pathways in colon cancer, breast cancer, prostate cancer and ovarian cancer (22–25). Subsequently, the effects of DFOG on STAT3 mRNA expression and p-STAT3 protein levels were assessed using RT-qPCR and western blotting. DFOG treatment resulted in a decrease in STAT3 mRNA expression (Fig. 2B) and a significant reduction in p-STAT3 protein levels (Fig. 2C).
To evaluate the effect of DFOG on self-renewal and tumor growth in NSCLC, sphere formation and colony formation assays were performed. DFOG treatment led to reduced sphere (Fig. 2D) and colony (Fig. 2E) formation rates in H460-derived SFCs. Additionally, the protein levels of the stem cell markers CD133 and CD44 were significantly decreased (Fig. 2F). Consistent with these results, DFOG treatment substantially decreased the protein levels of Oct4 and Sox2 (Fig. 2G) in H460-derived SFCs.
For in vivo assessment, DFOG was orally administered to mice with H460-derived SFC xenograft tumors, with DMSO administered in the control group. The volumes of xenografts are shown in Table II and the maximum diameter is shown in Table III. As shown in Fig. 2H-1, −2 and −3, DFOG significantly suppressed the growth of xenograft tumors. Mechanistically, DFOG exerted a dual effect by reducing the p-STAT3 levels (Fig. 2H-4 and −5) and enhancing miR-152-3p expression (Fig. 2H-6). These results suggested that DFOG inhibited the self-renewal and tumor growth of H460-derived SFCs both in vitro and in vivo, likely through modulation of miR-152-3p and its target, STAT3.
Table III.Maximum diameter measured of the xenograft (mm) in each mouse across all experimental groups. |
To further elucidate the role of miR-152-3p regulation in DFOG-mediated suppression of self-renewal, H460-derived SFCs were transfected with a miR-152-3p mimic. As shown in Fig. 3A, the miR-152-3p mimic, in combination with DFOG (5 µM), elevated miR-152-3p expression. Furthermore, both DFOG (5 µM) and the miR-152-3p mimic synergistically reduced STAT3 mRNA levels and p-STAT3 protein levels (Fig. 3B and C). Notably, miR-152-3p overexpression enhanced the inhibitory effects of DFOG on self-renewal, leading to a reduction in sphere (Fig. 3D) and colony (Fig. 3E) formation rates compared with the control group (0 µM). The combined action of miR-152-3p and DFOG resulted in decreased expression of the stemness markers CD133 and CD44 (Fig. 3F), as well as the pluripotent factors Oct4 and Sox2 (Fig. 3G). These results suggested that DFOG elevated miR-152-3p expression, which in turn suppressed STAT3 transcription and activity, thereby inhibiting self-renewal in H460-derived SFCs.
To further validate the role of miR-152-3p regulation in DFOG-induced suppression of self-renewal, H460-derived SFCs were transfected with miR-152-3p inhibitor or miR-inhibitor-NC, followed by treatment with or without DFOG (5 µM). As shown in Fig. 4A, miR-152-3p inhibitor transfection effectively reversed the DFOG-induced increase in miR-152-3p expression. Additionally, miR-152-3p inhibitor counteracted the reduction in STAT3 mRNA expression and p-STAT3 protein levels induced by DFOG (Fig. 4B and C). Notably, miR-152-3p inhibitor mitigated the inhibitory effects of DFOG on self-renewal, as evidenced by an increase in sphere (Fig. 4D) and colony formation rates (Fig. 4E). miR-152-3p inhibitor transfection also increased the expression of the stemness-associated markers CD133 and CD44 (Fig. 4F), and the pluripotent factors Oct4 and Sox2 (Fig. 4G), reversing the suppressive effect of DFOG. These results suggested that inhibiting the expression of miR-152-3p could counteract the inhibition of p-STAT3 activity and self-renewal induced by DFOG treatment in H460-derived SFCs.
To investigate whether the DFOG-induced suppression of self-renewal was linked to the regulation of STAT3 mRNA expression and activity, H460-derived SFCs were treated with S3I 201, a specific STAT3 inhibitor. As shown in Fig. 5A, S3I 201 (10 µM) treatment did not affect the DFOG-induced elevation of miR-152-3p expression. However, as illustrated in Fig. 5B and C, S3I 201 (10 µM), in combination with DFOG (5 µM), effectively decreased STAT3 mRNA expression and p-STAT3 protein levels. Inhibition of STAT3 activity enhanced the suppressive effects of DFOG on self-renewal compared with S3I 201 (10 µM) or DFOG (5 µM) treatment alone, leading to a significant reduction in sphere (Fig. 5D) and colony (Fig. 5E) formation rates. Furthermore, the combination of S3I 201 and DFOG resulted in decreased protein levels of the stemness markers CD133 and CD44 (Fig. 5F), as well as the pluripotent factors Oct4 and Sox2 (Fig. 5G). These results highlighted that modulation of STAT3 mRNA expression and activity by DFOG contributed to the suppression of self-renewal in H460-derived SFCs.
To verify the upstream and downstream relationship between miR-152-3p and STAT3, H460-derived SFCs were transfected with STAT3 cDNA and pcDNA3.1, followed by treatment with or without DFOG (5 µM). As shown in Fig. 6A, transfection with STAT3 cDNA did not affect the miR-152-3p expression induced by DFOG. However, STAT3 cDNA transfection significantly counteracted the reduction in STAT3 mRNA and protein levels caused by DFOG (Fig. 6B and C).
To determine whether miR-152-3p directly targets STAT3, a luciferase reporter assay was conducted in H460-derived SFCs to identify the specific binding site of the miR-152-3p seed sequence on the 3′-UTR of STAT3 mRNA. RNAhybrid predicted binding sites for miR-152-3p and STAT3 (Fig. 7A). As shown in Fig. 7B, luciferase activity was reduced in cells co-transfected with the miR-152-3p mimic and STAT3-3′-UTR-WT, while no change in luciferase activity was observed following co-transfection with STAT3-3′-UTR-MUT. Additionally, the luciferase activity was further decreased in NSCLC cells co-transfected with miR-152-3p mimic and STAT3-3′-UTR-WT after DFOG (5 µM) treatment, compared with cells treated with miR-152-3p mimic or DFOG alone (Fig. 7C). These results demonstrated that DFOG inhibited self-renewal by upregulating miR-152-3p, which directly suppressed STAT3 expression by disrupting its transcriptional activity (Fig. 7D).
The inhibitory effect of DFOG on self-renewal was also assessed in A549-derived SFCs. Consistently, DFOG dose-dependently increased miR-152-3p expression, and downregulated STAT3 mRNA expression and p-STAT3 protein levels in A549 cells (Fig. 8A-C). Additionally, sphere formation (Fig. 8D) and colony formation (Fig. 8E) rates, as well as the expression of the stem cell markers CD44 and CD133 (Fig. 8F), and the pluripotent factors Oct4 and Sox2 (Fig. 8G), were all suppressed. These results indicated that DFOG-induced inhibition of self-renewal in A549-derived SFCs was mediated through the upregulation of miR-152-3p, and the suppression of STAT3 mRNA and activity.
The present study provided evidence that DFOG inhibited self-renewal and tumor growth in NSCLC cells by upregulating miR-152-3p, thereby suppressing the expression and activity of STAT3. These findings have substantial implications for the potential use of DFOG as a therapeutic strategy for human NSCLC, particularly targeting cancer cells with self-renewal-related stemness properties.
Dysregulated miRNA expression contributes to the progression of breast cancer, liver cancer and lung cancer (31,35–37,40,41). Reduced miR-152-3p expression has been linked to multiple aspects of malignancy, including progression, proliferation, invasion and metastasis, in colon cancer, breast cancer, prostate cancer and ovarian cancer (22–25). However, further investigation is required, particularly in lung cancer. Our results demonstrated that DFOG upregulated miR-152-3p, miR-34a-5p and miR-148a-3p, with miR-152-3p exhibiting the most pronounced increase. To the best of our knowledge, the present study was the first to report that DFOG enhanced self-renewal and tumor growth traits in NSCLC cells by elevating miR-152-3p expression, thus revealing a novel mechanism through which DFOG exerts its inhibitory effects on self-renewal and tumor growth. Furthermore, the molecular mechanisms underlying DFOG-induced suppression of tumor cells involve multiple signaling pathways, including the inactivation of FoxM1 and NF-κB (17,18). One study indicated that the expression of miR-152 and STAT3 was associated with poor prognosis in epithelial ovarian cancer (31). STAT3, an oncogenic transcription factor mediating signaling from the cell surface to the nucleus, is frequently upregulated in various malignancies, including NSCLC (29,30). Notably, targeting STAT3 has been shown to reduce tumor stem cell properties (30,41,42). In the present study, a luciferase reporter assay identified STAT3 as a direct target of miR-152-3p. This finding was further corroborated by the reduced transcription of STAT3 following combined treatment with DFOG and a miR-152-3p mimic in H460-derived SFCs. The present results demonstrated that DFOG-induced upregulation of miR-152-3p inhibited self-renewal and tumor growth by downregulating STAT3 expression at the mRNA level and impairing its activity, as evidenced by decreased p-STAT3 protein levels in H460-derived SFCs. Overexpression of STAT3 reversed this effect, highlighting the interconnection among DFOG, miR-152-3p and STAT3. Collectively, these findings suggested that the inhibitory effects of DFOG on self-renewal and tumor growth were mediated through miR-152-3p upregulation and STAT3 downregulation, underscoring its potential as a preventive and therapeutic agent for NSCLC, particularly in targeting self-renewing tumor cells.
In conclusion, the present study elucidated the mechanism by which DFOG targeted STAT3 through miR-152-3p upregulation, effectively suppressing self-renewal and tumor growth in NSCLC. Therefore, DFOG may be a promising candidate for novel preventive and therapeutic interventions for NSCLC in humans.
Not applicable.
The present study was funded by the Science and Technology Innovation Leading Academics of National High-level Personnel of Special Support Program from Ministry of Science and Technology, P.R. China (grant no. GKFZ-2018-29), Department of Science and Technology of Guizhou Province [grant no. QKHJC-ZK(2022)-YB-666], and the Natural Science Foundation of Hunan Province (grant no. 2021JJ30462).
The data generated in the present study may be requested from the corresponding author.
QY, XL and XC conducted the experiments, contributed to data collection and drafted the manuscript. JX and JZ performed the data analysis and contributed to the study design. XL, XC and JX provided resources. JZ and QY confirm the authenticity of all the raw data. All authors have read and approved the final version of the manuscript.
All animal studies were approved (approval no. D2023045) by the Ethics Committee of Hunan Normal University (Changsha, China).
Not applicable.
The authors declare that they have no competing interests.
|
DFOG |
7-difluoromethoxyl-5,4′-di-n-octylygenistein |
|
NSCLC |
non-small cell lung carcinoma |
|
p-STAT3 |
phosphorylated-STAT3 |
|
Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer statistics, 2022. CA Cancer J Clin. 72:7–33. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Chansky K, Detterbeck FC, Nicholson AG, Rusch VW, Vallières E, Groome P, Kennedy C, Krasnik M, Peake M, Shemanski L, et al: The IASLC lung cancer staging project: External validation of the revision of the TNM stage groupings in the eighth edition of the TNM classification of lung cancer. J Thorac Oncol. 12:1109–1121. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Wang C, Wu Y, Shao J, Liu D and Li W: Clinicopathological variables influencing overall survival, recurrence and post-recurrence survival in resected stage I non-small-cell lung cancer. BMC Cancer. 20:1502020. View Article : Google Scholar : PubMed/NCBI | |
|
Wang X, Chen Y, Wang X, Tian H, Wang Y, Jin J, Shan Z, Liu Y, Cai Z, Tong X, et al: Stem cell factor SOX2 confers ferroptosis resistance in lung cancer via upregulation of SLC7A11. Cancer Res. 81:5217–5229. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Clara JA, Monge C, Yang Y and Takebe N: Targeting signalling pathways and the immune microenvironment of cancer stem cells-a clinical update. Nat Rev Clin Oncol. 17:204–232. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Schaal CM, Bora-Singhal N, Kumar DM and Chellappan SP: Regulation of Sox2 and stemness by nicotine and electronic-cigarettes in non-small cell lung cancer. Mol Cancer. 17:1492018. View Article : Google Scholar : PubMed/NCBI | |
|
Sunayama J, Matsuda K, Sato A, Tachibana K, Suzuki K, Narita Y, Shibui S, Sakurada K, Kayama T, Tomiyama A and Kitanaka C: Crosstalk between the PI3K/mTOR and MEK/ERK pathways involved in the maintenance of self-renewal and tumorigenicity of glioblastoma stem-like cells. Stem Cells. 28:1930–1939. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Nimmakayala RK, Leon F, Rachagani S, Rauth S, Nallasamy P, Marimuthu S, Shailendra GK, Chhonker YS, Chugh S, Chirravuri R, et al: Metabolic programming of distinct cancer stem cells promotes metastasis of pancreatic ductal adenocarcinoma. Oncogene. 40:215–231. 202 View Article : Google Scholar : PubMed/NCBI | |
|
Sharif T, Martell E, Dai C, Singh SK and Gujar S: Regulation of the proline regulatory axis and autophagy modulates stemness in TP73/p73 deficient cancer stem-like cells. Autophagy. 15:934–936. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Kaufhold S, Garbán H and Bonavida B: Yin Yang 1 is associated with cancer stem cell transcription factors (SOX2, OCT4, BMI1) and clinical implication. J Exp Clin Cancer Res. 35:842016. View Article : Google Scholar : PubMed/NCBI | |
|
Walcher L, Kistenmacher AK, Suo H, Kitte R, Dluczek S, Strauß A, Blaudszun AR, Yevsa T, Fricke S and Kossatz-Boehlert U: Cancer stem cells-Origins and biomarkers: Perspectives for targeted personalized therapies. Front Immunol. 11:12802020. View Article : Google Scholar : PubMed/NCBI | |
|
Fu Z, Cao X, Yang Y, Song Z, Zhang J and Wang Z: Upregulation of FoxM1 by MnSOD overexpression contributes to cancer Stem-like cell characteristics in the lung cancer H460 cell line. Technol Cancer Res Treat. 17:15330338187896352018. View Article : Google Scholar : PubMed/NCBI | |
|
Liu F, Cao X, Liu Z, Guo H, Ren K, Quan M, Zhou Y, Xiang H and Cao J: Casticin suppresses self-renewal and invasion of lung cancer stem-like cells from A549 cells through down-regulation of pAkt. Acta Biochim Biophys Sin (Shanghai). 46:15–21. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Wei D, Yang L, Lv B and Chen L: Genistein suppresses retinoblastoma cell viability and growth and induces apoptosis by upregulating miR-145 and inhibiting its target ABCE1. Mol Vis. 23:385–394. 2017.PubMed/NCBI | |
|
Ma CH, Zhang YX, Tang LH, Yang XJ, Cui WM, Han CC and Ji WY: MicroRNA-1469, a p53-responsive microRNA promotes Genistein induced apoptosis by targeting Mcl1 in human laryngeal cancer cells. Biomed Pharmacother. 106:665–671. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang L, Zhang J, Gong Y and Lv L: Systematic and experimental investigations of the anti-colorectal cancer mediated by genistein. Biofactors. 46:974–982. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Ning Y, Xu M, Cao X, Chen X and Luo X: Inactivation of AKT, ERK and NF-κB by genistein derivative, 7-difluoromethoxyl-5,4′-di-n-octylygenistein, reduces ovarian carcinoma oncogenicity. Oncol Rep. 38:949–958. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Xiang HL, Liu F, Quan MF, Cao JG and Lv Y: 7-difluoromethoxyl-5,4′-di-n-octyl genistein inhibits growth of gastric cancer cells through down-regulating forkhead box M1. World J Gastroenterol. 18:4618–4626. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Ren W, Hou J, Yang C, Wang H, Wu S, Wu Y, Zhao X and Lu C: Extracellular vesicles secreted by hypoxia pre-challenged mesenchymal stem cells promote non-small cell lung cancer cell growth and mobility as well as macrophage M2 polarization via miR-21-5p delivery. J Exp Clin Cancer Res. 38:622019. View Article : Google Scholar : PubMed/NCBI | |
|
Li P, Xing W, Xu J, Yuan D, Liang G, Liu B and Ma H: microRNA-301b-3p downregulation underlies a novel inhibitory role of long non-coding RNA MBNL1-AS1 in non-small cell lung cancer. Stem Cell Res Ther. 10:1442019. View Article : Google Scholar : PubMed/NCBI | |
|
Xie C, Zhu J, Jiang Y, Chen J, Wang X, Geng S, Wu J, Zhong C, Li X and Meng Z: Sulforaphane inhibits the acquisition of tobacco Smoke-induced lung cancer stem cell-like properties via the IL-6/ΔNp63α/Notch axis. Theranostics. 9:4827–4840. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Lai SW, Chen MY, Bamodu OA, Hsieh MS, Huang TY, Yeh CT, Lee WH and Cherng YG: Exosomal lncRNA PVT1/VEGFA axis promotes colon cancer metastasis and stemness by downregulation of tumor suppressor miR-152-3p. Oxid Med Cell Longev. 2021:99598072021. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang CF, Xie YX, Qian YC, Wang M, Liu LZ, Shu YQ, Bai XM and Jiang BH: TBX15/miR-152/KIF2C pathway regulates breast cancer doxorubicin resistance via promoting PKM2 ubiquitination. Cancer Cell Int. 21:5422021. View Article : Google Scholar : PubMed/NCBI | |
|
Moya L, Meijer J, Schubert S, Matin F and Batra J: Assessment of miR-98-5p, miR-152-3p, miR-326 and miR-4289 expression as biomarker for prostate cancer diagnosis. Int J Mol Sci. 20:11542019. View Article : Google Scholar : PubMed/NCBI | |
|
Lyu M, Li X, Shen Y, Lu J, Zhang L, Zhong S and Wang J: CircATRNL1 and circZNF608 inhibit ovarian cancer by sequestering miR-152-5p and encoding protein. Front Genet. 13:7840892022. View Article : Google Scholar : PubMed/NCBI | |
|
Yuan Q, Wang R, Li X, Sun F, Lin J, Fu Z and Zhang J: DNMT1/miR-152-3p/SOS1 signaling axis promotes self-renewal and tumor growth of cancer stem-like cells derived from non-small cell lung cancer. Clin Epigenetics. 16:552024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao L, Wu X, Zhang Z, Fang L, Yang B and Li Y: ELF1 suppresses autophagy to reduce cisplatin resistance via the miR-152-3p/NCAM1/ERK axis in lung cancer cells. Cancer Sci. 114:2650–2663. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Huang J, Lou J, Liu X and Xie Y: LncRNA PCGsEM1 contributes to the proliferation, migration and invasion of Non-small cell lung cancer cells via acting as a sponge for miR-152-3p. Curr Pharm Des. 27:4663–4670. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Huang R, Wang S, Wang N, Zheng Y, Zhou J, Yang B, Wang X, Zhang J, Guo L, Wang S, et al: CCL5 derived from tumor-associated macrophages promotes prostate cancer stem cells and metastasis via activating β-catenin/STAT3 signaling. Cell Death Dis. 11:2342020. View Article : Google Scholar : PubMed/NCBI | |
|
Ouyang S, Li H, Lou L, Huang Q, Zhang Z, Mo J, Li M, Lu J, Zhu K, Chu Y, et al: Inhibition of STAT3-ferroptosis negative regulatory axis suppresses tumor growth and alleviates chemoresistance in gastric cancer. Redox Biol. 52:1023172022. View Article : Google Scholar : PubMed/NCBI | |
|
Cao Y, Shi H, Ren F, Jia Y and Zhang R: Long non-coding RNA CCAT1 promotes metastasis and poor prognosis in epithelial ovarian cancer. Exp Cell Res. 359:185–194. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Al-Harbi B and Aboussekhra A: Cucurbitacin I (JSI-124)-dependent inhibition of STAT3 permanently suppresses the pro-carcinogenic effects of active breast cancer-associated fibroblasts. Mol Carcinog. 60:242–251. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Li X, Wang L, Cao X, Zhou L, Xu C, Cui Y, Qiu Y and Cao J: Casticin inhibits stemness of hepatocellular carcinoma cells via disrupting the reciprocal negative regulation between DNMT1 and miR-148a-3p. Toxicol Appl Pharmacol. 396:1149982020. View Article : Google Scholar : PubMed/NCBI | |
|
Chen MJ, Cheng YM, Chen CC, Chen YC and Shen CJ: MiR-148a and miR-152 reduce tamoxifen resistance in ER+ breast cancer via downregulating ALCAM. Biochem Biophys Res Commun. 483:840–846. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
He WW, Ma HT, Guo X, Wu WM, Gao EJ and Zhao YH: lncRNA SNHG3 accelerates the proliferation and invasion of non-small cell lung cancer by downregulating miR-340-5p. J Biol Regul Homeost Agents. 34:2017–2027. 2020.PubMed/NCBI | |
|
Chen Z, Ying J, Shang W, Ding D, Guo M and Wang H: miR-342-3p regulates the proliferation and apoptosis of NSCLC cells by targeting BCL-2. Technol Cancer Res Treat. 20:153303382110411932021. View Article : Google Scholar : PubMed/NCBI | |
|
Li YY, Tao YW, Gao S, Li P, Zheng JM, Zhang SE, Liang J and Zhang Y: Cancer-associated fibroblasts contribute to oral cancer cells proliferation and metastasis via Exosome-mediated paracrine miR-34a-5p. EBioMedicine. 36:209–220. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Li LW, Xiao HQ, Ma R, Yang M, Li W and Lou G: miR-152 is involved in the proliferation and metastasis of ovarian cancer through repression of ERBB3. Int J Mol Med. 41:1529–1535. 2018.PubMed/NCBI | |
|
Chang DL, Wei W, Yu ZP and Qin CK: miR-152-5p inhibits proliferation and induces apoptosis of liver cancer cells by up-regulating FOXO expression. Pharmazie. 72:338–343. 2017.PubMed/NCBI | |
|
Reichenbach N, Delekate A, Plescher M, Schmitt F, Krauss S, Blank N, Halle A and Petzold GC: Inhibition of Stat3-mediated astrogliosis ameliorates pathology in an Alzheimer's disease model. EMBO Mol Med. 11:e96652019. View Article : Google Scholar : PubMed/NCBI | |
|
Liu YX, Xu BW, Niu XD, Chen YJ, Fu XQ, Wang XQ, Yin CL, Chou JY, Li JK, Wu JY, et al: Inhibition of Src/STAT3 signaling-mediated angiogenesis is involved in the anti-melanoma effects of dioscin. Pharmacol Res. 175:1059832022. View Article : Google Scholar : PubMed/NCBI |