Colorectal cancer (CRC) is a malignant tumor of the digestive system with high incidence rate and mortality. Tanshinone IIA (Tan IIA) plays an anti-cancer role in a variety of cancer cells. Here, we aimed to elucidate the therapeutic effects and potential mechanism of Tan IIA in CRC V体育官网.
OUMS23 cells were treated with 0, 5, 10, or 20 μM Tan IIA, and CRC mice were exposed to 20 μM Tan IIA + SLC7A11 plasmid. Edu and flow cytometry analyses were performed to assay cell proliferation and apoptosis, respectively. An Iron Assay Kit was used for determining total iron and Fe2+ levels in intracellular and tumor tissues. Lipid reactive oxygen species (ROS) production was evaluated by flow cytometry. SLC7A11 expression was analyzed by reverse transcription-quantitative PCR (RT-qPCR), Western blot assay, and immunohistochemistry (IHC) VSports手机版. The activation status of the phosphatidylinositol-3-kinase (PI3K)/AKT/mammalian target of rapamycin (TOR) pathway was determined by Western blotting.
Tan IIA suppressed CRC proliferation in a dose-dependent manner. Moreover, Tan IIA inhibited SLC7A11 expression in OUMS23 cells and tumor tissues. Functional assays showed that Tan IIA induces CRC apoptosis, ferroptosis, and ROS release in intracellular and tumor tissues, while SLC7A11 plasmid transfection reverses these effects. Furthermore, SLC7A11 plasmid reversed the effects of Tan IIA on tumor volume and weight in CRC subcutaneous tumors. Further experiments revealed that SLC7A11 plasmid abolished the effects of Tan IIA on the PI3K/AKT/mTOR pathway, as confirmed by increased phospho (p)-AKT and p-mTOR expression, as well as increased p-AKT/AKT and p-mTOR/ mTOR ratios V体育安卓版.
Tan IIA induces ferroptosis in CRC by suppressing SLC7A11 expression through the PI3K/AKT/mTOR pathway V体育ios版. Therefore, Tan IIA may be an effective therapeutic agent in the treatment of CRC.
CRC is a common malignant tumor of the digestive system originating from colorectal mucosa epithelium with high incidence rate and mortality [1, 2]. CRC mostly occurs in middle-aged men, most of whom are 40 to 70 years old. At present, the main treatment methods for CRC include surgery, chemotherapy, and immunotherapy [3,4,5]. Among them, chemotherapy is the preferred treatment. However, drug resistance and the inconspicuous early clinical manifestations of CRC before cancer metastasis develops hinder early diagnosis and treatment [6]. Chemoresistance is one of the major problems during chemotherapy for CRC and significantly limits the efficacy of the treatment and influences the prognosis of patients VSports最新版本. More than 90% of patients with metastatic cancer may fail chemotherapy due to drug resistance [7]. Therefore, reducing the incidence rate and mortality of CRC and finding effective treatment methods are urgent major clinical scientific issues.
Ferroptosis, a newly discovered cell death mode, is different from cell apoptosis, cell necrosis, autophagy, and other death modes [8]. An abnormal increase in cellular iron levels may lead to imbalances in redox homeostasis and lipid peroxidation of the cell membrane, eventually leading to cell death [9]. Ferroptosis involves many physiological and pathological processes, including cancer cell death, neurotoxicity, and T cell immunity [10, 11]. It also plays an important role in the occurrence and development of various diseases [12, 13]. Shin et al. showed that nuclear factor erythroid 2-related factor 2 (Nrf2) inhibition reverses resistance to glutathione peroxidase 4 (GPX4) inhibitor-induced ferroptosis in head and neck cancer [14] V体育平台登录. Yang et al. revealed that cetuximab promotes Ras-selective lethal 3 (RSL3)-induced ferroptosis by suppressing the Nrf2/heme oxygenase 1 (HO-1) signaling pathway in KRAS mutant CRC [15]. However, the specific mechanism of ferroptosis in CRC needs to be further explored.
Tan IIA, a widely used extract of Salvia miltiorrhiza, has a good inhibitory effect on tumor proliferation, migration, and angiogenesis [16]. Nie et al. found that Tan IIA regulates human acute myeloid leukemia cell proliferation, cell cycle, and apoptosis through the microRNA (miR)-497-5p/AKT3 axis [17]. Zhou et al. reported that Tan IIA suppresses ovarian cancer growth both in vitro and in vivo [18]. Qi et al. showed that Tan IIA markedly decreases the ATP level, glucose uptake, and lactate production in non-small cell lung cancer cells in vitro, and that it inhibits tumor growth in a xenograft model in vivo [19]. Moreover, Tan IIA induces ferroptosis in gastric cancer both in vitro and in vivo [20] VSports注册入口. Previous studies also evidenced Tan IIA is a vital regulator in CRC [21,22,23]. Qian et al. showed that Tan IIA alleviates the biological characteristics of CRC via activation of the ROS/c-Jun N-terminal kinase (JNK) signaling pathway [24]. Tan IIA also effectively inhibits CRC angiogenesis in vivo [22, 23]. However, the effects of Tan IIA on ferroptosis in CRC remain unknown.
Multiple pathways are involved in the progression of CRC [24,25,26]. PI3K/AKT/mTOR signaling pathway plays a key role in cancer development including proliferation, survival, metastasis, and angiogenesis [27]. PI3K/AKT/mTOR pathway is overexpressed and considered as a therapeutic target for colorectal cancer [28] V体育官网入口. PI3K/AKT pathway inhibition could sensitize cancer cells to ferroptosis induction [29]. And recent studies have revealed that mTOR is closely related to the occurrence and development of ferroptosis [30, 31]. Moreover, Tan IIA can suppress PI3K/AKT/mTOR pathway in several cancer cells [32, 33]. In this study, PI3K/AKT/mTOR pathway was analyzed.
Here, we aimed to analyze the effects of Tan IIA on ferroptosis in CRC and to clarify the potential mechanism VSports在线直播. Our findings provide a therapeutic basis for CRC therapy.
OUMS23 (a human CRC cell line) cells were obtained from ATCC and cultured in DMEM medium (Gibco) containing 5% FBS (AS1044, ASPEN) and 1% penicillin/streptomycin at 37 °C with 5% CO2. Cells were treated with Tan IIA (0, 5, 10, and 20 μM) for 48 h [34]. Tan IIA was obtained from MedChemExpress (HY-N0135, purity 99.78%) and dissolved in DMSO for use.
To determine cell proliferation, EdU assay was performed. Briefly, OUMS23 cells were plated into 96-well plates and cultured for 2 h. Then, the cells were exposed to 50 μM EdU and fixed with 4% paraformaldehyde for 20 min. After that, the supernatant was removed by centrifugation for 5 min, and the cells were stained using an Apollo reaction kit. The results were analyzed using a fluorescence microscope.
To determine the effect of Tan IIA on normal human intestinal epithelial cells (HIEC6), MTT assay was performed. Briefly, after treatment, HIEC6 cells were cultured into 96-well plates and incubated for 48 at 37 °C. Then, 10 μl MTT solution was added to each well and continuously incubated for further 4 h. And 100 μl DMSO was added to each well to dissolve the formazan product. Finally, the optical density (OD) at the wavelength of 570 nm was measured by a multifunctional plate reader (BioTek, Richmond, USA).
To determine the effect of Tan IIA on OUMS23 cell apoptosis, FCM assay was applied. After digesting with trypsin, OUMS23 cells were collected by centrifugation at 4 ℃ for 5 min. Then, the cells were washed twice with PBS (AS1044, ASPEN) and stained using the Annexin V/propidium iodide (PI) Apoptosis Detection Kit (Beyotime). After that, the cells were gently mixed and cultured for 20 min at room temperature without light. Annexin V-FITC and PI fluorescence were measured using a FACSCalibur flow cytometer (BD Technologies).
To determine the total iron and Fe2+ levels in OUMS23 cells and tissues, iron assay was performed. The Iron Assay Kit (Elabscience) was used to measure total iron and Fe2+ levels in OUMS23 cells and tissues. The iron assay buffer and iron reducer were successively added to the cells and tissues according to the instructions. Then, the samples were thoroughly mixed and cultured for 30 min after the addition of iron reducer without light and an iron probe. Lastly, the absorbance was measured at 593 nm.
After digesting with trypsin, OUMS23 cells were collected by centrifugation at 4℃ for 5 min. Then, the cells were treated with DCFH-DA dye solution and cultured for 30 min. Finally, fluorescence was examined using FACSAria II flow cytometer (BD Technologies).
RNA from OUMS23 cells and tissues was isolated using the TRIpure Total RNA Extraction Reagent (EP013, ELK Biotechnology) as per the manufacturer’s instructions. Then, the total RNA was reversed to cDNA using an EntiLink™ 1 st Strand cDNA Synthesis Super Mix (ELK Biotechnology) according to the instructions. SLC7A11 and GAPDH expression levels were determined using the following specific primers:
H-ACTIN, F-5′-GTCCACCGCAAATGCTTCTA-3′ and
R-5′-TGCTGTCACCTTCACCGTTC-3′;
SLC7A11, F-5′TGTGGGGTCCTGTCACTATTTG-3′ and
R-5′-GATATCACAGCAGTAGCTGCAGG-3′.
Thermocycling conditions used for qPCR were as follows: initial denaturation at 95 °C for 30 s; followed by 40 cycles of 10 s at 95 °C, 30 s at 58 °C and 30 s at 72 °C; and a final extension for 10 min at 72 °C. Relative changes in target gene expression were analyzed using the 2−ΔΔCt method.
OUMS23 cells and tissues were lysed for 30 min using RIPA buffer (AS1004, ASPEN). Proteins were then resolved by SDS-PAGE (AS1012, ASPEN) and transferred onto PVDF membranes. The membranes were blocked with 5% skimmed milk for 2 h to avoid non-specific binding and then incubated with primary antibodies against SLC7A11 (ab175186, 1:1000; Abcam), p-AKT (#9018; 1:1000; CST), AKT (#2938; 1:2000; CST), p-mTOR (#5536; 1:500; CST), mTOR (#2983, 1:1000; CST), or β-actin (TDY051, 1:10,000, Beijing TDY Biotech co., LTD.) at 4℃ overnight. After washing in TBST for three times, the membranes were incubated with secondary antibodies for 2 h. The protein signals were visualized by the addition of ECL (AS1059, ASPEN).
OUMS23 cells were transfected with control and SLC7A11 plasmids using Lipofectamine® 3000 reagent (Thermo) for 48 h following the manufacturer’s protocol. After that, RT-qPCR was performed to evaluate cell transfection efficiency.
Male BALB/c nude mice (4–6 weeks old, n = 6) were purchased from Beijing Vital River Laboratory Animal Technology Co., Ltd and bred under standard conditions with free access to food and water. After subcutaneous injection of 2 × 106 OUMS23 cells into their axillary skin tissue, the mice were treated with Tan IIA (50 mg/kg) or Tan IIA in combination with the control or SLC7A11 plasmid. The mice were divided into four groups: Group I, Control group; Group II, 50 mg/kg Tan IIA; Group III, 50 mg/kg Tan IIA + control plasmid; and Group IV, 50 mg/kg Tan IIA + SLC7A11 plasmid. Seven days after OUMS23 cell injection, Tan IIA + control or SLC7A11 plasmid was intraperitoneally injected every other day, and the mice were killed 28 days after tumor cell inoculation. Mice were anesthetized and sacrificed by cervical dislocation. Death was verified by observing cardiac and respiratory arrest. Then, tumor volume and weight were measured. The experiments were terminated when the mice lost >15% of their body weight prior to the injection. None of the mice died during the study period. The experimental protocols were approved by the Ethics Committee of The First People's Hospital of Lianyungang.
Tissue samples were fixed with 4% paraformaldehyde for 24 h, paraffin embedded, and sectioned. Then, the slices were placed in citrate buffer and baked in a 62 ℃ oven for 90 to 120 min. After that, the slides were incubated with 5% BSA for 30 min, followed by incubation with SLC7A11 (1:200, Triple Eagle, 26864-1-AP) at 4 °C overnight. Next, the sections were incubated with the secondary antibody for 30 min and examined by optical microscopy (OLYMPUS).
SPSS20.0 software was used for statistical analysis. The data are displayed as the mean ± standard deviation (SD) from three independent experiments, and comparisons between groups were carried out using one-way analysis of variance (ANOVA) or Student's t-test. Statistical significance was set at *P < 0.05 and **P < 0.01.
First, the effect of Tan IIA on normal human intestinal epithelial cells (HIEC6) was detected by MTT assay, and the results indicated that Tan IIA (0, 5, 10, and 20 μM) has no toxic effect on HIEC6 cells (Supplementary Fig. 1). We then analyzed the effects of Tan IIA on the proliferation and apoptosis of CRC cells. OUMS23 cells were exposed to Tan IIA (0, 5, 10, and 20 μM) for 48 h. The results from the Edu assay showed that Tan IIA inhibited OUMS23 cell proliferation in a dose-dependent manner (Fig. 1A). In addition, as illustrated in Fig. 1B, Tan IIA induced more apoptotic cell death than the Control. These findings reveal the protective effects of Tan IIA on CRC cells.
Effects of Tan IIA on OUMS23 cell proliferation and apoptosis. OUMS23 cells were exposed to various concentration of Tan IIA. A OUMS23 cells were labeled with EdU and their proliferation was examined. B Flow cytometry analysis of apoptotic cells. **P < 0.01 vs. the 0 μM Tan IIA treatment group
Ferroptosis is a form of iron-dependent cell death, which is different from necrosis and apoptosis [8]. Several studies have suggested that ferroptosis is a vital modality of CRC cell death [15, 35]. Therefore, we investigated the effects of Tan IIA on ferroptosis in OUMS23 cells. Tan IIA treatment increased the intracellular concentrations of total iron (Fig. 2A) and Fe2+ levels (Fig. 2B) in OUMS23 cells. Furthermore, lipid ROS levels increased after Tan IIA treatment (Fig. 2C). Thus, these findings suggested that Tan IIA suppresses cell proliferation via the induction of ferroptosis in OUMS23 cells.
Effects of Tan IIA on OUMS23 cell ferroptosis. Total iron (A) and ferrous iron (B) levels were determined in OUMS23 cells after treatment with Tan IIA. C The generation of lipid ROS in OUMS23 cells was analyzed using flow cytometry analysis. *P < 0.05, **P < 0.01 vs. the 0 μM Tan IIA treatment group
Ferroptosis is related to many diseases and can be regulated by multiple critical factors, including SLC7A11 [36]. Thus, we examined whether Tan IIA affects SLC7A11 expression in OUMS23 cells. Tan IIA reduced the expression of SLC7A11 in a dose-dependent manner in OUMS23 cells (Fig. 3A, B). To further analyze the roles of SLC7A11 in OUMS23 cells, the cells were transfected with a control or SLC7A11 plasmid and exposed to 20 μM Tan IIA. As displayed in Fig. 3C, D, SLC7A11 expression was up-regulated in SLC7A11 plasmid-transfected cells. In addition, we observed that Tan IIA substantially decreased the SLC7A11 expression in OUMS23 cells (Fig. 3E, F) and increased the intracellular total iron (Fig. 3G), Fe2+ (Fig. 3H), and ROS (Fig. 3I) levels. However, these effects were significantly suppressed by transfection with the SLC7A11 plasmid.
Effects of SLC7A11 plasmid transfection on Tan IIA-induced OUMS23 cell ferroptosis. A, B RT-qPCR and Western blot analysis of SLC7A11 levels in the Control and Tan IIA groups. C, D SLC7A11 levels in control plasmid- and SLC7A11 plasmid-transfected cells were analyzed using RT-qPCR and Western blot assay. E, F Evaluation of SLC7A11 levels using RT-qPCR and Western blot in Tan IIA + control plasmid and Tan IIA + SLC7A11 plasmid groups. Total iron (G) and ferrous iron (H) levels were determined. I Flow cytometry analysis quantifying lipid ROS production in OUMS23 cells. **P < 0.01 vs. the 0 μM Tanshinone IIA treatment group; ##P < 0.01 vs. the Control plasmid group; &&P < 0.01 vs. the Control group; $, $$P < 0.05, 0.01 vs. the Tan IIA + control plasmid group
Previous reports have suggested that multiple pathways are involved in the progression of CRC, including the PI3K/AKT pathway [37]. Consistently, we found that Tan IIA inhibited the p-AKT and p-mTOR expression in a dose-dependent manner in OUMS23 cells (Fig. 4A), as well as the ratio of p-AKT/AKT and p-mTOR/mTOR (Fig. 4B, C). In contrast, we observed the opposite results in SLC7A11 plasmid-transfected cells (Fig. 4D, F). These findings suggested that the PI3K/AKT/mTOR pathway acts as a vital player in CRC progression.
The PI3K/AKT/mTOR pathway is involved in the progression of CRC. OUMS23 cells were transfected with SLC7A11 plasmid or control plasmid and exposed to 20 μM Tan IIA. A Western blot analysis of p-AKT and p-mTOR expression. B, C The p-AKT/AKT and p-mTOR/mTOR ratios. D Detection of p-AKT and p-mTOR levels. E, F Quantification of the p-AKT/AKT and p-mTOR/mTOR ratios. *,**P < 0.05, 0.01 vs. the 0 μM Tan IIA treatment group; ##P < 0.01 vs. the Control group; &&P < 0.01 vs. the Tan IIA + control plasmid
Next, we conducted in vivo experiments to analyze the roles of SLC7A11 in CRC progression. The subcutaneous tumor models were treated with Tan IIA and infected with control plasmid or SLC7A11 plasmid. Figure 5A displays a representative diagram of the subcutaneous tumors. Moreover, tumor volumes and weights were measured. Tan IIA treatment decreased the volume and weight of the tumors in mice inoculated with OUMS23 cells. However, these tumor-suppressive effects were remarkably eliminated by SLC7A11 plasmid transfection (Fig. 5B, C). In conclusion, our data indicated that SLC7A11 overexpression blocks Tan IIA-mediated tumor inhibition.
Tan IIA suppresses CRC growth in vivo. A Representative images of the tumors are shown. B, C Tumor volumes and weights in each group were measured. **P < 0.01 vs. Control; ##P < 0.01 vs. Tan IIA + control plasmid
We further analyzed whether SLC7A11 affects cell death through its effects on ferroptosis. As shown in Fig. 6, Tan IIA prominently reduced the total iron (Fig. 6A), Fe2+ (Fig. 6B), and ROS (Fig. 6C) levels in tumor tissues, and these effects were reversed by transfection with the SLC7A11 plasmid.
Effects of SLC7A11 plasmid and Tan IIA on ferroptosis in CRC subcutaneous tumors. Total iron (A), ferrous iron (B), and lipid ROS (C) levels in CRC subcutaneous tumors were determined. **P < 0.01 vs. Control; #, ##P < 0.05, 0.01 vs. Tan IIA + control plasmid
We also determined the effects of SLC7A11 plasmid transfection on Tan IIA regulation of the PI3K/AKT/mTOR pathway in CRC subcutaneous tumors. Data from the IHC assay and RT-qPCR analysis demonstrated that Tan IIA significantly reduced the SLC7A11 mRNA and protein levels (Fig. 7A–C). Furthermore, the protein expression of p-AKT and p-mTOR (Fig. 7D), and the p-AKT/AKT and p-mTOR/mTOR ratio (Fig. 7E, F) in tumor tissues were decreased. However, these effects were eliminated by transfection with the SLC7A11 plasmid. These findings suggested that Tan IIA induces ferroptosis by inhibiting SLC7A11 expression via the PI3K/AKT/mTOR pathway in CRC.
Effects of SLC7A11 plasmid and Tan IIA on the PI3K/AKT/mTOR pathway in CRC subcutaneous tumors. A The expression of SLC7A11 as determined by IHC assays. B Quantification of SLC7A11 expression. C SLC7A11 levels measured by RT-qPCR. D Western blot analysis of p-AKT and p-mTOR expression. E, F Values of the p-AKT/AKT and p-mTOR/mTOR ratios. bar = 50 μm. **P < 0.01 vs. Control; ##P < 0.01 vs. Tan IIA + control plasmid
CRC is the third most common malignant tumor and the fourth leading cause of tumor-related death, with a high incidence rate and mortality [38]. The treatment methods of CRC include surgery, radiotherapy, molecular targeted therapy, and immunotherapy [39, 40]. At present, oxaliplatin is the first-line chemotherapy drug for CRC patients, but drug resistance remains an obstacle in the treatment of CRC [41]. At the same time, the symptoms of CRC are not obvious until metastasis develops, which also hinders early diagnosis. Therefore, new therapeutic targets for CRC are urgently needed.
Tan IIA is the main component isolated from Salvia miltiorrhiza. Tan IIA plays a protective role in angina pectoris and cerebral ischemia through its vasodilatory effect and anti-inflammatory activity [42]. In addition, Tan IIA regulates tumor development in gastric cancer [43], colon cancer [44], and CRC cancer [21]. In addition, Xie et al. found that the mechanisms of Tan IIA cytotoxicity include anti-proliferation, apoptosis induction, and endoplasmic reticulum stress induction in various cancer cell lines [45]. Our previous study revealed that Tan IIA significantly inhibits the proliferation and metastasis of CRC cells [46]. In this study, consistent with previous findings, we found that Tan IIA inhibits OUMS23 cell proliferation and induces apoptosis in a dose-dependent manner.
Ferroptosis, a newly discovered type of non-apoptotic programmed cell death that regulates cancer progression, may be a promising strategy for tumor treatment [8, 12, 35]. It is considered a beneficial therapeutic target for lung cancer and a new therapeutic target for bladder cancer [47]. Recent studies indicated that Tan IIA induces ferroptosis and ameliorates cisplatin resistance in gastric cancer cells through suppressing SLC7A11 expression [48, 49]. However, the effect of Tan IIA on ferroptosis in CRC remains unknown. Iron is a vital executor of ferroptosis. The intracellular iron level is regulated by iron regulatory transporters, and Fe2+ is particularly important for stimulating ferroptosis [42]. Therefore, we studied the effect of Tan IIA on ferroptosis in OUMS23 cells. Our data demonstrate that Tan IIA increases the concentration of total iron and Fe2+ in OUMS23 cells. GPX4 is the central mediator of ferroptosis and induces cancer cell death accompanied by the production of lipid ROS [50]. We found that Tan IIA-treated cells have increased lipid ROS levels. These findings indicate that Tan IIA induces ferroptosis in OUMS23 cells.
SLC7A11 is highly expressed in human tumors, including CRC [51]. SLC7A11 inhibition can induce ferroptosis. In contrast, SLC7A11 overexpression may protect cancer cells from ferroptosis. Sun et al. showed that lidocaine promotes ferroptosis in ovarian and breast cancer by targeting the miR-382-5p/SLC7A11 axis [52]. In addition, studies have shown that Tan IIA can promote ferroptosis by down-regulating the expression of SLC7A11 in breast cancer and gastric cancer [53, 54]. In our study, we found that Tan IIA decreases the expression of SLC7A11 in OUMS23 cells in a dose-dependent manner. To analyze the roles of SLC7A11 in OUMS23 cells, OUMS23 cells were transfected with control plasmid or SLC7A111 plasmid and exposed to 20 μM Tan IIA. SLC7A11 expression was down-regulated in Tan IIA-treated cells and increased in SLC7A11 plasmid-transfected cells. Moreover, SLC7A11 plasmid transfection reversed the effects of Tan IIA on ferroptosis, as confirmed by decreased intracellular total iron, Fe2+, and ROS levels. Thus, Tan IIA induces ferroptosis in CRC cells by inhibiting SLC7A11 expression.
In this study, PI3K/AKT/mTOR pathway was analyzed. Consistent with this, we found that Tan IIA inhibits p-AKT and p-mTOR expression and the ratio of p-AKT/AKT and p-mTOR/mTOR in OUMS23 cells in a dose-dependent manner. However, we observed the opposite results in the SLC7A11 plasmid transfection group. Our findings demonstrate that the PI3K/AKT/mTOR pathway plays an important role in CRC progression. The roles of SLC7A11 have been investigated both in vitro and in vivo. Based on the above findings, we established a nude mouse model of CRC subcutaneous tumors and clarified the potential regulatory mechanism between Tan IIA and SLC7A11. The xenograft tumor model was treated with Tan IIA and infected with control plasmid or SLC7A11 plasmid. We measured the volume and weight of solid tumors and found that the addition of the SLC7A11 plasmid reversed the effect of Tan IIA on tumor growth and ferroptosis in CRC subcutaneous tumors, suggesting that Tan IIA could inhibit CRC growth by regulating SLC7A11, and Tan IIA may be a potential therapeutic agent in the treatment of CRC. Immunofluorescence, RT-qPCR, and Western blot analyses revealed that the SLC7A11 plasmid reverses the effects of Tan IIA on the PI3K/AKT/mTOR pathway in CRC subcutaneous tumors.
There were also some limitations of the current study. First, only one CRC cell line (OUMS23) was used in this study to study the effect of Tan IIA on CRC cells, and studying more CRC cell lines will make the results of this study more convincing. Besides, the specific mechanisms underlying the regulatory effects of Tan IIA on SLC7A11, especially the potential involvement of microRNAs or other signaling pathways in this process has not been explored. Moreover, the subcutaneous xenograft tumor model in nude mice enables human tumor cells to find a living carrier to study the growth and metastasis of human tumor with the help of nude mouse. However, due to the lack of T cell immunity and heterogeneous growth environment, the subcutaneous xenograft tumor model in nude mice cannot fully represent the complex microenvironment of colorectal cancer.
Taken together, our findings demonstrate that Tan IIA induces ferroptosis by suppressing SLC7A11 expression through the PI3K/AKT/mTOR pathway in CRC. Our findings may supply a powerful experimental basis for the use of Tan IIA in the treatment of CRC, and Tan IIA may as a candidate for clinical treatment of CRC.
The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.
Colorectal cancer
Glutathione peroxidase 4
Heme oxygenase 1
Immunohistochemistry
C-Jun N-terminal kinase
MicroRNA
Mammalian target of rapamycin
Nuclear factor erythroid 2-related factor 2
Propidium iodide
Phosphatidylinositol 3-kinase
Reactive oxygen species
Ras-selective lethal 3
Reverse transcription-quantitative PCR
Tanshinone IIA
Liang X, Hendryx M, Qi L, Lane D, Luo J. Association between prediagnosis depression and mortality among postmenopausal women with colorectal cancer. PLoS ONE. 2020;15:e0244728. https://doi.org/10.1371/journal.pone.0244728.
Kroenke CH, Prado CM, Meyerhardt JA, Weltzien EK, Xiao J, Cespedes FE, Caan BJ. Muscle radiodensity and mortality in patients with colorectal cancer. Cancer. 2018;124:3008–15. VSports app下载 - https://doi.org/10.1002/cncr.31405.
Crone V, Hasselager RP, Fransgaard T, Gogenur I. Anaesthetic technique and outcomes after colorectal cancer surgery. Dan Med J. 2020;67(4):A04190255.
Aoullay Z, Slaoui M, Razine R, Er-Raki A, Meddah B, Cherrah Y. Therapeutic characteristics, chemotherapy-related toxicities and survivorship in colorectal cancer patients. Ethiop J Health Sci. 2020;30:65–74. V体育平台登录 - https://doi.org/10.4314/ejhs.v30i1.9.
Fan A, Wang B, Wang X, Nie Y, Fan D, Zhao X, Lu Y. Immunotherapy in colorectal cancer: current achievements and future perspective. Int J Biol Sci. 2021;17:3837–49. https://doi.org/10.7150/ijbs.64077.
Sun T, Li K, Xu G, Zhu K, Wang Q, Dang C, Yuan D. Postoperative oxaliplatin-based hyperthermic intraperitoneal chemotherapy: an effective and safe palliative treatment option for colorectal cancer with peritoneal metastasis. World J Surg Oncol. 2021;19:200. https://doi.org/10.1186/s12957-021-02320-4.
Longley DB, Johnston PG. Molecular mechanisms of drug resistance. J Pathol. 2005;205(2):275–92. https://doi.org/10.1002/path.1706.
Wang Y, Wei Z, Pan K, Li J, Chen Q. The function and mechanism of ferroptosis in cancer. Apoptosis. 2020;25:786–98. https://doi.org/10.1007/s10495-020-01638-w.
Li D, Zhang M, Chao H. Significance of glutathione peroxidase 4 and intracellular iron level in ovarian cancer cells-"utilization" of ferroptosis mechanism. Inflamm Res. 2021;70:1177–89. https://doi.org/10.1007/s00011-021-01495-6.
Liu P, Yuan J, Feng Y, Chen X, Wang G, Zhao L. Ferroptosis contributes to isoflurane-induced neurotoxicity and learning and memory impairment. Cell Death Discov. 2021;7:72. https://doi.org/10.1038/s41420-021-00454-8.
Lei T, Qian H, Lei P, Hu Y. Ferroptosis-related gene signature associates with immunity and predicts prognosis accurately in patients with osteosarcoma. Cancer Sci. 2021;112:4785–98. https://doi.org/10.1111/cas.15131 (VSports app下载).
Jiang X, Stockwell BR, Conrad M. Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 2021;22:266–82. https://doi.org/10.1038/s41580-020-00324-8.
Wu X, Li Y, Zhang S, Zhou X. Ferroptosis as a novel therapeutic target for cardiovascular disease. Theranostics. 2021;11:3052–9. "V体育2025版" https://doi.org/10.7150/thno.54113.
Shin D, Kim EH, Lee J, Roh JL. Nrf2 inhibition reverses resistance to GPX4 inhibitor-induced ferroptosis in head and neck cancer. Free Radic Biol Med. 2018;129:454–62. https://doi.org/10.1016/j.freeradbiomed.2018.10.426.
Yang J, Mo J, Dai J, Ye C, Cen W, Zheng X, Jiang L, Ye L. Cetuximab promotes RSL3-induced ferroptosis by suppressing the Nrf2/HO-1 signalling pathway in KRAS mutant colorectal cancer. Cell Death Dis. 2021;12:1079. https://doi.org/10.1038/s41419-021-04367-3.
Ni H, Ruan G, Sun C, Yang X, Miao Z, Li J, Chen Y, Qin H, Liu Y, Zheng L, Xing Y, Xi T, Li X. Tanshinone IIA inhibits gastric cancer cell stemness through inducing ferroptosis. Environ Toxicol. 2022;37:192–200. https://doi.org/10.1002/tox.23388.
Nie ZY, Zhao MH, Cheng BQ, Pan RF, Wang TR, Qin Y, Zhang XJ. Tanshinone IIA regulates human AML cell proliferation, cell cycle, and apoptosis through miR-497-5p/AKT3 axis. Cancer Cell Int. 2020;20:379. https://doi.org/10.1186/s12935-020-01468-5.
Zhou J, Jiang YY, Wang XX, Wang HP, Chen H, Wu YC, Wang L, Pu X, Yue GZ, Zhang L. Tanshinone IIA suppresses ovarian cancer growth through inhibiting malignant properties and angiogenesis. Ann Transl Med. 2020;8:1295. https://doi.org/10.21037/atm-20-5741 (VSports app下载).
Qi H, Chen Z, Qin Y, Wang X, Zhang Z, Li Y. Tanshinone IIA inhibits cell growth by suppressing SIX1-induced aerobic glycolysis in non-small cell lung cancer cells. Oncol Lett. 2022;23:184. https://doi.org/10.3892/ol.2022.13304.
Guan Z, Chen J, Li X, Dong N. Tanshinone IIA induces ferroptosis in gastric cancer cells through p53-mediated SLC7A11 down-regulation. Biosci Rep. 2020;40:BSR20201807. https://doi.org/10.1042/BSR20201807.
Qian J, Cao Y, Zhang J, Li L, Wu J, Yu J, Huo J. Tanshinone IIA alleviates the biological characteristics of colorectal cancer via activating the ROS/JNK signaling pathway. Anticancer Agents Med Chem. 2022. https://doi.org/10.2174/1871520622666220421093430.
Sui H, Zhao J, Zhou L, Wen H, Deng W, Li C, Ji Q, Liu X, Feng Y, Chai N, Zhang Q, Cai J, Li Q. Tanshinone IIA inhibits β-catenin/VEGF-mediated angiogenesis by targeting TGF-β1 in normoxic and HIF-1α in hypoxic microenvironments in human colorectal cancer. Cancer Lett. 2017;403:86–97. https://doi.org/10.1016/j.canlet.2017.05.013.
Liu L, Gao H, Wen T, Gu T, Zhang S, Yuan Z. Tanshinone IIA attenuates AOM/DSS-induced colorectal tumorigenesis in mice via inhibition of intestinal inflammation. Pharm Biol. 2021;59:89–96. https://doi.org/10.1080/13880209.2020.1865412 (VSports app下载).
Yan H, Talty R, Johnson CH. Targeting ferroptosis to treat colorectal cancer. Trends Cell Biol. 2023;33:185–8. https://doi.org/10.1016/j.tcb.2022.11.003.
Koppula P, Zhuang L, Gan B. Cystine transporter SLC7A11/xCT in cancer: ferroptosis, nutrient dependency, and cancer therapy. Protein Cell. 2021;12:599–620. https://doi.org/10.1007/s13238-020-00789-5.
Ma Z, Lou S, Jiang Z. PHLDA2 regulates EMT and autophagy in colorectal cancer via the PI3K/AKT signaling pathway. Aging (Albany NY). 2020;12:7985–8000. VSports注册入口 - https://doi.org/10.18632/aging.103117.
Li N, Lu B, Luo C, Cai J, Lu M, Zhang Y, Chen H, Dai M. Incidence, mortality, survival, risk factor and screening of colorectal cancer: a comparison among China, Europe, and northern America. Cancer Lett. 2021;522:255–68. https://doi.org/10.1016/j.canlet.2021.09.034.
Ebrahimzadeh S, Ahangari H, Soleimanian A, Hosseini K, Ebrahimi V, Ghasemnejad T, Soofiyani SR, Tarhriz V, Eyvazi S. Colorectal cancer treatment using bacteria: focus on molecular mechanisms. BMC Microbiol. 2021;21:218. https://doi.org/10.1186/s12866-021-02274-3.
Dong J, Li Y, Xiao H, Zhang S, Wang B, Wang H, Li Y, Fan S, Cui M. Oral microbiota affects the efficacy and prognosis of radiotherapy for colorectal cancer in mouse models. Cell Rep. 2021;37: 109886. https://doi.org/10.1016/j.celrep.2021.109886 (V体育平台登录).
Li Y, Gan Y, Liu J, Li J, Zhou Z, Tian R, Sun R, Liu J, Xiao Q, Li Y, Lu P, Peng Y, Peng Y, Shu G, Yin G. Downregulation of MEIS1 mediated by ELFN1-AS1/EZH2/DNMT3a axis promotes tumorigenesis and oxaliplatin resistance in colorectal cancer. Signal Transduct Target Ther. 2022;7:87. https://doi.org/10.1038/s41392-022-00902-6.
Chen Z, Gao X, Jiao Y, Qiu Y, Wang A, Yu M, Che F, Li S, Liu J, Li J, Zhang H, Yu C, Li G, Gao Y, Pan L, Sun W, Guo J, Cao B, Zhu Y, Xu H. Tanshinone IIA exerts anti-inflammatory and immune-regulating effects on vulnerable atherosclerotic plaque partially via the TLR4/MyD88/NF-kappaB signal pathway. Front Pharmacol. 2019;10:850. https://doi.org/10.3389/fphar.2019.00850.
Guan Z, Chen J, Li X, Dong N. Tanshinone IIA induces ferroptosis in gastric cancer cells through p53-mediated SLC7A11 down-regulation. Biosci Rep. 2020;40(8):BSR20201807. "VSports" https://doi.org/10.1042/BSR20201807.
Qian J, Cao Y, Zhang J, Li L, Wu J, Wei G, Yu J, Huo J. Tanshinone IIA induces autophagy in colon cancer cells through MEK/ERK/mTOR pathway. Transl Cancer Res. 2020;9:6919–28. https://doi.org/10.21037/tcr-20-1963.
Song Q, Yang L, Han Z, Wu X, Li R, Zhou L, Liu N, Sui H, Cai J, Wang Y, Ji Q, Li Q. Tanshinone IIA inhibits epithelial-to-mesenchymal transition through hindering beta-arrestin1 mediated beta-catenin signaling pathway in colorectal cancer. Front Pharmacol. 2020;11: 586616. https://doi.org/10.3389/fphar.2020.586616.
Xie J, Liu J, Liu H, Liang S, Lin M, Gu Y, Liu T, Wang D, Ge H, Mo SL. The antitumor effect of tanshinone IIA on anti-proliferation and decreasing VEGF/VEGFR2 expression on the human non-small cell lung cancer A549 cell line. Acta Pharm Sin B. 2015;5:554–63. https://doi.org/10.1016/j.apsb.2015.07.008.
Zhang Y, Zhao Y, Liu J, Li C, Feng Y, Jiang S, Sun X, Hu X, Wang Y. Transgelin-2 involves in the apoptosis of colorectal cancer cells induced by Tanshinone-IIA. Anal Cell Pathol (Amst). 2022;2022:9358583. https://doi.org/10.1155/2022/9358583.
Zeng F, Lan Y, Wang N, Huang X, Zhou Q, Wang Y. Ferroptosis: a new therapeutic target for bladder cancer. Front Pharmacol. 2022;13:1043283. https://doi.org/10.3389/fphar.2022.1043283.
Guan Z, Chen J, Li X, Dong N. Tanshinone IIA induces ferroptosis in gastric cancer cells through p53-mediated SLC7A11 down-regulation. Biosci Rep. 2020;40:BSR20201807. https://doi.org/10.1042/BSR20201807.
Qin H, Yuan Y, Yuan M, Yi S, Yang Y, Zhang Y. Tanshinone IIA ameliorates cisplatin-induced toxicology and cisplatin resistance via regulating SLC7A11 expression. Environ Toxicol. 2023. https://doi.org/10.1002/tox.24049.
Park E, Chung SW. ROS-mediated autophagy increases intracellular iron levels and ferroptosis by ferritin and transferrin receptor regulation. Cell Death Dis. 2019;10:822. https://doi.org/10.1038/s41419-019-2064-5.
Ursini F, Maiorino M. Lipid peroxidation and ferroptosis: the role of GSH and GPx4. Free Radic Biol Med. 2020;152:175–85. https://doi.org/10.1016/j.freeradbiomed.2020.02.027.
Zhang L, Liu W, Liu F, Wang Q, Song M, Yu Q, Tang K, Teng T, Wu D, Wang X, Han W, Li Y. IMCA induces ferroptosis mediated by SLC7A11 through the AMPK/mTOR pathway in colorectal cancer. Oxid Med Cell Longev. 2020;2020:1675613. "V体育平台登录" https://doi.org/10.1155/2020/1675613.
Sun D, Li YC, Zhang XY. Lidocaine promoted ferroptosis by targeting miR-382-5p /SLC7A11 axis in ovarian and breast cancer. Front Pharmacol. 2021;12: 681223. "V体育2025版" https://doi.org/10.3389/fphar.2021.681223.
Fang JY, Richardson BC. The MAPK signalling pathways and colorectal cancer. Lancet Oncol. 2005;6(5):322–7. https://doi.org/10.1016/S1470-2045(05)70168-6.
Zhao H, Ming T, Tang S, Ren S, Yang H, Liu M, Tao Q, Xu H. Wnt signaling in colorectal cancer: pathogenic role and therapeutic target. Mol Cancer. 2022;21:144. https://doi.org/10.1186/s12943-022-01616-7.
Yerlikaya A, Zeren S. Molecular pathways, targeted therapies, and proteomic investigations of colorectal cancer. Curr Mol Med. 2023;23:2–12. https://doi.org/10.2174/1566524022666211224120614.
Yu L, Wei J, Liu P. Attacking the PI3K/Akt/mTOR signaling pathway for targeted therapeutic treatment in human cancer. Semin Cancer Biol. 2022;85:69–94. https://doi.org/10.1016/j.semcancer.2021.06.019.
Narayanankutty A. PI3K/Akt/mTOR pathway as a therapeutic target for colorectal cancer: a review of preclinical and clinical evidence. Curr Drug Targets. 2019;20:1217–26. https://doi.org/10.2174/1389450120666190618123846.
Su H, Peng C, Liu Y. Regulation of ferroptosis by PI3K/Akt signaling pathway: a promising therapeutic axis in cancer. Front Cell Dev Biol. 2024;12:1372330. https://doi.org/10.3389/fcell.2024.1372330.
Yi J, Zhu J, Wu J, Thompson CB, Jiang X. Oncogenic activation of PI3K-AKT-mTOR signaling suppresses ferroptosis via SREBP-mediated lipogenesis. Proc Natl Acad Sci U S A. 2020;117(49):31189–97. https://doi.org/10.1073/pnas.2017152117.
Liu H, Liu C, Wang M, Sun D, Zhu P, Zhang P, Tan X, Shi G. Tanshinone IIA affects the malignant growth of Cholangiocarcinoma cells by inhibiting the PI3K-Akt-mTOR pathway. Sci Rep. 2021;11:19268. https://doi.org/10.1038/s41598-021-98948-z (VSports app下载).
Pan Y, Chen L, Li R, Liu Y, Nan M, Hou L. Tanshinone IIa induces autophagy and apoptosis via PI3K/Akt/mTOR axis in acute promyelocytic leukemia NB4 cells. Evid Based Complement Alternat Med. 2021;2021:3372403. https://doi.org/10.1155/2021/3372403.
Guan Z, Chen J, Li X, Dong N. Tanshinone IIA induces ferroptosis in gastric cancer cells through p53-mediated SLC7A11 down-regulation. Biosci Rep. 2020;40(8):BSR20201807. VSports在线直播 - https://doi.org/10.1042/BSR20201807.
Luo N, Zhang K, Li X, Hu Y, Guo L. Tanshinone IIA destabilizes SLC7A11 by regulating PIAS4-mediated SUMOylation of SLC7A11 through KDM1A, and promotes ferroptosis in breast cancer. J Adv Res. 2025;69:313–27. https://doi.org/10.1016/j.jare.2024.04.009 (VSports手机版).
Not applicable.
The present study was supported by 2021 Jiangsu Traditional Chinese Medicine Science and Technology Development Plan Project (grant no. MS2021065).
Department of Colorectal Surgery, The First People’s Hospital of Lianyungang, No. 6 Zhenhua Road, Haizhou District, Lianyungang, 222002, China
Tingrui Ge, Huazhuan Li, Ping Xiang, Dong Yang, Jingyi Zhou & Yonggang Zhang
Search author on:PubMed Google Scholar
Search author on:PubMed Google Scholar
Search author on:PubMed Google Scholar
Search author on:PubMed Google Scholar
Search author on:PubMed Google Scholar
Search author on:PubMed Google Scholar
Tingrui Ge contributed to Conceptualization, Data curation, Funding acquisition, Investigation, Methodology, Project administration, Writing – original draft, and Writing – review & editing. Huazhuan Li contributed to Data curation, Resources, and Supervision. Ping Xiang contributed to Methodology, Software, and Visualization. Dong Yang contributed to Software and Supervision. Jingyi Zhou contributed to Formal analysis and Validation. Yonggang Zhang contributed to Data curation, Supervision, and Writing – review & editing. All authors read and approved the final manuscript.
Correspondence to Yonggang Zhang.
This study is reported in accordance with ARRIVE guidelines. This study was approved by Ethics Committee of The First People's Hospital of Lianyungang.
Not applicable.
The authors declare no competing interests.
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Additional file 2: Supplementary Fig. 1. Effect of Tan IIA on normal human intestinal epithelial cellswas detected by MTT assay.
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
Ge, T., Li, H., Xiang, P. et al. Tanshinone IIA induces ferroptosis in colorectal cancer cells through the suppression of SLC7A11 expression via the PI3K/AKT/mTOR pathway. Eur J Med Res 30, 576 (2025). https://doi.org/10.1186/s40001-025-02842-7
Received:
Accepted:
Published:
DOI: https://doi.org/10.1186/s40001-025-02842-7
Anyone you share the following link with will be able to read this content:
Sorry, a shareable link is not currently available for this article.
Provided by the Springer Nature SharedIt content-sharing initiative
ISSN: 2047-783X