Open Access

Tanshinone IIA enhances the inhibitory effect of imatinib on proliferation and motility of acute leukemia cell line TIB‑152 in vivo and in vitro by inhibiting the PI3K/AKT/mTOR signaling pathway

Retraction in: /10.3892/or.2024.8725

  • Authors:
    • Zhi Teng
    • Shijuan Xu
    • Qin Lei
  • View Affiliations

  • Published online on: December 31, 2019     https://doi.org/10.3892/or.2019.7453
  • Pages: 503-515
  • Copyright: © Teng et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Acute lymphoblastic leukemia (ALL) is a malignant hematological disease. Tanshinone IIA (Tan IIA) has antitumor activity in vitro and in vivo. The aim of the present study was to investigate the effects of Tan IIA in combination with imatinib (IM) on the proliferation, apoptosis, migration and invasion of acute T lymphocytic leukemia TIB‑152 cells in vivo and in vitro, and analyze the potential underlying mechanism. Tan IIA and IM, alone and in combination, significantly inhibited proliferation, migration and invasion of TIB‑152 cells, and promoted apoptosis; the effect of co‑treatment with Tan IIA plus IM was enhanced. IGF‑1 promoted the proliferation, migration and invasion of TIB‑152 cells and inhibited apoptosis, while Tan IIA treatment significantly reversed these effects. In vivo experiments demonstrated that treatment with Tan IIA and IM, alone or in combination, significantly inhibited tumor growth in TIB‑152 xenograft mice; the growth inhibition of Tan IIA plus IM was the strongest observed. Western blot analysis revealed that the combination of Tan IIA and IM resulted in significantly lower levels of p‑PI3K, p‑AKT and p‑mTOR in cells and tissues compared with the IM and Tan alone treatment groups. In addition, the combination of Tan IIA and IM significantly decreased the levels of Ki67, cleaved caspase‑3, VEGF and MMP‑9 in cells and tissues, and the level of caspase‑3 was significantly increased. Taken together, the results revealed that Tan IIA enhanced the inhibitory effect of imatinib on TIB‑152 cell proliferation, migration and invasion, and induced apoptosis, which may be associated with inhibition of the PI3K/AKT/mTOR signaling pathway.

Introduction

Acute lymphoblastic leukemia (ALL) is a malignant hematological disease that originates in B- or T-line lymphoid progenitor cells. ALL can be divided into acute B lymphocytic leukemia (B-ALL) and acute T lymphocytic leukemia (T-ALL), of which T-ALL accounts for ~80% (1). ALL patients have a short remission period and a high recurrence rate, and the long-term survival rate of traditional chemotherapy is <20% (2). The emergence of tyrosine kinase inhibitors (TKIs), such as imatinib (IM) has improved the remission rate and reduced the recurrence rate of ALL patients (3). However, there are some limitations in the treatment of ALL with IM, such as drug resistance and side effects (4). In addition, to achieve complete remission and restore normal hematopoiesis of ALL, ≥4 drugs in combination therapy are often used as induction and remission treatment of high-risk or very high-risk children (<10 years) with ALL and in most young adults (24–30 years) with ALL (5). Rapidly and completely reducing the leukemic cell load prior to drug resistance development is considered to be the key to remission. Therefore, finding a better combination of chemotherapy drugs is crucial for the treatment of ALL.

In recent years, plant-derived compounds have become clinically useful anticancer drugs (68). Tanshinone IIA (Tan IIA) is an important active ingredient in Salvia miltiorrhiza and there have been reports on its antitumor effects. Studies confirmed that Tan IIA has antitumor activity in vitro and in vivo, and can induce differentiation, promote apoptosis and inhibit proliferation in various tumor cells (9,10), including leukemic cells (11). Tan IIA exerts a significant growth inhibitory effect on various leukemia cells, such as SUP-B15 (human Ph acute T lymphocytic leukemia cell line), K562 (chronic myeloid leukemia cell line), CEM (human leukemia cell line) and NB4 (acute promyelocytic leukemia cell beads) (12). Tan IIA treatment of KBM-5 cells (human chronic myeloid leukemia cells) can cause S-G1 phase arrest, DNA damage and caspase-3/9 activation in mitochondria. Tan IIA activates JNK and p38/MAPK, and induced apoptosis can be reversed by JNK inhibitors, suggesting that Tan IIA induces mitochondria-dependent apoptosis that is associated with activation of JNK (13). Tan IIA induces apoptosis in U937 acute myeloid leukemia cells possibly by activating pregnane X receptor, which in turn inhibits nuclear transcription factor (NF)-κB activity, resulting in the downregulation of Bcl-2 (14). In addition, Tan IIA works synergistically with other antitumor drugs. In the all-trans retinoic acid-resistant acute promyelocytic leukemia cell line MR2, the combination of Tan IIA and arsenic trioxide was found to enhance apoptosis and downregulate P-glycoprotein expression (15). These findings suggest that Tan IIA may be used as an adjunct to enhance the efficacy of chemotherapy drugs.

The PI3K/AKT/mTOR signaling pathway is a major signaling pathway involved in cell proliferation, apoptosis and metastasis, and its cascade reaction pathway occupies an important position in the signal transduction process (16). As a member of the lipid kinase family, PI3K is a heterodimer composed of two subunits, including a regulatory and a catalytic subunit (17). At rest, PI3K is ubiquitous in the cytosol. When cells are stimulated by growth factors, PI3K is activated and aggregates on the cell membrane, converting 3,4-diphosphophosphatidylinositol into 3,4,5-triphosphate phosphatidylinositol (PIP3), and then PIP3 can be used to activate downstream AKT. AKT is an evolutionarily highly conserved serine/threonine protein kinase that is mostly located in the cytosol at rest. Activated or phosphorylated (p-) PI3K generates PIP3 on the cell membrane. PIP3 interacts with the pleckstrin homology domain of AKT and phosphorylates the Thr308 and Ser473 sites via 3-phosphatidylinositol-dependent protein kinase 1. p-AKT translocates into the cytoplasm or nucleus, and regulates cell survival and apoptosis by regulating protein synthesis and gene transcription (18). mTOR is a target for rapamycin downstream of AKT in mammalian cells. p-AKT activates downstream mTOR signaling by direct phosphorylation of the Ser2448 of mTOR or by inhibition of nodular sclerosis complex (TSC)2 to form a complex with TSC1. After mTOR is activated, it phosphorylates elF4E-binding protein 1 and p70 ribosomal protein S6 kinase downstream, and initiates the synthesis of various proteins, including cyclin 1, hypoxia-inducible factor-1, and vascular endothelial growth factor (VEGF). Therefore, the PI3K/AKT/mTOR signaling pathway is closely associated with cell proliferation, differentiation, apoptosis and migration (19,20). A growing body of evidence suggests that abnormal activation of the PI3K/AKT/mTOR signaling pathway is important in the over-proliferation and apoptosis of tumor cells, and contributes to the treatment resistance of various cancers, including leukemia (20,21). Tumor treatment strategies for key molecular targets of this signaling pathway are urgently required.

The aim of the present study was to investigate the effects of Tan IIA combined with IM on the proliferation, apoptosis, migration and invasion of human acute T lymphocytic leukemia cells TIB-152. A tumor xenograft growth assay was also used to study the therapeutic effect of TAN IIA in combination with IM in vivo. In addition, we examined the changes in the PI3K/AKT/mTOR signaling pathway in cells and tissues after Tan IIA plus IM treatment and suggested a potential mechanism of action. The presented results may provide a basis for the development of novel therapeutic options for acute T-lymphocytic leukemia in the future.

Materials and methods

Cell lines

TIB-152, a human acute leukemia T cell line was obtained from the Cell Bank of Type Culture Collection of the Chinese Academy of Sciences. TBI-512 c was maintained in RPMI-1640 (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% fetal bovine serum (FBS), 100 U/ml penicillin and 100 mg/ml streptomycin (all from Gibco; Thermo Fisher Scientific, Inc.) and cultured in a humidified incubator at 37°C with 5% CO2.

A total of 20 female athymic BALB/c nude mice (age, 6–7 weeks) were obtained from Beijing Institute of Life Sciences of the Chinese Academy of Sciences and were bred in specific pathogen-free environment in the experimental animal center of the 215 Hospital of Shanxi Nuclear Industry. All animal procedures were approved by the Institutional Animal Care and Use Committee of the 215 Hospital of Shanxi Nuclear Industry and conformed to the guidelines of the US National Institutes of Health.

Drugs

IM was purchased from Cayman Chemical Company, formulated into a 1 µM stock solution with DMSO (Sigma- Aldrich; Merck KGaA) and stored at −20°C in the dark. Tan IIA (purity, ≥99.5%) was purchased from the Chinese National Institute for the Control of Pharmaceutical and Biological Products, prepared into a 20 mM stock solution with methanol (Sigma-Aldrich; Merck KGaA) and stored at −20°C in the dark.

Cytotoxicity assay

Cell viability was evaluated by CCK-8 assay (Beyotime Institute of Biotechnology) in triplicate. Cells (2×105) were cultured in 96-well plates and the medium was removed after 24 h. Cells were treated with Tan IIA at 0, 0.625, 1.25, 2.5, 5, 10, 20, 40, 60, 80, 100 and 120 µM or with IM at 0, 0.078, 0.156, 0.312, 0.625, 1.25, 2.5, 5, 10, 15, 20 and 25 µM for 48 h. At the end of the cultivation, 10 µl of CCK-8 solution was added to the wells and samples were incubated for 4 h at 37°C. The optical density (OD) was measured at 490 nm. The IC50 was determined from survival curves.

Cell proliferation assay

Cells in the logarithmic growth phase were tested and divided into 4 groups as follows: i) Conventional culture group (TIB-152), ii) 20 µM Tan IIA monotherapy group [Tan IIA (20 µM)], iii) 5 µM IM monotherapy group [IM (5 µM)], and iv) 20 µM Tan IIA combined with 5 µM IM group (IM + Tan). Cell proliferation was analyzed using the CCK-8 assay. Cells were cultured in 96-well plates at 5×103 cells/well in 100 µl growth medium. Following incubation overnight, Tan IIA (20 µM), IM (5 µM) and IM + Tan groups were established. The TIB-152 group was treated with 0.1% DMSO. For rescue experiments, TBI-512 cells were pre-treated with the PI3K pathway-specific activator IGF-1 (10 nM) 24 h before exposure to Tan IIA or/and IM. At 24, 48, 72 and 96 h of treatment, the culture medium was removed and replaced with 100 µl RPMI-1640 containing 10 µl CCK-8 solution. After incubation at 37°C for 1 h, the OD was measured at 490 nm. This assay was conducted in triplicate and six wells were used per condition per time.

Western blot analysis

Proteins were extracted from cells or tissues using ice-cold lysis buffer consisting of 1% Triton X-100, 1% deoxycholate and 0.1% SDS. The protein content of the cell or tissue lysates was determined using BCA protein quantification kit. Proteins (50 µg) were resolved on 12% SDS-PAGE gels and transferred to a nitrocellulose membrane. The membrane was blocked in PBS with 5% skimmed milk for 1 h at room temperature and then incubated overnight at 4°C with the primary antibodies against Ki67 (1:1,000; monoclonal; ZRB1007; Sigma-Aldrich; Merck KGaA), VEGF (1:500; monoclonal; SAB1402390; Sigma-Aldrich; Merck KGaA), MMP-9 (1:500; polyclonal; SAB4501896; Sigma-Aldrich; Merck KGaA), cleaved caspase-3 (1:1,000; polyclonal; sc-22140; Santa Cruz Biotechnology, Inc.), PI3K (1:500; monoclonal; SAB5300225; Sigma-Aldrich; Merck KGaA), p-PI3K (p85; 1:500; polyclonal; SAB4502195; Sigma-Aldrich; Merck KGaA), AKT (1:1,000; polyclonal; SAB4500796; Sigma-Aldrich; Merck KGaA), p-AKT (Ser473; 1:500; monoclonal; sc293125; Santa Cruz Biotechnology, Inc.), mTOR (1:1,000; polyclonal; sc517464; Santa Cruz Biotechnology, Inc.), p-mTOR (Ser2448; 1:1,000; monoclonal; sc293133; Santa Cruz Biotechnology, Inc.) or GAPDH (1:5,000; polyclonal; sc-20375; Santa Cruz Biotechnology, Inc.). The membranes were incubated with HRP-conjugated goat anti-rabbit IgG secondary antibody (1:5,000; polyclonal; sc-2922; Santa Cruz Biotechnology, Inc.) for 1 h at room temperature. Protein bands were visualized using an enhanced chemiluminescence kit (GE Healthcare). Images were analyzed using Quantity One version 4.4 (Bio-Rad Laboratories, Inc.).

Cell apoptosis assay

Cell apoptosis was detected by an Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) apoptosis detection kit (Nanjing KeyGen Biotech Co., Ltd.) according to the manufacturer's instructions. In brief, TIB-152 cells were seeded in 6-well plates (2×105 cells/well) with RPMI-1640 supplemented with 10% FBS. Tan IIA (20 µM), IM (5 µM) or IM (5 µM) + Tan (20 µM) was added and the cells were cultured for 24 h. The TIB-152 group was treated with 0.1% DMSO. For rescue experiments, TBI-512 cells were pre-treated with IGF-1 (10 nM) 24 h before exposure to Tan IIA or/and IM. After 24 h of incubation, the cells were collected, centrifuged at 1,000 × g for 5 min and then resuspended in 500 µl binding buffer (provided with kit), followed by the addition of Annexin V-FITC (5 µl) and PI (5 µl). The samples were then incubated in the dark at room temperature for 15 min. Cell apoptosis assay was performed within 1 h on a flow cytometer (FACSCalibur system) equipped with Cell Quest software (version 5.1; BD Biosciences). The total percentage of apoptotic cells was defined as the sum of early and late apoptotic cells (22).

Transwell assays

The invasion capacity was determined using Transwell chambers (8-µm pore size). TBI-512 cells (1×105) were cultured in 96-well plates with serum-free RPMI-1640 for 24 h, resuspended in fresh serum-free RPMI-1640 and placed in the upper chamber of the Matrigel-coated Transwell insert (5×104), with 0.1% DMSO, 20 µM Tan IIA, 5 µM IM or 5 µM IM plus 20 µM Tan. For rescue experiments, cells were pre-treated with IGF-1 (10 nM) 24 h before exposure to Tan IIA or/and IM. The lower chambers were filled with 500 µl RPMI-1640 containing 10% FBS. After 12 h, the surface of the membrane was scrubbed gently with a cotton swab and cells that invaded to the lower surface were fixed with 4% paraformaldehyde for 30 min at room temperature. Fixed cells were washed with PBS and stained with 0.1% crystal violet solution for 20 min at 37°C. Cells were then photographed and counted under a light microscope (magnification, ×200) and the mean number of invading cells was determined by counting five random fields of each well.

Wound healing assay

Migration capacities were determined using wound healing assays. TIB-152 cells were maintained in RPMI-1640 with 10% FBS. The treatment groups were established as described and cells were cultured for 48 h. For rescue experiments, TBI-512 cells were pre-treated with IGF-1 (10 nM) 24 h before exposure to Tan IIA or/and IM. Subsequently, cells (1×106) were seeded in 24-well plates and cultured in serum-free medium for 24 h. A wound was inflicted in the confluent cell layer using a 200-µl pipette tip and the wells were gently washed with PBS to remove all floating cells. Pictures of the wounds were captured at 0 and 24 h. Gap width was measured using Image-Pro Plus 6.0 (Media Cybernetics, Inc.).

Tumor xenograft growth assay in vivo

Female athymic BALB/c nude mice (age, 6–7 weeks; weight, 18–22 g) were housed under standard conditions (room temperature, 22°C; 12-h light/dark cycle) and controlled humidity (relative, 50%) with free access to food and water. TBI-152 cells (4×106) were injected into the right flank of the mice. Mice were randomized into four groups (n=5/group). Treatment commenced on the second day after inoculation. Mice in the three drug treatment groups were injected intraperitoneally with Tan IIA (50 mg/kg), IM (50 mg/kg) or IM + Tan, three times per week for 3 weeks (23). In the control group, the mice were injected intraperitoneally with saline. The mice were weighed weekly. Tumor volume was measured every 3 days using fine digital calipers and the volume of the tumor was calculated according to the following formula: Volume=length × width2 ×0.52. After completion of the drug treatment, the mice were sacrificed and tumors were photographed and stored at −80°C for further for analysis. All animal procedures were approved by the Institutional Animal Care and Use Committee of the 215 Hospital of Shanxi Nuclear Industry and followed the guidelines of the US National Institutes of Health.

TUNEL staining

Tumor tissues were fixed overnight at 4°C in 4% paraformaldehyde, embedded in paraffin and cut into 4-µm sections. TUNEL staining (Roche Diagnostics) was performed according to the manufacturer's instructions. Paraffin-embedded tissue sections were dewaxed in xylene for 5–10 min, followed by replacing with fresh xylene and dewaxing for a further 5–10 min. Subsequently, the sections were washed with absolute ethanol for 5 min, 90% ethanol for 2 min, 70% ethanol for 2 min, and distilled water for 2 min. All procedures were performed at 37°C. Subsequently, 20 µg/ml of DNase-free proteinase K was added dropwise to the sections and allowed to act at 37°C for 30 min. The coverslips were then placed in 4% paraformaldehyde for 30 min and immersed in 0.2% Triton X-100 for 15 min at room temperature. TdT reaction mix (100 µl) was added to the coverslips for 1 h at 37°C and coverslips were immersed in 2X SSC for 15 min at 37°C, 0.3% H2O2 for 30 min at 37°C and 100 µl streptavidin-HRP for 30 min at 37°C. Then, 100 µl DAB was added to the coverslips until a light brown background developed for 30 min at 37°C. The nuclei of apoptotic cells were stained dark brown. Images were analyzed using Image-Pro Plus 6.0 software (Media Cybernetics, Inc.). Sections were then photographed and counted under a light microscope (magnification, ×200), and the number of TUNEL-positive cells in 10 microscopic fields were counted. Apoptosis rate (%)=(apoptotic cells/total cells).

Immunohistochemical analysis

Paraffin-embedded 4-µm tumor tissue sections were used for immunohistochemical staining. The sections were dewaxed, placed in 3% H2O2 for 15 min at room temperature and washed 3 times with PBS. Then, the sections were incubated in 0.1 mol/l citrate buffer (pH 6.0) for 10 min at 100°C. The samples were probed with a primary antibody against Ki67 (1:100; SAB5300423; Sigma-Aldrich; Merck KGaA), cleaved caspase-3 (1:500; MAB4703, Sigma-Aldrich; Merck KGaA), VEGF (1:100; SAB1402390; Sigma-Aldrich; Merck KGaA) and MMP-9 (1:100; SAB5200294; Sigma-Aldrich; Merck KGaA) overnight at 4°C and then incubated with HRP-conjugated IgG (I-10677; 1:500; Invitrogen; Thermo Fisher Scientific, Inc.) at room temperature for 1 h. Positive cells were observed using a light microscope (magnification, ×200) and evaluation was performed using Photoshop CS6 (Adobe Systems, Inc.) and Image-Pro Plus 6.0 software (Media Cybernetics, Inc.).

Statistical analysis

Data were analyzed using SPSS 20.0 (IBM Corp.) and are expressed as the mean ± standard deviation representative of ≥3 independent experiments. Statistical differences between two groups were analyzed by unpaired Student's t-test and multiple comparisons were analyzed by two-way ANOVA followed by Tukey's test. P<0.05 was considered to indicate statistically significant differences.

Results

Cytotoxicity of Tan IIA and IM on TIB-152 cells

The structures of IM and Tan IIA are presented in Fig. 1A and B, respectively. To analyze the cytotoxic effects of Tan IIA and IM on TIB-152 cells, the cells we treated with varying concentrations of Tan IIA and IM, and cell viability was analyzed using CCK-8 assays 48 h later. CCK-8 assays demonstrated that IM at ≥0.625 µM (Fig. 1C) and Tan IIA at ≥10 µM (Fig. 1D) significantly suppressed cell viability. Tan IIA was determined to have an IC50 of 19.456±1.24 µM and the IC50 of IM was 4.922±0.36 µM. For subsequent experiments, 5 µM IM and 20 µM Tan IIA were considered as optimum concentrations.

Tan IIA enhances the effect of IM on the proliferation and apoptosis of TIB-152 cells

To analyze the effect of Tan IIA in combination with IM on the proliferation of TIB-152 cells, CCK-8 assays were performed (Fig. 2A). Cells treated with Tan IIA (20 µM) and IM (5 µM), alone or in combination, exhibited significant inhibition of cell proliferation in a dose-dependent manner compared with the TIB-152 group. From the growth curve, it was observed that the growth rate in the IM + Tan group was significantly lower compared with that in the IM (5 µM) and Tan IIA (20 µM) groups (Fig. 2A). To elucidate whether the decreased proliferation was due to the upregulation of cell apoptosis, an Annexin V/PI flow cytometry assay was performed and IM + Tan co-treatment was found to significantly increase the apoptosis rate compared with the IM (5 µM) and the TIB-152 groups at 48 h (Fig. 2B and C). The results demonstrated that, in TIB-152 cells, Tan IIA acted as a chemosensitizer for IM.

Ki67 is closely associated with and can be used as a reliable marker for cell proliferation (24). Caspase-3 is an important terminal cleavage enzyme in the process of apoptosis (25). To gain further insight into the molecular mechanism of how Tan IIA affects proliferation and apoptosis in conjunction with IM, associated protein expression was detected by western blotting. The results revealed that IM and Tan IIA treatment significantly decreased the levels of proliferation-associated and increased the levels of apoptosis-associated proteins compared with the TIB-152 group (Fig. 2H and I). Ki67 levels were further significantly decreased and cleaved caspase-3 levels were significantly increased in the IM + Tan group compared with those in the IM (5 µM) group.

Tan IIA enhances the inhibitory effect of IM on invasion and migration of TIB-152 cells

We investigated whether Tan IIA combined with IM affected the migration and invasion of TIB-152 cells in vitro. Transwell assays were performed to measure the invasive capability and wound healing assay to measure the migratory capability. As shown in Fig. 2D-G, IM + Tan significantly reduced the number of invading cells and the wound closure rate compared with the IM (5 µM) and TIB-152 groups.

Extracellular matrix degradation induced by MMPs is required for cell invasion and VEGF plays a key role in mediating cell migration (26,27). Therefore, western blotting was used to analyze the effects of Tan IIA and IM on the expression of VEGF and MMP-9. The results demonstrated that treatment with Tan IIA and IM, alone or in combination, significantly decreased the levels of VEGF and MMP-9 compared with the TIB-152 group, and IM + Tan significantly decreased protein levels compared with the IM (5 µM) group (Fig. 2H and J). These indicate that Tan IIA enhanced the inhibition of cell migration and invasion exerted by IM.

Tan IIA enhances the inhibitory effect of IM on the PI3K/AKT/mTOR signaling pathway in TIB-152 cells

To further evaluate the effects of IM + Tan, the expression of proteins associated with the PI3K/AKT/mTOR signaling pathway was analyzed by western blotting. We found that treatment with Tan IIA or IM significantly decreased the phosphorylation of PI3K, AKT and mTOR compared with the control (Fig. 3), while the total protein levels of PI3K, AKT and mTOR were not significantly affected. These results confirmed that Tan IIA and IM inhibited the activation of the PI3K/AKT/mTOR signaling pathway. Additionally, IM + Tan further significantly decreased the phosphorylated protein levels compared with the IM (% µM) group, suggesting that Tan IIA enhanced the inhibitory effect of IM on the PI3K/AKT/mTOR signaling pathway.

Tan IIA reverses the effect of PI3K pathway activator IGF-1 on the biological characteristics of TIB-152 cells

It was further investigated whether Tan IIA regulated the proliferation, apoptosis, migration and invasion of TIB-152 cells by inhibiting the activation of the PI3K/AKT/mTOR signaling pathway to enhance the anticancer effect of IM. Rescue experiments were performed by treating cells with the PI3K pathway activator IGF-1 prior to treatment with Tan IIA or/and IM. As shown in Fig. 4A and B, the levels of p-PI3K, p-AKT and p-mTOR in the IGF-1 (10 mM) group were significantly higher compared with those in the TIB-152 group, confirming that IGF-1 activated the PI3K/AKT/mTOR signaling pathway. IM treatment significantly inhibited the activation of PI3K/AKT/mTOR by IGF-1 and IM + Tan significantly enhanced the effect of IM, resulting in protein levels comparable to those of the TIB-152 group.

Further analysis of the biological characteristics of TIB-152 cells revealed that proliferation, migration and invasion in the Tan + IM + IGF-1 and IM + IGF-1 groups were significantly reduced compared with the IGF-1 group, and the apoptosis rate was significantly increased (Fig. 4). Furthermore, the results from the Tan + IM + IGF-1 group on cell biological performance were significantly better compared with the IM + IGF-1 group. Therefore, Tan IIA appears to enhance the effect of IM on the biological characteristics of TIB-152 cells by inhibiting PI3K/AKT/mTOR signaling pathway activation.

Tan IIA enhances the inhibitory effect of IM on TIB-152 tumor growth in xenograft mice

Next, tumor growth assays were used to study the antitumor activity of Tan IIA in combination with IM in vivo and to further elucidate its mechanism of action. Compared with the control group, the growth rate of transplanted tumors in the three treatment groups was slower and the tumor volume in the Tan + IM group increased the slowest (Fig. 5A and B). At day 21 after treatment, IM or Tan IIA treatment significantly decreased tumor growth compared with the control and Ta + IM further significantly decreased the observed tumor volume compared with the IM (50 mg/kg) group. The results indicated that Tan IIA enhanced the antitumor effect of IM on TIB-152 ×enograft mice.

To further explore the mechanism by which Tan IIA inhibited tumor growth in vivo, the expression of representative proteins was analyzed by TUNEL, immunohistochemistry and western blotting (Fig. 5C-L). It was observed that the protein levels of p-PI3K, p-AKT, p-mTOR, Ki67, VEGF and MMP-9 were significantly lower in the Tan IIA, IM and Tan + IM groups compared with the control, while cleaved caspase-3 levels were significantly elevated. These results were consistent with the results from the in vitro experiments.

Discussion

Tumor development and progression are the result of a combination of cell hyperproliferation and apoptotic pathways. Apoptosis and its role in tumorigenesis and treatment are attracting increasing attention, and drug therapy based on the mechanism of tumor cell apoptosis has made progress (28). Metastasis is the leading cause of death among cancer patients, and cell migration and invasion are hallmarks of cancer metastasis (29). In the present study, experiments were performed to investigate the effects of Tan II in combination with IM on these processes in vivo and in vitro, and it was observed that IM + Tan inhibited cell proliferation, migration and invasion, promoted apoptosis and inhibited tumor growth of a TIB-152 ×enograft in vivo compared with the control. The results suggested that Tan IIA synergistically enhanced the antitumor effect of IM. The effects of Tan IIA may be mediated through inhibition of PI3K/AKT/mTOR signaling pathway activation in vivo and in vitro.

Tan IIA is a diterpenequinone extracted from the root of Salvia miltiorrhiza (30). Due to its cardioprotective and antiatherosclerotic properties, Tan IIA has become a research hotspot in the field of cardiovascular and neurological diseases (31). It was previously demonstrated that Tan IIA has good antitumor properties in vivo and in vitro. It can inhibit the growth of various tumor cell lines, including gastric cancer, non-small-cell lung cancer, leukemia and prostate cancer (31,32). Shan et al (12) used MTT assays to investigate the effects of Tan IIA on the proliferation of human leukemia K562 cells. They observed that Tan IIA inhibited excessive cell proliferation and that the inhibitory effect was dose-dependent. Furthermore, they demonstrated that Tan IIA inhibited the proliferation of K562 cells by inhibiting the AKT/mTOR signaling pathway. Other studies reported that the combination of Tan IIA with a variety of commonly used chemotherapy drugs, such as cisplatin and 5-fluorouracil, can enhance the therapeutic effects of these drugs (33,34). Lv et al (35) reported that Tan IIA may act synergistically with 17-demethoxygeldanamycin to inhibit tumor growth by inducing cell cycle arrest and autophagy. As a TKI, IM is widely used in the treatment of leukemia, but comes with disadvantages that include toxic side effects and drug resistance (36). Combination therapy is commonly used in the clinical setting (37). Therefore, the present study used the acute T lymphocytic leukemia cell line TIB-152 to assess the effect of Tan IIA in combination with IM on cells at doses associated with fewer toxic side effects. We observed that Tan IIA and IM significantly inhibited the proliferation of TIB-152 cells and this effect was the strongest when the treatments were combined, suggesting a synergistic interaction. Caspase-3 is an important terminal cleavage enzyme in the process of apoptosis (25) and upregulation of caspase-3 activity promotes apoptosis and exerts antitumor effects (38). Tan IIA induces increased caspase-3 activity in the leukemia cell line THP-1, promoted apoptosis, and exerted a growth inhibitory effect on THP-1 cells, suggesting that it may be of value as a potential anti-leukemic agent (39). Consistently with these findings, the results of the present study demonstrated that Tan IIA in combination with IM significantly promoted the expression of caspase-3, indicating that Tan IIA may promote the activation of apoptotic protein kinases to inhibit the proliferation of tumor cells.

Zhang et al (40) found that Tan IIA plays an antitumor role by promoting apoptosis and inhibiting cell metastasis. MMP-induced degradation of the extracellular matrix is a hallmark of cell migration and invasion and it has been demonstrated that the metastatic ability of cancer cells can be regulated by MMPs (41). VEGF is a major regulator of tumor angiogenesis and invasion, and can enhance growth, invasion and metastasis of tumor cells by promoting the expression of MMP-9 in vascular endothelial cells (42). Increased expression of VEGF and MMP-9 in advanced leukemia, such as chronic myeloid leukemia, is closely associated with disease progression (43). In the present study, Tan IIA in combination with IM significantly reduced MMP-9 and VEGF protein expression, which may be associated with the inhibition of cell migration and invasion induced by Tan IIA.

Cell biological functions, such as cell proliferation, apoptosis and metastasis, are regulated by various signal transduction pathways in the cell. PI3K/AKT/mTOR is one of the most important signal transduction pathways and its abnormal activation is closely associated with the occurrence and development of malignant tumors (44). This pathway is recognized as a key point in tumor regulation and is an important target for anticancer drug selection (19,44). The PI3K/AKT/mTOR signaling pathway is abnormally activated in various tumors, such as leukemia, lymphoma and breast cancer (44). In leukemia, multiple targets in the PI3K/AKT/mTOR signaling pathway are abnormally activated and these constitute important targets for the development of therapeutic drugs (45,46). In the present study, it was observed that Tan IIA and IM reduced the phosphorylation of PI3K, AKT and mTOR in TBI-152 cells, and Tan IIA enhanced the inhibitory effect of IM on the PI3K/AKT/mTOR signaling pathway. Activation of the PI3K/AKT/mTOR signaling pathway directly regulates specific gene expression by affecting downstream protein kinases, such as apoptosis-associated cellular molecules, including caspase-3, caspase-9, Bcl-2 and Bax, cell cycle-associated factors, including cyclin D1 and E, and by participating in the regulation of cell proliferation and apoptosis (47,48). In addition, activation of the PI3K/AKT signaling pathway promotes VEGF synthesis (49) and VEGF binds to its receptor to feedback activation of PI3K. Activation of PI3K/AKT and VEGF further promote MMP expression and mediate tumor cell infiltration and metastasis (50). Tan IIA inhibits the PI3K/AKT signaling pathway, induces cytochrome c release, activates caspase-9 and −3, and induces apoptosis through the mitochondrial signaling pathway in gastric carcinoma cells and glioma cells (29,51). Tan IIA inhibits MMP-9 activity by inhibiting the AKT signaling pathway, thereby inhibiting human aortic smooth muscle cell migration and invasion (52). In order to confirm that the enhancement of the IM antitumor effect by Tan IIA in TIB-152 cells was mediated through the inhibition of the PI3K/AKT/mTOR signaling pathway, rescue experiments were performed. The results demonstrated that the proliferation, migration and invasion of TIB-152 cells were significantly increased after treatment with the PI3K-specific activator IGF-1 and the apoptosis rate was significantly reduced. IM + Tan treatment reversed these effects, confirming that the abnormal activation of PI3K/AKT/mTOR signaling may be associated with the progression of ALL.

Munagala et al (53) observed that intraperitoneal injection of Tan IIA inhibited the growth of cervical cancer xenograft in nude mice. Zhou et al (54) reported that the antitumor mechanism of Tan IIA inhibited the growth of human colon cancer subcutaneous xenograft in nude mice by downregulating the expression of VEGF. These studies previously confirmed the antitumor effect of Tan IIA in vivo. We also found that Tan IIA significantly inhibited the growth of TIB-152 ×enograft tumors, and significantly reduced the phosphorylation of PI3K, AKT and mTOR, and the expression of apoptosis-associated proteins in vivo. These results were consistent with the results of the in vitro experiments. The findings suggested that Tan IIA may be a PI3K/AKT/mTOR inhibitor and act synergistically with IM to exert antitumor effects. Moreover, the activation of the Lyn/PI3K/AKT signaling pathway exerts a protective effect on leukemia cells undergoing aggressive oxidative stress, indicating that, when the PI3K/AKT signaling pathway is inhibited, TIB-152 cells are more vulnerable to Tan IIA + IM treatment (55).

However, the pharmacological studies of Tan IIA remain at preclinical research stage and there is a limited number of clinical trials investigating complex disease pathogenesis. In addition, the process of tumor progression is extremely complex, involving multiple signaling cascades and cross talks of the PI3K/AKT/mTOR signaling pathway with other signaling pathways (5658). Therefore, the efficacy of Tan IIA in inhibiting ALL requires further investigation. In a follow-up study, it may be interesting to explore the intervention of Tan IIA on ALL cells in order to elucidate its antitumor mechanism of action.

In conclusion, it was demonstrated that the combination of Tan IIA and IM synergistically increased the apoptosis of TIB-152 cells and inhibited cell proliferation, migration and invasion. The potential mechanism may be through inhibiting the PI3K/AKT/mTOR signaling pathway in vivo and in vitro. These findings may provide a new basis for the clinical application of Tan IIA and IM in the treatment of ALL.

Acknowledgements

The authors would like to thank Dr ZhiTeng and Dr Qin Lei (The 215 Hospital of Shanxi Nuclear Industry, Shanxi, China) for providing technical support for the present study.

Funding

Not funding was received.

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

ZT analyzed and interpreted the principal data regarding cell functional analysis and western blotting. SX and QL were involved in the in vivo experiments and the statistical analysis. QL was responsible for the design of the study and drafting the manuscript. All authors have read and approved the final version of the manuscript.

Ethics approval and consent to participate

The animal experiments in the present study were approved by The Animal Care and Research Committee of the 215 Hospital of Shanxi Nuclear Industry (Shanxi, China). All experiments were performed in compliance with relevant laws and guidelines. All experiments were conducted following the institutional guidelines of the 215 Hospital of Shanxi Nuclear Industry.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Yang T, Yao S, Zhang X and Guo Y: Andrographolide inhibits growth of human T-cell acute lymphoblastic leukemia Jurkat cells by downregulation of PI3K/AKT and upregulation of p38 MAPK pathways. Drug Des Devel Ther. 10:1389–1397. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Mullighan CG, Phillips LA, Su X, Ma J, Miller CB, Shurtleff SA and Downing JR: Genomic analysis of the clonal origins of relapsed acute lymphoblastic leukemia. Science. 322:1377–1380. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Yilmaz M, Kantarjian H and Jabbour E: Treatment of acute lymphoblastic leukemia in older adults: Now and the future. Clin Adv Hematol Oncol. 15:266–274. 2017.PubMed/NCBI

4 

Fujimaki K, Hattori Y and Nakajima H: 10-year complete remission in a philadelphia chromosome-positive acute lymphoblastic leukemia patient using imatinib without high-intensity chemotherapy or allogeneic stem cell transplantation. Int J Hematol. 107:709–711. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Zhang SH, An FY, Xu JX, Kong LJ, He HL, Chai YH and Zhao WL: Clinical features and prognostic factors of children with acute lymphoblastic leukemia in high-risk group. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 25:365–370. 2017.(In Chinese). PubMed/NCBI

6 

Anand P, Sundaram C, Jhurani S, Kunnumakkara AB and Aggarwal BB: Curcumin and cancer: An ‘old-age’ disease with an ‘age-old’ solution. Cancer Lett. 267:133–164. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Ireson CR, Jones DJ, Orr S, Coughtrie MW, Boocock DJ, Williams ML, Farmer PB, Steward WF and Gescher AJ: Metabolism of the cancer chemopreventive agent curcumin in human and rat intestine. Cancer Epidemiol Biomarkers Prev. 11:105–111. 2002.PubMed/NCBI

8 

Tabernero J, Kunzmann V, Scheithauer W, Reni M, Shiansong Li J, Ferrara S and Djazouli K: Nab-paclitaxel plus gemcitabine for metastatic pancreatic cancer: A subgroup analysis of the Western European cohort of the MPACT trial. Onco Targets Ther. 10:591–596. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Zhang RW, Liu ZG, Xie Y, Wang LX, Li MC and Sun X: In vitro inhibition of invasion and metastasis in colon cancer cells by TanIIA. Genet Mol Res. 15:2016. View Article : Google Scholar :

10 

Zu Y, Wang J, Ping W and Sun W: Tan IIA inhibits H1299 cell viability through the MDM4-IAP3 signaling pathway. Mol Med Rep. 17:2384–2392. 2018.PubMed/NCBI

11 

Munagala R, Aqil F, Jeyabalan J, Vadhanam M and Gupta R: Increased anti-tumor activity by novel systemic delivery and molecular targets of tanshinone II A. Cancer Res. 70:56902011.

12 

Shan QQ, Gong YP, Guo Y, Lin J, Zhou RQ and Yang X: Anti-tumor effect of tanshinone IIA, tetrandrine, honokiol, curcumin, oridonin and paeonol on leukemia cell lines. Sichuan Da Xue Xue Bao Yi Xue Ban. 43:362–366. 2012.(In Chinese). PubMed/NCBI

13 

Yun SM, Jeong SJ, Kim JH, Jung JH, Lee HJ, Sohn EJ, Lee MH and Kim SH: Activation of c-Jun N-terminal kinase mediates tanshinone IIA-induced apoptosis in KBM-5 chronic myeloid leukemia cells. Biol Pharm Bull. 36:208–214. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Liu C, Li J, Wang L, Wu F, Huang L, Xu Y, Ye J, Xiao B, Meng F, Chen S and Yang M: Analysis of tanshinone IIA induced cellular apoptosis in leukemia cells by genome-wide expression profiling. BMC Complement Altern Med. 12:52012. View Article : Google Scholar : PubMed/NCBI

15 

Li L, Zhang ZH and Zhao WD: Apoptosis of MR2 cells induced by tanshinone II A combined with arsenic trioxide. Sichuan Da Xue Xue Bao Yi Xue Ban. 40:812–816. 2009.(In Chinese). PubMed/NCBI

16 

Keppler-Noreuil KM, Parker VE, Darling TN and Martinez-Agosto JA: Somatic overgrowth disorders of the PI3K/AKT/mTOR pathway & therapeutic strategies. Am J Med Genet C Semin Med Genet. 172:402–421. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Goda S, Kaneshita Y, Inoue H, Domae E, Ikeo T, Iida J and Domae N: Enamel matrix derivative protein stimulated wound healing via phosphoinositide 3-kinase. J Periodontol. 80:1631–1637. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Molgaard S, Ulrichsen M, Olsen D and Glerup S: Detection of phosphorylated Akt and MAPK in cell culture assays. MethodsX. 3:386–398. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Zhang Y, Kwok-Shing Ng P, Kucherlapati M, Chen F, Liu Y, Tsang YH, de Velasco G, Jeong KJ, Akbani R, Hadjipanayis A, et al: A pan-cancer proteogenomic atlas of PI3K/AKT/mTOR pathway alterations. Cancer Cell. 31:820–832.e3. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Costa RLB, Han HS and Gradishar WJ: Targeting the PI3K/AKT/mTOR pathway in triple-negative breast cancer: A review. Breast Cancer Res Treat. 169:397–406. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Park S, Chapuis N, Tamburini J, Bardet V, Cornillet-Lefebvre P, Willems L, Green A, Mayeux P, Lacombe C and Bouscary D: Role of the PI3K/AKT and mTOR signaling pathways in acute myeloid leukemia. Haematologica. 95:819–828. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Robles-Escajeda E, Das U, Ortega NM, Parra K, Francia G, Dimmock JR, Varela-Ramirez A and Aguilera RJ: A novel curcumin-like dienone induces apoptosis in triple-negative breast cancer cells. Cell Oncol (Dordr). 39:265–277. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Jiang P, Chen M, Lu J, Chen C and Jiao BH: Effect of tanshinone IIA on MMP-2 and iNOS expression and free radical release in hippocampus of rat Alzheimer's disease model. Acad J Second Mil Med Univ. 30:380–384. 2010. View Article : Google Scholar

24 

Petrelli F, Viale G, Cabiddu M and Barni S: Prognostic value of different cut-off levels of Ki-67 in breast cancer: A systematic review and meta-analysis of 64,196 patients. Breast Cancer Res Treat. 153:477–491. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Mirzayans R, Andrais B, Kumar P and Murray D: The growing complexity of cancer cell response to DNA-damaging agents: Caspase 3 mediates cell death or survival? Int J Mol Sci. 17:E7082016. View Article : Google Scholar : PubMed/NCBI

26 

Castro MG, Campos LE, Rodriguez YI and Alvarez SE: In vitro methods to study the modulation of migration and invasion by sphingosine-1-phosphate. Methods Mol Biol. 1697:117–131. 2018. View Article : Google Scholar : PubMed/NCBI

27 

Evans IM, Kennedy SA, Paliashvili K, Santra T, Yamaji M, Lovering RC, Britton G, Frankel P, Kolch W and Zachary IC: Vascular endothelial growth factor (VEGF) promotes assembly of the p130Cas interactome to drive endothelial chemotactic signaling and angiogenesis. Mol Cell Proteomics. 16:168–180. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Gregory CD: Apoptosis in cancer pathogenesis and anti-cancer therapy. New Perspectives and Opportunities. Springer International Publishing; Cham: pp. p2472016

29 

Su CC and Chiu TL: Tanshinone IIA decreases the protein expression of EGFR, and IGFR blocking the PI3K/Akt/mTOR pathway in gastric carcinoma AGS cells both in vitro and in vivo. Oncol Rep. 36:1173–1179. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Zhang S, Liu Q, Luo H, Chen P, Wu X, Yang M, Kong W and Guo W: UFLC-MS/MS analysis of four tanshinone components in Salvia miltiorrhizae after ultrasound-assisted extraction. J Chromatogr B Analyt Technol Biomed Life Sci. 1017-1018:204–210. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Buenafe OE, Orellana-Paucar A, Maes J, Huang H, Ying X, De Borggraeve W, Crawford AD, Luyten W, Esguerra CV and de Witte P: Tanshinone IIA exhibits anticonvulsant activity in zebrafish and mouse seizure models. ACS Chem Neurosci. 4:1479–1487. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Gong Y, Li Y, Abdolmaleky HM, Li L and Zhou JR: Tanshinones inhibit the growth of breast cancer cells through epigenetic modification of aurora A expression and function. PLoS One. 7:e336562012. View Article : Google Scholar : PubMed/NCBI

33 

Su CC: Tanshinone IIA potentiates the efficacy of 5-FU in Colo205 colon cancer cells in vivo through downregulation of P-gp and LC3-II. Exp Ther Med. 3:555–559. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Chang TW, Lin CY, Tzeng YJ and Lur HS: Synergistic combinations of tanshinone IIA and trans-resveratrol toward cisplatin-comparable cytotoxicity in HepG2 human hepatocellular carcinoma cells. Anticancer Res. 34:5473–5480. 2014.PubMed/NCBI

35 

Lv C, Zeng HW, Wang JX, Yuan X, Zhang C, Fang T, Yang PM, Wu T, Zhou YD, Nagle DG and Zhang WD: The antitumor natural product tanshinone IIA inhibits protein kinase C and acts synergistically with 17-AAG. Cell Death Dis. 9:1652018. View Article : Google Scholar : PubMed/NCBI

36 

Hochhaus A and La Rosée P: Imatinib therapy in chronic myelogenous leukemia: Strategies to avoid and overcome resistance. Leukemia. 18:1321–1331. 2004. View Article : Google Scholar : PubMed/NCBI

37 

Jabbour E, Cortes J and Kantarjian H: Treatment selection after imatinib resistance in chronic myeloid leukemia. Target Oncol. 4:3–10. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Pu X, Storr SJ, Zhang Y, Rakha EA, Green AR, Ellis IO and Martin SG: Caspase-3 and caspase-8 expression in breast cancer: Caspase-3 is associated with survival. Apoptosis. 22:357–368. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Liu JJ, Zhang Y, Lin DJ and Xiao RZ: Tanshinone IIA inhibits leukemia THP-1 cell growth by induction of apoptosis. Oncol Rep. 21:1075–1781. 2009.PubMed/NCBI

40 

Zhang Y, Wei RX, Zhu XB, Cai L, Jin W and Hu H: Tanshinone IIA induces apoptosis and inhibits the proliferation, migration, and invasion of the osteosarcoma MG-63 cell line in vitro. Anticancer Drugs. 23:212–219. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Rizwan A, Cheng M, Krishnamachary B, Jiang L, Bhujwalla Z and Kristine G: Cancer cell adhesion and degradome interact to metastasize. Cancer Res. 74:31642014.

42 

Shi YL, Xu T, Li LP and Chen XP: Over-expression of VEGF and MMP-9 in Residual Tumor Cells of Hepatocellular Carcinoma after Embolization with Lipidol. J Huazhong Univ Sci Technolog Med Sci. 33:90–95. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Yang L, Dong ZR, Wen SP, Pan L, Zhang XJ, Luo JM and Xu SR: Relationship between VEGF and MMP-2, MMP-9 in 82 patients with acute myeloid leukemia. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 14:15–20. 2006.(In Chinese). PubMed/NCBI

44 

Lonetti A, Cappellini A, Bertaina A, Locatelli F, Pession A, Buontempo F, Evangelisti C, Evangelisti C, Orsini E, Zambonin L, et al: Improving nelarabine efficacy in T cell acute lymphoblastic leukemia by targeting aberrant PI3K/AKT/mTOR signaling pathway. J Hematol Oncol. 9:1142016. View Article : Google Scholar : PubMed/NCBI

45 

Lin M: Experimental study of atorvastatin regulation of HL-60 leukemia cell apoptosis through PI3K/AKT/mTOR. J Hainan Med Univ. 22:9–12. 2016.

46 

Tabe Y, Tafuri A, Sekihara K, Yang H and Konopleva M: Inhibition of mTOR kinase as a therapeutic target for acute myeloid leukemia. Expert Opin Ther Targets. 21:705–714. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Jiang S, Wang Q, Feng M, Li J, Guan Z, An D, Dong M, Peng Y, Kuerban K and Ye L: C2-ceramide enhances sorafenib-induced caspase-dependent apoptosis via PI3K/AKT/mTOR and Erk signaling pathways in HCC cells. Appl Microbiol Biotechnol. 101:1535–1546. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Reddy EP, Divakar SA, Reddy MVR, Cosenza SC, Baker SJ, Akula B and Parekh S: Abstract 4519: Targeting of cyclin D/Rb/E2F and PI3K/AKT/MTOR pathways with ON 123300 as a therapeutic strategy for mantle cell lymphoma. Cancer Res. 74:45192014.

49 

Di J, Gao K, Qu D, Yang J and Zheng J: Rap2B promotes angiogenesis via PI3K/AKT/VEGF signaling pathway in human renal cell carcinoma. Tumour Biol. 39:10104283177016532017. View Article : Google Scholar : PubMed/NCBI

50 

Zhou H, Wu J, Wang T, Zhang X and Liu D: CXCL10/CXCR3 axis promotes the invasion of gastric cancer via PI3K/AKT pathway-dependent MMPs production. Biomed Pharmacother. 82:479–488. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Ding L, Wang S, Wang W, Lv P, Zhao D, Chen F, Meng T, Dong L and Qi L: Tanshinone IIA affects autophagy and apoptosis of glioma cells by inhibiting phosphatidylinositol 3-Kinase/Akt/Mammalian target of rapamycin signaling pathway. Pharmacology. 99:188–195. 2017. View Article : Google Scholar : PubMed/NCBI

52 

Jin UH, Suh SJ, Chang HW, Son JK, Lee SH, Son KH, Chang YC and Kim CH: Tanshinone IIA from Salvia miltiorrhiza BUNGE inhibits human aortic smooth muscle cell migration and MMP-9 activity through AKT signaling pathway. J Cell Biochem. 104:15–26. 2008. View Article : Google Scholar : PubMed/NCBI

53 

Munagala R, Aqil F, Jeyabalan J and Gupta RC: Tanshinone IIA inhibits viral oncogene expression leading to apoptosis and inhibition of cervical cancer. Cancer Lett. 356:536–546. 2015. View Article : Google Scholar : PubMed/NCBI

54 

Zhou LH, Hu Q, Sui H, Ci SJ, Wang Y, Liu X, Liu NN, Yin PH, Qin JM and Li Q: Tanshinone II-a inhibits angiogenesis through down regulation of COX-2 in human colorectal cancer. Asian Pac J Cancer Prev. 13:4453–4458. 2012. View Article : Google Scholar : PubMed/NCBI

55 

Ruiz-Medina BE, Lerma D, Hwang M, Ross JA, Skouta R, Aguilera RJ, Kirken RA, Varela-Ramirez A and Robles-Escajeda E: Green barley mitigates cytotoxicity in human lymphocytes undergoing aggressive oxidative stress, via activation of both the Lyn/PI3K/Akt and MAPK/ERK pathways. Sci Rep. 9:60052018. View Article : Google Scholar

56 

Xu Y, Li N, Xiang R and Sun P: Emerging roles of the p38 MAPK and PI3K/AKT/mTOR pathways in oncogene-induced senescence. Trends Biochem Sci. 39:268–276. 2014. View Article : Google Scholar : PubMed/NCBI

57 

Watson AL, Anderson LK, Greeley AD, Keng VW, Rahrmann EP, Halfond AL, Powell NM, Collins MH, Rizvi T, Moertel CL, et al: Co-targeting the MAPK and PI3K/AKT/mTOR pathways in two genetically engineered mouse models of schwann cell tumors reduces tumor grade and multiplicity. Oncotarget. 5:1502–1514. 2014. View Article : Google Scholar : PubMed/NCBI

58 

Saini KS, Loi S, de Azambuja E, Metzger-Filho O, Saini ML, Ignatiadis M, Dancey JE and Piccart-Gebhart MJ: Targeting the PI3K/AKT/mTOR and Raf/MEK/ERK pathways in the treatment of breast cancer. Cancer Treat Rev. 39:935–946. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2020
Volume 43 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Teng Z, Xu S and Lei Q: Tanshinone IIA enhances the inhibitory effect of imatinib on proliferation and motility of acute leukemia cell line TIB‑152 in vivo and in vitro by inhibiting the PI3K/AKT/mTOR signaling pathway Retraction in /10.3892/or.2024.8725. Oncol Rep 43: 503-515, 2020
APA
Teng, Z., Xu, S., & Lei, Q. (2020). Tanshinone IIA enhances the inhibitory effect of imatinib on proliferation and motility of acute leukemia cell line TIB‑152 in vivo and in vitro by inhibiting the PI3K/AKT/mTOR signaling pathway Retraction in /10.3892/or.2024.8725. Oncology Reports, 43, 503-515. https://doi.org/10.3892/or.2019.7453
MLA
Teng, Z., Xu, S., Lei, Q."Tanshinone IIA enhances the inhibitory effect of imatinib on proliferation and motility of acute leukemia cell line TIB‑152 in vivo and in vitro by inhibiting the PI3K/AKT/mTOR signaling pathway Retraction in /10.3892/or.2024.8725". Oncology Reports 43.2 (2020): 503-515.
Chicago
Teng, Z., Xu, S., Lei, Q."Tanshinone IIA enhances the inhibitory effect of imatinib on proliferation and motility of acute leukemia cell line TIB‑152 in vivo and in vitro by inhibiting the PI3K/AKT/mTOR signaling pathway Retraction in /10.3892/or.2024.8725". Oncology Reports 43, no. 2 (2020): 503-515. https://doi.org/10.3892/or.2019.7453