Inhibition of Forkhead box protein M1 by thiostrepton increases chemosensitivity to doxorubicin in T-cell acute lymphoblastic leukemia

  • Authors:
    • Jian‑Yong Wang
    • Xiu‑Hong Jia
    • Hai‑Yan Xing
    • You‑Jie Li
    • Wen‑Wen Fan
    • Na Li
    • Shu‑Yang Xie
  • View Affiliations

  • Published online on: March 11, 2015     https://doi.org/10.3892/mmr.2015.3469
  • Pages: 1457-1464
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive type of blood malignancy, deriving from T-cell progenitors in the thymus, and comprises 10‑15% of pediatric and 25% of adult primary ALL cases. Despite advances, 20% of pediatric and the majority of adult patients with T-ALL succumb to mortality from resistant or relapsed disease, and the survival rate for patients with resistant or relapsed T-ALL remains poor. Alterations in the expression of Forkhead box protein M1 (FoxM1) have been detected in several types of cancer, and the inhibition of FoxM1 has been investigated as therapeutic strategy in cancer. The present study investigated the effects of the inhibition of FoxM1 by thiostrepton in human T-ALL Jurkat cells. The cells were treated with different concentrations of thiostrepton, either alone or in combination with doxorubicin. Cell viability was measured using CCK-8 assays and the cell cycle distribution, apoptosis and cell-associated mean fluorescence intensity of intracellular doxorubicin were assessed using flow cytometric analysis. The mRNA and protein expression levels were detected by reverse transcription‑quantitative polymerase chain reaction and western blot analyses. The inhibition of FoxM1 by thiostrepton significantly decreased the proliferation of the Jurkat cells proliferation in a time- and dose-dependent manner. Cell arrest at the G2/M phase, and apoptosis was significantly increased in the thiostrepton-treated Jurkat cells. Thiostrepton reduced the half maximal inhibitory concentration of doxorubicin in the Jurkat cells, and significantly enhanced the cytotoxicity of doxorubicin within the Jurkat cells by enhancing doxorubicin-induced apoptosis and increasing the accumulation of intracellular doxorubicin. Furthermore, the inhibition of FoxM1 by thiostrepton enhanced doxorubicin-induced apoptosis, possibly through a caspase-3-dependent pathway, and increased the accumuation of intracellular doxorubicin, possibly through downregulating the expression of glutathione S-transferase pi. Collectively, the results of the present study suggested that targeting FoxM1 with thiostrepton resulted in potent antileukemia activity and chemosensitizing effects in human T-ALL Jurkat cells.

Introduction

Acute lymphoblastic leukemia (ALL) occurs most frequently in children, with incidence peaks between the ages of 2 and 5 years, and is one of the most common childhood malignancies worldwide (1,2). T-cell ALL (T-ALL) is an aggressive type of blood malignancy, deriving from T-cell progenitors in the thymus, and comprises 10–15% of pediatric and 25% of adult primary ALL cases (3). The age of the patient at diagnosis, leukocyte count, ethnicity, gender and immunophenotype are clinical prognostic parameters, which are utilized to classify the patients into different risk groups (1). T-ALL is classified as a high-risk group in ALL (2). With the currently used intensive multi-agent chemotherapeutic procedures, the 5-year event-free survival (EFS) of children with T-ALL is 70–75%, whereas the EFS is 30–40% for adults <60 years of age, and 10% in those >60 years old (4). However, these types of therapy are highly toxic, and cases of relapse often develop resistance to chemotherapy, with a poor prognosis (2). Targeting leukemia-specific molecular abnormalities may further improve outcomes and reduce the adverse side effects of therapy.

Forkhead box protein M1 (FoxM1), characterized by a 100 amino acid winged-helix DNA binding domain, is a newly unified family member of the Forkhead transcription factors (5). The protein expression of FoxM1 is usually restricted to proliferating cells and is absent in quiescent or terminally differentiated cells, however, elevated expression levels of FoxM1 have been detected in a wide range of different types of human tumor and has been implicated in cellular transformation, tumor initiation and progression (6). By regulating the G1/S and G2/M transitions and M phase progression, antagonizing cellular senescence, stimulating stem cell-like characteristics (including self-renewal), promoting multiple steps of cancer progression by inducing mitogenic and survival signals, and promoting tumor invasion, migration, and angiogenesis, FoxM1 is important in tumorigenesis and cancer progression (6). Accumulating evidence has demonstrated that increased expression levels of FoxM1 are associated with a poor prognosis in various types of cancer (711), including acute myeloid leukemia (AML) (12). In addition, FoxM1 deregulation has also been associated with the development of cancer drug resistance (13). A previous study confirmed that suppression of FoxM1 enhanced the chemosen-sitivity of various types of cancer cell to the DNA-damaging reagent, doxorubicin (14). Although a number of investigations have examined the role of FoxM1 in AML (12,1517), the role of FoxM1 in ALL, particularly pediatric ALL, remains to be elucidated.

Thiostrepton, a natural product originally isolated from Streptomyces azureus, has received significant attention due to its potent anticancer activity as a FoxM1 inhibitor (18,19). Thiostrepton can downregulate the mRNA and protein expression levels of FoxM1 (20). The mechanism by which thiostrepton affects FoxM1 has been confirmed. Thiostrepton hinders the proteasomal degradation of a negative regulator of FoxM1 (NRFM), which either directly or indirectly inhibits the activity of FoxM1 as a transcription factor (21). Notably, thiostrepton appears to exert minimal toxicity against non-malignant cells (22). Thiostrepton has exhibited anticancer activity in rodent xenograft models, without observable toxicity (23,24). Collectively, these data suggest that thiostrepton is a suitable treatment for T-ALL, particularly in children. In the present study, the antileukemia effects of thiostrepton were examined in human T-ALL Jurkat cells. Crucially, the ability of thiostrepton to sensitize Jurkat cells to doxorubicin, which is commonly used for the treatment of ALL, was assessed.

Materials and methods

Chemicals and reagents

Thiostrepton (off-white powder, 1 g), purchased from Merck Millipore (Darmstadt, Germany), was freshly dissolved in dimethyl sulfoxide (DMSO; Sigma-Aldrich, St. Louis, MO, USA) to produce a 20 mmol/l stock solution. Doxorubicin (Haizheng Medicine Co. Ltd., Zhengjiang, China) was dissolved in a stock concentration of 10 mmol/l with ddH2O and divided into five aliquots. Rabbit monoclonal antibody against FoxM1 (cat. no. 5436) was obtained from Cell Signaling Technology, Inc. (Danvers, MA, USA). Rabbit polyclonal antibodies against glutathione S-transferase pi (GSTpi; cat. no. PB0144), survivin (cat. no. BA4055–2), caspase-3 (cat. no. BA2885–2) and β-actin (cat. no. BA2305) were obtained from Boster Biological Technology, Ltd. (Wuhan, China).

Cell lines and cell culture

The human T-ALL Jurkat cells (Key Laboratory of Tumour Molecular Biology of Binzhou Medical University (Binzhou, China) were cultured in RPMI-1640 medium (HyClone Laboratories, Inc., Logan, UT, USA), supplemented with 10% heat-inactivated fetal bovine serum (FBS) (GE Healthcare Bio-Sciences, Logan, UT, USA) at 37°C in 5% CO2.

Cell viability assay

Cell viability was assessed using a Cell Counting Kit-8 (CCK-8; Dojindo Molecular Technologies, Inc., Shanghai, China), according to the manufacturer’s instructions. The Jurkat cells were seeded into 96-well flat plates (1.5×104 cells/well) and then treated with or without various concentrations of thiostrepton (0, 1, 2 or 3 μm/l) for 24, 48 or 72 h at 37°C. The cells in the control group were treated with the equivalent quantity of dimethyl sulfoxide, at a final concentration of 0.02%. The CCK-8 kit was then used, according to the manufacturer’s instructions. The optical density (OD) was measured at 450 nm using a fluorescence spectrofluorometer (F-7000; Hitachi High-Technologies Corporation, Tokyo, Japan), and the reference OD was subtracted. RPMI-1640 containing 10% FBS with 0.02% DMSO served as the reference group. Cell viability was then calculated as follows: Cell viability = (ODsample - ODreference) / (ODcontrol - ODreference) × 100%. The half maximal inhibitory concentration of cells (IC50) was obtained using a probit regression analysis method (25). Each experiment was performed in triplicate and repeated three times.

Cell cycle analysis

The Jurkat cells were seeded into 6-well plates in RPMI-1640, containing 10% fetal bovine serum (FBS), with or without the agents, for 24 h at 37°C. The cells were harvested and fixed with 500 μl 70% cold (−20°C) ethanol (Shenzhen Kingstone Experimental Equipment Co., Ltd., Shenzhen, China) at 4°C overnight, and then incubated with RNase A (KeyGen Biotech Co., Ltd., Nanjing, China) for 30 min at 37°C, followed by incubation with 100 μg/ml propidium iodide (PI; KeyGen Biotech Co., Ltd.) at room temperature for 30 min. The cell cycle distribution was detected using a flow cytometer (FACS FC500; Beckman Coulter, Brea, CA, USA).

Detection of apoptosis

The Jurkat cells (2.5×105 cells/ml) were seeded into 6-well plates in RPMI-1640 containing 10% FBS, with or without the agents, for 24 h. Tells were collected and washed twice with phosphate-buffered saline (PBS), suspended in 500 μl binding buffer (KeyGen Biotech Co., Ltd.), 5 μl annexin V-fluorescein isothiocyanate and 5 μl PI (KeyGen Biotech Co., Ltd.) for 15 min in the dark at room temperature. The apoptotic cells were analyzed using a flow cytometer.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was isolated from the cells using TRIzol reagent (Invitrogen Life Technologies, Carlsbad, CA, USA). RT into complementary DNA (cDNA) was performed using a PrimeScript™ RT reagent kit with gDNA eraser (Takara Bio, Inc., Otsu, Japan). qPCR was performed using a SYBR® Premix Ex Taq™ kit (Takara Bio, Inc.) on an ABI PRISM 7500 real-time PCR system (Applied Biosystems, Foster City, CA, USA). The cycling parameters were as follows: 95°C for 30 sec, then 40 cycles of 95°C for 5 sec, 60°C for 34 sec. The isolation of RNA, RT and qPCR were performed, according to the manufacturer’s instructions. The primers used (Table I) were designed using Primer 5 version 5.6.0 software and synthesized by Sangon Biotech Co, Ltd. (Shanghai, China). GAPDH was used as an internal control. qPCR for each gene of each cDNA sample was assayed in triplicate. The results were calculated using the 2−∆∆Ct method. The following equations were used: ΔCt = Ct(target gene) - Ct(actin); ΔΔCt = ΔCt(thiostrepton-treated cells) - ΔCt(untreated control).

Table I

Primers used in reverse transcription-quantitative polymerase chain reaction.

Table I

Primers used in reverse transcription-quantitative polymerase chain reaction.

GenePrimer sequenceProduct length (bp)
FoxM1F: 5′-AGGAAGTGGCAGAGTCCAAC-3′
R: 5′-TGCTGTGATGATGCTGTGAA-3′
128
GSTpiF: 5′-CACTCAAAGCCTCCTGCCTA-3′
R: 5′-TGCTGGTCCTTCCCATAGAG-3′
127
GAPDHF: 5′-TGGTATCGTGGAAGGACTCA-3′
R: 5′-CAGTAGAGGCAGGGATGATG-3′
131

[i] FoxM1, forkhead box protein M1; GSTpi, glutathione S-transferase pi; F, forward; R, reverse.

Western blot analysis

The ells were lysed in 100 μl radioimmunoprecipitation assay buffer (Beyotime Biotechnology, Shanghai, China)., supplemented with 1 μl phenylmeth-ylsulfonyl fluoride (Beyotime Biotechnology), and the protein concentration of the lysate was determined using a Bicinchoninic Acid Protein Assay kit (Beyotime Biotechnology). The proteins (50 μg/lane) were separated by 8–12% SDS-PAGE (Beyotime Biotechnology) and transferred to polyvinylidine difluoride membranes (EMD Millipore, Bedford, MA, USA). The membranes were blocked with 5% skimmed milk for 2 h, and then incubated overnight at 4°C with specific antibodies. The primary antibodies used were rabbit monoclonal antibody against FoxM1 (1:1,000), and rabbit polyclonal antibodies against GSTpi (1:200), survivin (1:200), caspase-3 (1:200) and β-actin (1:1,000). The following day, the membrane was incubated in horesradish peroxidase-labeled goat anti-rabbit immunoglobulin G (1:5,000; cat. no. ZB-5301; Beijing ZhongShan-Golden Bridge Technology Co., Ltd., Beijing, China) for 1 h at room temperature. Finally, images were captured using a FluorChem FC2 gel imaging system (Alpha Innotech, San Leandro, CA, USA).

Flow cytometric analysis of intracellular doxorubicin accumulation

The intracellular accumulation of doxorubicin was measured using a standard fluorescent method (26). The Jurkat cells were cultured in RPMI-1640 containing 10 FBS, with increasing concentrations of thiostrepton (0, 1, 2 or 3 μM) for 24 h at 37°C. The medium was then replaced with RPMI-1640 containing 10% FBS and doxorubicin (0.2 μM), in which the cells were incubated for 1 h at 37°C. The cells were then harvested and washed twice in ice-cold PBS. The cell-associated mean fluorescence intensity (MFI) of intracellular doxorubicin was determined using the FACS FC500 flow cytometer with excitation and emission wavelengths of 485 and 585 nm, respectively.

Statistical analysis

Statistical analyses were performed using SPSS 19.0 software (IBM SPSS, Armonk, NY, USA). Data are expressed as the mean ± standard deviation. The significance of differences between groups were determined using Student’s t-test or analysis of variance. P<0.05 was considered to indicate a statistically significant difference. The synergistic or antagonistic effects of the two drugs were evaluated, according to the formula [Q = Ea + b / (Ea + Eb - EaxEb)]. In this equation, Ea and Eb represent the individual inhibitory rates of drugs A and B, respectively, and Ea + b represents the inhibitory rate of the combined drug therapy on tumor cell proliferation. Q-values ranging between 0.85 and 1.15 indicated that the effects of the two drugs were simply additive, whereas Q-values >1.15 indicated a synergistic effect, and Q-values <0.85 indicated an antagonistic effect for the combined drug therapy.

Results

Thiostrepton suppresses the viability of Jurkat cells

The Jurkat cells were treated with various concentrations of thiostrepton (0–4 μM) for 24, 48 or 72 h, and the effect of thiostrepton on cell viability was evaluated using a CCK-8 assay. The results demonstrated that the Jurkat cells were sensitive to thiostrepton, and that thiostrepton inhibited cell growth in a time- and dose-dependent manner (P<0.05; Fig. 1). The IC50 values for thiostrepton in the Jurkat cells at 24, 48 and 72 h were 2.03, 1.52 and 1.29 μM, respectively.

Thiostrepton-mediated inhibition of FoxM1 induces cell cycle arrest at the G2/M phase and leads to apoptosis of Jurkat cells

To further characterize the mechanism underlying the antileukemia activity of thiostrepton on the Jurkat cells, the mRNA and protein expression levels of FoxM1 were determined following treatment with various concentrations of thiostrepton (0, 1, 2 or 3 μM) for 24 h, to examine whether thiostrepton affected cell cycle distribution or apoptosis. The results revealed that thiostrepton inhibited the expression of FoxM1 in a dose-dependent manner, and induced gradual dose-dependent G2/M arrest and apoptosis in the Jurkat cells (Fig. 2).

Inhibition of FoxM1 by thiostrepton enhances the antiproliferative effects of doxorubicin on Jurkat cells

Based on the aforementioned findings, the present study subsequently investigated whether the Jurkat cells with reduced expression of FoxM1 were more sensitive to doxorubicin, a common antileukemic drug used in the treatment of ALL. As shown in Fig. 1, treatment with 1 μM thiostrepton for 24 h resulted in a 7.70±0.95% reduction of viable cells (P<0.05) compared with the control group, therefore, the concentration of 1 μM was considered to be a non-cytotoxic dose. The Jurkat cells were incubated with various concentrations of doxorubicin (0–0.8 μm/l), with or without thiostrepton (1 μm/l), for 24 h. Cell proliferation was measured by performing CCK-8 assays. The results demonstrated that the combined treatment reduced the IC50 value of doxorubicin in the Jurkat cells between 0.295 and 0.198 μmol/l (Fig. 3). Thiostrepton and doxorubicin were identified to synergistically inhibit cell growth, with Q values often >1.15 (Table II). It was suggested that the inhibition of FoxM1 by thiostrepton increased the chemosensitivity of Jurkat cells to doxorubicin.

Table II

Interaction between doxorubicin and thiostrepton in combined treatment with different doses of doxorubicin.

Table II

Interaction between doxorubicin and thiostrepton in combined treatment with different doses of doxorubicin.

Doxorubicin (μM)aQ-value
0.0250.86
0.051.17
0.11.32
0.21.61
0.31.43
0.41.25
0.81.07

{ label (or @symbol) needed for fn[@id='tfn2-mmr-12-01-1457'] } Q>1.15 indicates to a synergistic interaction;

a At each dose of doxorubicin, 1 μM thiostrepton was used.

Thiostrepton enhances doxorubicin-induced caspase-mediated apoptosis in Jurkat cells

The present study then examined whether thiostrepton enhances doxorubicin-induced cell apoptosis. The Jurkat cells were treated with a low concentration of either thiostrepton (1 μm/l) or doxorubicin (0.2 μm/l), or treated with the two drugs in combination for 24 h. Flow cytometric analysis revealed that thiostrepton increased doxorubicin-induced apoptosis between 16.98±2.87 and 36.07±4.18% (P<0.05; Fig. 4A). To determine the mechanism leading to the cell apoptosis, the protein expression levels of survivin and caspase-3 were examined. As shown in Fig. 4B, decreases in the expression of survivin were significantly greater in the cells treated with thiostrepton and doxorubicin in combination, compared with those in the single treatment groups. In addition, combination treatment resulted in greater increases in the protein expression of caspase-3 compared with either drug alone (Fig. 4B).

Thiostrepton increases the intracellular accumulation of doxorubicin in Jurkat cells

The capability of thiostrepton to promote doxorubicin accumulation within Jurkat cells was observed (Fig. 5). The doxorubicin-associated MFI was significantly increased in Jurkat cells, which were treated with various concentrations of thiostrepton (1, 2 or 3 μM) for 24 h, which explained the enhanced effects of thiostrepton on doxorubicin cytotoxicity in the Jurkat cells. In addition, thiostrepton was observed to elevate the concentration of doxorubicin in the Jurkat cells in a dose-dependent manner (P<0.05).

Inhibition of FoxM1 by thiostrepton decreases the gene and protein expression levels of GSTpi within the Jurkat cells

To better understand the mechanism by which thiostrepton functionally elevated drug accumulation within the Jurkat cells, the mRNA and protein expression levels of GSTpi were quantitatively measured. The Jurkat cells were treated with various concentrations of thiostrepton (0, 1, 2 or 3 μM) for 24 h. The gene and protein expression levels of GSTpi in the Jurkat cells decreased significantly when the cells were treated with thiostrepton (P<0.05; Fig. 6). The results indicated that the inhibition of FoxM1 by thiostrepton downregulated the expression of GSTpi in a dose-dependent manner in the Jurkat cells (P<0.05).

Discussion

T-ALL is a genetically heterogeneous type of cancer, with 20% of childhood patients and the majority of adult patients succumbing to mortality from disease resistance or relapse (27). Current standard therapies have unsatisfactory effects on survival rates, and therapy-associated side-effects have led to a focus on identifying novel therapeutic approaches for T-ALL. The present study was the first, to the best of our knowledge, to demonstrate thiostrepton-induced inhibition of FoxM1 in human T-ALL Jurkat cells. The results revealed that thiostrepton inhibited Jurkat cell proliferation and induced apoptosis in a dose-dependent manner. It was also demonstrated that thiostrepton enhanced the chemo-sensitivity of the Jurkat cells to doxorubicin by enhancing doxorubicin-induced apoptosis and increasing the intracellular accumulation of doxorubicin.

The present study investigated the antileukemic effects of thiostrepton on Jurkat cell proliferation. Thiostrepton significantly inhibited the proliferation of Jurkat cells, and the IC50 value of thiostrepton in the Jurkat cells following 48 h treatment was 1.52 μmol/l, suggesting the sensitivity of the Jurkat cells to thiostrepton was comparable to that of other types of cancer cell (28,29). Furthermore, the results of the present study indicated that thiostrepton induced G2/M cell cycle phase arrest and increased apoptosis in the Jurkat cells. These results demonstrated the efficient antileukemic activity of thiostrepton on Jurkat cells, and suggested that the effects of thiostrepton may depend on the downregulation of FoxM1 target genes, which have been found to be involved in the regulation of cell growth, apoptosis, cell cycle and progression (30).

Previous studies have suggested that silencing FoxM1 enhances sensitivity to chemotherapeutic agents in various types of cancer cells (14,29), therefore, the present study investigated whether the thiostrepton-mediated inhibition of FoxM1 sensitized the Jurkat cells to the effects of chemotherapy. the IC50 value of doxorubicin in the Jurkat cells decreased between 0.295 and 0.198 μmol/l when thiostrepton (1 μmol/l) was used in combination. These results demonstrated that thiostrepton enhanced the doxorubicin cytotoxicity effect within the Jurkat cells through a synergistic effect of thiostrepton and doxorubicin. Therefore, a combination therapy approach using doxorubicin and thiostrepton can improve the effect of doxorubicin and also reduce the adverse side-effects of doxorubicin, by allowing it to be used at lower doses in treating human T-ALL.

The ability to induce leukemic cell death is critical for successful treatment with chemotherapeutic drugs. In the present study, flow cytometric analysis revealed a more marked increase in apoptosis in cells treated with a combination of thiostrepton and doxorubicin, compared with those treated with either drug alone. To further examine the mechanism underlying the increased efficacy of combination treatment, proteins, which are known to regulate apoptosis, were examined. Survivin is known for its role in preventing apoptosis and conferring resistance to chemotherapeutic agents in various cell lines (31). Specifically, its anti-apoptotic function appears to be associated with the ability to directly or indirectly inhibit caspases (31). In the present study, the cells treated with thiostrepton and doxorubicin combined exhibited a greater decrease in the protein expression of survivin compared with the cells treatedwith doxorubicin only, and thiostrepton also enhanced doxorubicin-induced increases in the protein expression of caspase-3. These results suggested that the inhibition of FoxM1 by thiostrepton sensitized the Jurkat cells to doxorubicin, partly through enhanced apoptosis, possibly via a caspase-3-dependent pathway.

In the present study, thiostrepton significantly elevated the concentration of doxorubicin in Jurkat cells in a dose-dependent manner, while promoting doxorubicin accumulation within the Jurkat cells. To determine the mechanism by which thiostrepton functionally elevated drug accumulation within the Jurkat cells, the expression of GSTpi was measured. Previous studies have demonstrated that the mRNA and protein expression levels of GSTpi are high in Jurkat cells (32). GSTpi is a cytosolic isoform of glutathione s-transferases (GSTs) and its overexpression is associated with resistance to chemotherapy in various types of human cell (33). GSTs belong to a large family of functional enzymes, which catalyze the S-conjugation of glutathione (GSH) with a wide variety of electrophilic compounds (34). In addition to increasing GST enzyme activity, alteration of cellular GSH levels has also been observed to contribute the resistance of cells to doxorubicin (35). The results of the present study further revealed that the mRNA and protein expression levels of the GSTpi gene were significantly inhibited by thiostrepton. These results indicated that thiostrepton elevated the sensitivity of Jurkat cells to doxorubicin, partly by increasing the accumulation of intracellular doxorubicin, possibly via downregulating the expression of GSTpi.

In conclusion, the results of the present study demonstrated that thiostrepton significantly inhibited the proliferation, and induced the apoptosis of Jurkat cells. The inhibition of FoxM1 by thiostrepton also chemosensitized the Jurkat cells to doxorubicin by enhancing doxorubicin-induced apoptosis and elevating the accumulation of intracellular doxorubicin. These results suggested that the inhibition of FoxM1 by thiostrepton have wide therapeutic and/or adjuvant applications for T-ALL chemotherapy. Thiostrepton may be particularly useful in young children with resistant or relapsed T-ALL, who currently require more aggressive chemotherapeutic agents.

Acknowledgments

This study was supported by the NCET-10-0919, ‘Taishan Scholar’ position, National Natural Science Foundation (nos. 31371321 and 81200601), the Shandong Science and Technology Committee (no. 2010GSF10264), the Foundation of Shandong Educational Committee (nos. J10LC60 and J11LC01) and the Binzhou Science and Technology Committee of China (no. 2011ZC0905).

References

1 

Inaba H, Greaves M and Mullighan CG: Acute lymphoblastic leukaemia. Lancet. 381:1943–1955. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Bhojwani D and Pui CH: Relapsed childhood acute lymphoblastic leukaemia. Lancet Oncol. 14:e205–217. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Van Vlierberghe P and Ferrando A: The molecular basis of T cell acute lymphoblastic leukemia. J Clin Invest. 122:3398–3406. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Martelli AM, Lonetti A, Buontempo F, et al: Targeting signaling pathways in T-cell acute lymphoblastic leukemia initiating cells. Adv Biol Regul. 56:6–21. Apr 30–2014.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

5 

Wierstra I and Alves J: FOXM1, a typical proliferation-associated transcription factor. Biol Chem. 388:1257–1274. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Koo CY, Muir KW and Lam EW: FOXM1: From cancer initiation to progression and treatment. Biochim Biophys Acta. 1819:28–37. 2012. View Article : Google Scholar

7 

Priller M, Pöschl J, Abrão L, et al: Expression of FoxM1 is required for the proliferation of medulloblastoma cells and indicates worse survival of patients. Clin Cancer Res. 17:6791–6801. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Xue YJ, Xiao RH, Long DZ, et al: Overexpression of FoxM1 is associated with tumor progression in patients with clear cell renal cell carcinoma. J Transl Med. 10:2002012. View Article : Google Scholar : PubMed/NCBI

9 

Xia JT, Wang H, Liang LJ, et al: Overexpression of FOXM1 is associated with poor prognosis and clinicopathologic stage of pancreatic ductal adenocarcinoma. Pancreas. 41:629–635. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Xu N, Jia D, Chen W, et al: FoxM1 is associated with poor prognosis of non-small cell lung cancer patients through promoting tumor metastasis. PLoS One. 8:e594122013. View Article : Google Scholar : PubMed/NCBI

11 

Li X, Qiu W, Liu B, et al: Forkhead box transcription factor 1 expression in gastric cancer: FOXM1 is a poor prognostic factor and mediates resistance to docetaxel. J Transl Med. 11:2042013. View Article : Google Scholar : PubMed/NCBI

12 

Liu LL, Zhang DH, Mao X, Zhang XH and Zhang B: Over-expression of FoxM1 is associated with adverse prognosis and FLT3-ITD in acute myeloid leukemia. Biochem Biophys Res Commun. 446:280–285. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Myatt SS and Lam EW: The emerging roles of forkhead box (Fox) proteins in cancer. Nat Rev Cancer. 7:847–859. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Halasi M and Gartel AL: Suppression of FOXM1 sensitizes human cancer cells to cell death induced by DNA-damage. PLoS One. 7:e317612012. View Article : Google Scholar : PubMed/NCBI

15 

Nakamura S, Hirano I, Okinaka K, et al: The FOXM1 transcriptional factor promotes the proliferation of leukemia cells through modulation of cell cycle progression in acute myeloid leukemia. Carcinogenesis. 31:2012–2021. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Zhang X, Zeng J, Zhou M, et al: The tumor suppressive role of miRNA-370 by targeting FoxM1 in acute myeloid leukemia. Mol Cancer. 11:562012. View Article : Google Scholar : PubMed/NCBI

17 

Mencalha AL, Binato R, Ferreira GM, Du Rocher B and Abdelhay E: Forkhead box M1 (FoxM1) gene is a new STAT3 transcriptional factor target and is essential for proliferation, survival and DNA repair of K562 cell line. PloS one. 7:e481602012. View Article : Google Scholar : PubMed/NCBI

18 

Bhat UG, Halasi M and Gartel AL: FoxM1 is a general target for proteasome inhibitors. PLoS One. 4:e65932009. View Article : Google Scholar : PubMed/NCBI

19 

Hegde NS, Sanders DA, Rodriguez R and Balasubramanian S: The transcription factor FOXM1 is a cellular target of the natural product thiostrepton. Nat Chem. 3:725–731. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Hedge NS, Sanders DA, Rodriguez R and Balasubramanian S: The transcription factor FOXM1 is a cellular target of the natural product thiostrepton. Nat Chem. 3:725–731. 2011. View Article : Google Scholar

21 

Gartel AL: Thiostrepton, proteasome inhibitors and FOXM1. Cell Cycle. 10:4341–4342. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Halasi M, Schraufnagel DP and Gartel AL: Wild-type p53 protects normal cells against apoptosis induced by thiostrepton. Cell Cycle. 8:2850–2851. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Ahmed M, Uddin S, Hussain AR, et al: FoxM1 and its association with matrix metalloproteinases (MMP) signaling pathway in papillary thyroid carcinoma. J Clin Endocrinol Metab. 97:E1–13. 2012. View Article : Google Scholar

24 

Wang M and Gartel AL: Micelle-encapsulated thiostrepton as an effective nanomedicine for inhibiting tumor growth and for suppressing FOXM1 in human xenografts. Mol Cancer Ther. 10:2287–2297. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Liu X, Li X, Wang L, et al: Realgar induces apoptosis in the chronic lymphocytic leukemia cell line MEC1. Mol Med Rep. 8:1866–1870. 2013.PubMed/NCBI

26 

Ji BS, He L and Liu GQ: Reversal of p-glycoprotein-mediated multidrug resistance by CJX1, an amlodipine derivative, in doxorubicin-resistant human myelogenous leukemia (K562/DOX) cells. Life Sci. 77:2221–2232. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Van Vlierberghe P, A mbesi-I mpiombato A, De Keersmaecker K, et al: Prognostic relevance of integrated genetic profiling in adult T-cell acute lymphoblastic leukemia. Blood. 122:74–82. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Bhat UG, Halasi M and Gartel AL: Thiazole antibiotics target FoxM1 and induce apoptosis in human cancer cells. PLoS One. 4:e55922009. View Article : Google Scholar : PubMed/NCBI

29 

Lin J, Zheng Y, Chen K, Huang Z, Wu X and Zhang N: Inhibition of FOXM1 by thiostrepton sensitizes medulloblastoma to the effects of chemotherapy. Oncol Rep. 30:1739–1744. 2013.PubMed/NCBI

30 

Halasi M and Gartel AL: FOX(M1) news - it is cancer. Mol Cancer Ther. 12:245–254. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Zaffaroni N, Pennati M and Daidone MG: Survivin as a target for new anticancer interventions. J Cell Mol Med. 9:360–372. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Zhou L, Jing Y, Styblo M, Chen Z and Waxman S: Glutathione-S-transferase pi inhibits As2O3-induced apoptosis in lymphoma cells: involvement of hydrogen peroxide catabolism. Blood. 105:1198–1203. 2005. View Article : Google Scholar

33 

Lourenco GJ, Lorand-Metze I, Delamain MT, et al: Polymorphisms of glutathione S-transferase mu 1, theta 1 and pi 1 genes and prognosis in Hodgkin lymphoma. Leuk Lymphoma. 51:2215–2221. 2010. View Article : Google Scholar

34 

Ballerini S, Bellincampi L, Bernardini S, Iori R, Cortese C and Federici G: Analysis of GSTP1–1 polymorphism using real-time polymerase chain reaction. Clin Chim Acta. 329:127–132. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Song YN, Guo XL, Zheng BB, et al: Ligustrazine derivate DLJ14 reduces multidrug resistance of K562/A02 cells by modulating GSTπ activity. Toxicol In Vitro. 25:937–943. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2015
Volume 12 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang JY, Jia XH, Xing HY, Li YJ, Fan WW, Li N and Xie SY: Inhibition of Forkhead box protein M1 by thiostrepton increases chemosensitivity to doxorubicin in T-cell acute lymphoblastic leukemia. Mol Med Rep 12: 1457-1464, 2015
APA
Wang, J., Jia, X., Xing, H., Li, Y., Fan, W., Li, N., & Xie, S. (2015). Inhibition of Forkhead box protein M1 by thiostrepton increases chemosensitivity to doxorubicin in T-cell acute lymphoblastic leukemia. Molecular Medicine Reports, 12, 1457-1464. https://doi.org/10.3892/mmr.2015.3469
MLA
Wang, J., Jia, X., Xing, H., Li, Y., Fan, W., Li, N., Xie, S."Inhibition of Forkhead box protein M1 by thiostrepton increases chemosensitivity to doxorubicin in T-cell acute lymphoblastic leukemia". Molecular Medicine Reports 12.1 (2015): 1457-1464.
Chicago
Wang, J., Jia, X., Xing, H., Li, Y., Fan, W., Li, N., Xie, S."Inhibition of Forkhead box protein M1 by thiostrepton increases chemosensitivity to doxorubicin in T-cell acute lymphoblastic leukemia". Molecular Medicine Reports 12, no. 1 (2015): 1457-1464. https://doi.org/10.3892/mmr.2015.3469