Open Access

Arsenic trioxide and triptolide synergistically induce apoptosis in the SKM‑1 human myelodysplastic syndrome cell line

  • Authors:
    • Hai‑Ying Hua
    • Hua‑Qiang Gao
    • Ai‑Ning Sun
    • Jian‑Nong Cen
    • Li‑Li Wu
  • View Affiliations

  • Published online on: September 26, 2016     https://doi.org/10.3892/mmr.2016.5779
  • Pages: 4180-4186
  • Copyright: © Hua 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

Although certain combination therapies comprising arsenic trioxide (As2O3) with other agents exist for the treatment of several types of human cancer, few As2O3 combination therapies are clinically effective for myelodysplastic syndromes (MDS). Triptolide (TL) may be an effective therapeutic agent for the treatment of MDS. However, to date, there is no combination therapy for MDS with As2O3 and TL. Therefore, the aim of the present study was to investigate this combination therapy on the apoptosis of MDS SKM‑1 cells. The MDS SKM‑1 cells were treated with As2O3, TL or the two in combination at various concentrations, or were mock‑treated. Cell viability, cell apoptosis, levels of reactive oxygen species (ROS) and the expression of the cell apoptosis‑associated genes, B cell lymphoma‑2 (Bcl‑2), Bcl‑2‑associated X protein (Bax) and caspase‑3, were determined using an MTT assay, flow cytometric analysis of annexin V‑fluorescein isothiocyanate/propidium iodide double‑stained cells, flow cytometic analysis of intracellular 2',7'‑dichlorodihydrofluorescein diacetate fluorescence and reverse transcription‑quantitative polymerase chain reaction analysis, respectively. Combination index (CI) analysis was performed to determine whether effects were synergistic (CI<1). The combination treatment was found to synergistically inhibit MDS SKM‑1 cell growth, induce cell apoptosis, increase ROS levels, upregulate the expression levels of Bax and caspase‑3, and downregulate the mRNA expression of Bcl‑2. In conclusion, the combination treatment of As2O3 and TL synergistically induced apoptosis in the MDS SKM‑1 cells.

Introduction

Myelodysplastic syndromes (MDS) are clonal stem cell disorders characterized by peripheral cytopenias with dysplasia in one or more cell lineages, including erythrocytic, granulocytic and megakaryocytic lineages, leading to the progression to acute myelogenous leukemia (AML) with a poor prognosis (14). At present, allogeneic hematopoietic stem-cell transplantation is the only treatment option, which can induce long-term remission (5,6). However, its use is only possible in a minority of patients with MDS due to the advanced age of presentation, limited availability of donor sources, high rate of treatment-associated mortality (~39% at 1 year), suboptimal disease-free survival rates (~29% at 5 years) and chronic graft-versus-host disease (~15% at 1 year) (6). Aberrant DNA methylation is frequently associated with MDS; therefore, demethylating agents, including as azacytidine and decitabine, are used to treat patients with MDS. However, treatment of patients with a higher risk of MDS with azacitidine (7,8) only increases the overall survival rate to 24.5 months, compared with 15.0 months with conventional care, supportive care, treatment with low-dose cytarabine or intensive chemotherapy. In addition, treatment with decitabine (9) prolongs the median duration of the progression of AML or associated mortality rates to 12 months, compared with 6.8 months following supportive care alone. In addition, the rates of complete remission (9–17%) following treatment with demethylating agents (79) are similar to those following conventional care with low-dose cytarabine (11–18%) (10), and substantially lower, compared with those following induction chemotherapy in patients with AML (>50%) (11). Lenalidomide, a derivative of thalidomide, reduces transfusion requirements, and reverses cytologic and cytogenetic abnormalities in patients who have MDS with the 5q31 deletion (12). However, lenalidomide increases the risk of developing other malignancies, including AML and B-cell lymphoma (13). Thus, a more effective treatment option for MDS is urgently required.

Arsenic trioxide (As2O3) is a traditional Chinese medicine, which is effective in the clinical management of patients with acute promyelocytic leukemia (APL) (14,15). However, in two-phase II multicenter trials, rates of hematological improvement with As2O3 were 20–29%, with moderate toxicity reported (16,17). As2O3 induces the apoptosis of nonpromyelocytic leukemia and other types of malignant tumor cells (1820) through the inhibition of B cell lymphoma-2 (Bcl-2) (21), and the upregulation of Bcl-2-associated X protein (Bax) (22) and caspase-3 (23).

Extracts of the Chinese herb, Tripterygium wilfordii Hook F are used to treat autoimmune and/or inflammatory diseases, and triptolide (TL) is the active substance of these extracts in vitro and in vivo (24). Several studies have demonstrated that TL may be an effective therapeutic agent for the treatment of MDS (25), several types of human pancreatic (26) and adrenal (27) cancer, and T cell lymphocytic leukemia (28) via inducing cell apoptosis through the activation of caspase-3 and generation of reactive oxygen species (ROS) (2527).

Although certain combination therapies involving As2O3 and other agents, are ongoing for several types of human cancer, few As2O3 combination therapies are clinically effective. These include combination therapy of As2O3 with ascorbic acid in nonrefractory APL hematologic malignancies and multiple myeloma (18), but not in other AML except nonrefractory APL, acute lymphoid leukemia (18), chronic myeloid leukemia and chronic lymphoid leukemia (18). The use of phase 2 combination therapy with As2O3 and gemtuzumab ozogamicin for the treatment of MDS and secondary AML has been found to have acceptable response rates and toxicity, however, the median overall survival rate was only 9.7 months (29).

The aim of the present study was to investigate the effect of As2O3 in combination with TL on the apoptosis of MDS SKM-1 cells by evaluating the gene expression levels of Bcl-2, Bax and caspase-3, and the generation of ROS.

Materials and methods

Reagents and cell culture

TL (purity >99.0%; Chinese Academy of Medical Sciences, Nanjing, China) was dissolved in dimethyl sulfoxide (DMSO; Sigma-Aldrich; Thermo Fisher Scientific, Inc., Waltham, MA, USA) to form a 1 mM stock solution. As2O3 powder (Beijing Double-Crane Pharmaceutical Co., Ltd., Beijing, China) was dissolved in phosphate-buffered saline (PBS). The MDS SKM-1 cell line was obtained from the Cell Bank of the Japanese Collection of Research Bioresources (Osaka, Japan). The SKM-1 cells were cultured in RPMI 1640 medium (Life Technologies; Thermo Fisher Scientific, Inc.) supplemented with 10% fetal calf serum and 1% penicillin/streptomycin at 37°C in a humidified incubator with 5% CO2. Cells in the second to fourth passages and logarithmic growth phase, with >95% viability on trypan blue staining, were used for the following experiments.

Cell treatment and cell viability assessment using an MTT assay

The cells were seeded at a density of 4–6×104 cells/well in 96-well plates, cultured RPMI 1640 medium (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% fetal calf serum and 1% penicillin/streptomycin mixture at 37°C in humidified incubator with 5% CO2 for 48 h and treated with various concentrations of As2O3 (0.25, 0.5, 2, 8 or 32 µM), TL (10, 20, 40, 80 or 160 ng/ml) or As2O3+TL (0.25+10 ng/ml, 0.5+20 ng/ml, 2.0+40 ng/ml, 8+80 ng/ml or 32+160 ng/ml), or were mock-treated with RPMI-1640 medium containing 0.002% DMSO. Following treatment for 48 h, cell viability was assessed using a CellTiter 96 AQueous One Solution Cell Proliferation Assay kit (Promega, Nanjing, China), according to the manufacturer's protocol. The absorbance at 490 nm was measured using a SpectraMAX M5 spectrophotometer (Molecular Devices, LLC, Sunnyvale, CA, USA).

Flow cytometric analysis of MDS SKM-1 cell apoptosis

Following treatment of the cells for 48 h with As2O3, TL, As2O3 and TL, or mock treatment with RPMI-1640 media, the cells were collected by centrifugation at 1,300 × g for 3 min at room temperature, washed twice with PBS (BD Biosciences, Beijing, China), and resuspended in binding buffer (Novagen; EMD Millipore, Billerica, MA, USA) at 1×106 cells/ml. Subsequently, the cells were stained with 5 µl of annexin V-fluorescein isothiocyanate (FITC) and 5 µl of propidium iodide (PI), incubated in the dark at room temperature for 15 min, and mixed with binding buffer (400 µl). Analysis of apoptosis was then performed on a Calibur flow cytometer (BD Biosciences). Early and late apoptotic cells were calculated based on annexin V-positivity/PI-negativity and annexin V-positivity/PI-positivity, respectively.

Intracellular ROS

The cells (3×105/well) in 6-well plates were treated with As2O3, TL, As2O3 and TL or mock treatment, cultured in RPMI 1640 medium, supplemented with 10% FCS and 1% penicillin/streptomycin mixture at 37°C in humidified incubator with 5% CO2 for 48 h. Following treatment, the cells were washed once with PBS and treated with 100 nM 2′,7′-dichlorodihydrofluorescein diacetate in a cell culture incubator for 30 min at 37°C with 5% CO2. Following trypsinization, the cells were washed once with PBS and centrifuged at 1,300 × g for 3 min. The cell pellets were then resuspended in 1 ml PBS and analyzed on a Calibur flow cytometer (BD Biosciences).

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

Following the treatment of the cells for 48 h, total RNA was extracted using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. RT-qPCR analysis was performed on an ABI 7900 sequence detection system (Applied Biosystems; Thermo Fisher Scientific, Inc.) using a qRT-PCR kit (Qiagen, Beijing, China), according to the manufacturer's protocol. The Abl gene was used as an internal control. The primer sequences were as follows: Bcl-2, forward (F) 5′-AGGATCATGCTGTACTTAA-3′ and reverse (R) 5′-ATGAGGCACGTTATTATTAG-3′; Bax, F 5′-CGAACTGGACAGTAACAT-3′ and R 5′-CTCGGAAAAAGACCTCTC-3′; caspase-3, F 5′-TTGTAGAAGTCTAACTGGAA-3′ and R 5′-CCATGTCATCATCAACAC-3′; Abl, F 5′-GATACGAAGGGAGGGTGTACCA-3′ and R 5′-CTCGGCCAGGGTGTTGAA-3′. The 25 µl PCR reaction system included PCR mix 12.5 µl, F primer 0.5 µl, R primer 0.5 µl, probe 0.3 µl, ddH2O 7.2 µl, cDNA 4 µl. The reaction parameters of Bcl-2, Bax and caspase-3 were as follows: 94°C 5 min, 94°C 40 sec, 56°C 55 sec, 72°C 1 min for 45 cycles, 72°C extension 7 min; 94°C 5 min, 94°C 40 sec, 58°C 55 sec, 72°C 1 min for 45 cycles, 72°C extension 7 min; 94°C 5 min, 94°C 40 sec, 50°C 55 sec, 72°C 1 min for 45 cycles, 72°C extension 7 min. The results were reported as 2−∆∆Cq relative to the gene expression of Abl (30).

Statistical analysis

Statistical analysis was performed with SPSS version 16.0 (SPSS, Inc., Chicago, IL, USA). Data are expressed as the mean ± standard error of the mean. Statistical analysis was performed using one-way analysis of variance followed by the least significant difference post-hoc test and Student's t-test. Factorial design analysis of variance was used to determine additive or synergistic effects. P<0.05 was considered to indicate a statistically significant difference.

Results

As2O3 and TL synergistically inhibit the growth of MDS SKM-1 cells

To examine whether TL enhances the chemosensitivity of MDS SKM-1 cells to As2O3, the present study examined the growth of MDS SKM-1 cells following treatment with As2O3 in combination with TL. The combination treatment of As2O3+TL substantially suppressed SKM-1 cell growth, compared with the cells treated with As2O3 or TL alone (Fig. 1A). To evaluate whether the cell growth inhibition induced by the combination of TL+As2O3 was additive or synergistic, the CI values were determined according to the Chou-Talalay combination index equation CI=(C)1/(CX)1+(C)2/(CX)2+(C)1(C)2/(CX)1(CX)2 (31), where CI <1 defines synergism. The CI analysis revealed that the CI values ranged between 0.70 and 0.87 (Fig. 1B). These results indicated that As2O3 and TL synergistically inhibited MDS SKM-1 cell growth.

As2O3 and TL synergistically induce apoptosis in MDS SKM-1 cells

To examine whether As2O3 and TL synergistically inhibit MDS SKM-1 cell growth through the induction of cell apoptosis by treatment with As2O3 in combination with TL, cell apoptosis was assessed using flow cytometry with annexin V-FITC/PI double staining. The combination treatment of As2O3+TL substantially induced SKM-1 cell apoptosis, compared with either As2O3 or TL alone (Fig. 2A). CI analysis revealed that the CI values ranged between 0.65 and 0.85 (Fig. 2B). The results indicated that As2O3 and TL synergistically induced MDS SKM-1 cell apoptosis.

As2O3 and TL synergistically induce apoptosis via the generation of ROS in MDS SKM-1 cells

Treatment with As2O3 in combination with TL substantially increased the intracellular ROS levels, compared with either As2O3 or TL alone (Fig. 3A; P<0.01). CI analysis revealed that the CI values ranged between 0.60 and 0.86 (Fig. 3B). The results indicated that As2O3 and TL synergistically induced MDS SKM-1 cell apoptosis via the generation of ROS.

As2O3 and TL synergistically regulate the expression of apoptosis-associated genes in MDS SKM-1 cells

To determine whether As2O3 in combination with TL synergistically regulates the expression of apoptosis-associated genes, the mRNA expression levels of Bax, Bcl-2 and caspase-3 were measured in the cells treated with As2O3, TL or As2O3+TL for 48 h. As shown in Fig. 4, treatment with As2O3+TL led to significant increases in the expression levels of Bax and caspase-3, and a significant decrease in the mRNA expression of Bcl-2, compared with either As2O3 or TL alone (P<0.01; Fig. 4A-C). These results demonstrated that the combination of As2O3 and TL significantly induced apoptotic activity via inhibiting Bcl-2 and promoting the expression of Bax and caspase-3. CI analysis revealed that the CI values were 0.57–0.82 for Bax (Fig. 4A), 0.53–0.78 for Bcl-2 (Fig. 4B) and 0.56–0.82 for caspase-3 (Fig. 4C). These results indicated that As2O3 and TL synergistically induced MDS SKM-1 cell apoptosis via increasing the mRNA expression levels of Bax and caspase-3 and decreasing the mRNA expression of Bcl-2.

Discussion

To investigate whether TL enhances the chemosensitivity of MDS SKM-1 cells to As2O3, the present study treated MDS SKM-1 cells with As2O3, TL or the two in combination. It was found that As2O3/TL synergistically inhibited SKM-1 cell growth through upregulation of ROS levels and cell apoptosis, as evidenced by synergistically increased expression levels of Bax and caspase-3, and decreased mRNA expression of Bcl-2.

The present study found that As2O3+ TL synergistically induced MDS SKM-1 cell apoptosis, determined from analysis of annexin V-FITC/PI double staining using flow cytometry. Of note, As2O3, in combination with a mitogen-activated protein kinase kinase or proteinase (32) inhibitor, has been shown experimentally to have a synergistic effect on the induction of AML cell apoptosis. The present study also found that the combination treatment with As2O3 and TL resulted in a significant increase in the mRNA expression levels of Bax and caspase-3, and a significant decrease in the mRNA expression of Bcl-2, compared with the cells treated with either As2O3 or TL alone. To evaluate whether the combination of TL and As2O3 increased the mRNA expression levels of Bax and caspase-3 and decreased the mRNA expression of Bcl-2 in an additive or synergistic manner, the CI values were determined. The results indicated that As2O3+TL synergistically induced MDS SKM-1 cell apoptosis via increasing the mRNA expression levels of Bax and caspase-3 and decreasing the expression of Bcl-2 (CI<1). These results suggested that the synergistic cell apoptosis induced by the combination treatment resulted from inhibiting the mRNA expression of Bcl-2 and promoting the mRNA expression levels of Bax and caspase-3. It has been reported previously that As2O3 induces cell apoptosis via the upregulation of Bax (21) and the Bax/Bcl-2 ratio (22), and the downregulation of Bcl-2 (18). Caspase-3 is a member of the cysteine-aspartic acid protease family (33), and sequential activation of caspase proteins is central to the apoptosis of a variety of cancer cells (2123,26). TL induces human breast and prostate cancer cell apoptosis (33), and TL in combination with tumor-necrosis factor-related apoptosis-inducing ligand enhances the apoptosis of cholangiocarcinoma cells by increasing the activity caspase-3 (34). In addition, the combination treatment of low-dose 1,25-dihydroxyvitamin D(3) combined with As2O3 synergistically inhibits AML cell proliferation via cell apoptosis mediated by the increased expression levels of Bax and caspase-3, and decreased expression of Bcl-2 (34).

The present study also found that the combination treatment of As2O3 with TL synergistically increased the generation of ROS in the cells. Therefore, it was hypothesized that the induction of cell apoptosis by the combination treatment in the present study was mediated by the generation of ROS. It is well known that the presence of increased intracellular ROS in the mitochondria is involved in the induction of apoptosis in cancer cells, and that an increased intracellular ROS concentration has been shown to cause an increase in the Bax/Bcl-2 ratio and activation of caspase-3 (35,36). In the present study, it was found that, compared with the cells treated with either As2O3 or TL alone, the generation of intracellular ROS was significantly increased following exposure to As2O3 and TL in combination. TL has been found to induce human adrenal cancer NCI-H295 cell apoptosis through the ROS pathway (27), and treatments involving the combination of As2O3 and sulindac (34) or phytosphingosine (37) have been shown to enhance apoptotic cell death via increasing intracellular ROS.

In conclusion, the present study demonstrated that treatment with As2O3 in combination with TL synergistically induced MDS SKM-1 cell apoptosis via the induction of intracellular ROS, which upregulated the expression of Bax, downregulated the expression of Bcl-2 and upregulated the expression of caspase-3 (Fig. 5). These findings may provide a strategy to develop a novel combination therapy against MDS.

Acknowledgements

The present study was supported by a grant (grant no. LZ09103) from the Jiangsu Provincial Bureau of traditional Chinese Medicine (Jaingsu, China).

References

1 

Cogle CR, Craig BM, Rollison DE and List AF: Incidence of the myelodysplastic syndromes using a novel claims-based algorithm: High number of uncaptured cases by cancer registries. Blood. 117:7121–7125. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Jädersten M and Hellström-Lindberg E: Myelodysplastic syndromes: Biology and treatment. J Intern Med. 265:307–328. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Tefferi A and Vardiman JW: Myelodysplastic syndromes. N Engl J Med. 361:1872–1885. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Gore SD and Hermes-DeSantis ER: Enhancing survival outcomes in the management of patients with higher-risk myelodysplastic syndromes. Cancer Control. 16:(Suppl). S2–S10. 2009.

5 

Chang C, Storer BE, Scott BL, Bryant EM, Shulman HM, Flowers ME, Sandmaier BM, Witherspoon RP, Nash RA, Sanders JE, et al: Hematopoietic cell transplantation in patients with myelodysplastic syndrome or acute myeloid leukemia arising from myelodysplastic syndrome: Similar outcomes in patients with de novo disease and disease following prior therapy or antecedent hematologic disorders. Blood. 110:1379–1387. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Warlick ED, Cioc A, Defor T, Dolan M and Weisdorf D: Allogeneic stem cell transplantation for adults with myelodysplastic syndromes: Importance of pretransplant disease burden. Biol Blood Marrow Transplant. 15:30–38. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Fenaux P, Mufti GJ, Hellstrom-Lindberg E, Santini V, Finelli C, Giagounidis A, Schoch R, Gattermann N, Sanz G, List A, et al: Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: A randomised, open-label, phase III study. Lancet Oncol. 10:223–232. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Gore SD, Fenaux P, Santini V, Bennett JM, Silverman LR, Seymour JF, Hellström-Lindberg E, Swern AS, Beach CL and List AF: A multivariate analysis of the relationship between response and survival among patients with higher-risk myelodysplastic syndromes treated within azacitidine or conventional care regimens in the randomized AZA-001 trial. Haematologica. 98:1067–1072. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Kantarjian H, Issa JP, Rosenfeld CS, Bennett JM, Albitar M, DiPersio J, Klimek V, Slack J, de Castro C, Ravandi F, et al: Decitabine improves patient outcomes in myelodysplastic syndromes: Results of a phase III randomized study. Cancer. 106:1794–1803. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Zwierzina H, Suciu S, Loeffler-Ragg J, Neuwirtova R, Fenaux P, Beksac M, Harousseau J, Nuessler V, Cermak J, Solbu G, et al: Low-dose cytosine arabinoside (LD-AraC) vs. LD-AraC plus granulocyte/macrophage colony stimulating factor vs. LD-AraC plus Interleukin-3 for myelodysplastic syndrome patients with a high risk of developing acute leukemia: Final results of a randomized phase III study (06903) of the EORTC Leukemia Cooperative Group. Leukemia. 19:1929–1933. 2005. View Article : Google Scholar : PubMed/NCBI

11 

Beran M, Shen Y, Kantarjian H, O'Brien S, Koller CA, Giles FJ, Cortes J, Thomas DA, Faderl S, Despa S and Estey EH: High-dose chemotherapy in high-risk myelodysplastic syndrome: Covariate-adjusted comparison of five regimens. Cancer. 92:1999–2015. 2001. View Article : Google Scholar : PubMed/NCBI

12 

List A, Dewald G, Bennett J, Giagounidis A, Raza A, Feldman E, Powell B, Greenberg P, Thomas D, Stone R, et al: Lenalidomide in the myelodysplastic syndrome with chromosome 5q deletion. N Engl J Med. 355:1456–1465. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Badros AZ: Lenalidomide in myeloma-a high-maintenance friend. N Engl J Med. 366:1836–1838. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Lallemand-Breitenbach V, Zhu J, Chen Z and de Thé H: Curing APL through PML/RARA degradation by As2O3. Trends Mol Med. 18:36–42. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Shen ZX, Chen GQ, Ni JH, Li XS, Xiong SM, Qiu QY, Zhu J, Tang W, Sun GL, Yang KQ, et al: Use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL): II. Clinical efficacy and pharmacokinetics in relapsed patients. Blood. 89:3354–3360. 1997.PubMed/NCBI

16 

Schiller GJ, Slack J, Hainsworth JD, Mason J, Saleh M, Rizzieri D, Douer D and List AF: Phase II multicenter study of arsenic trioxide in patients with myelodysplastic syndromes. J Clin Oncol. 24:2456–2464. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Vey N, Bosly A, Guerci A, Feremans W, Dombret H, Dreyfus F, Bowen D, Burnett A, Dennis M, Ribrag V, et al: Arsenic trioxide in patients with myelodysplastic syndromes: A phase II multicenter study. J Clin Oncol. 24:2465–2471. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Takahashi S: Combination therapy with arsenic trioxide for hematological malignancies. Anticancer Agents Med Chem. 10:504–510. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Xia J, Li Y, Yang Q, Mei C, Chen Z, Bao B, Ahmad A, Miele L, Sarkar FH and Wang Z: Arsenic trioxide inhibits cell growth and induces apoptosis through inactivation of notch signaling pathway in breast cancer. Int J Mol Sci. 13:9627–9641. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Hoffman E and Mielicki WP: Arsenic trioxide: Impact on the growth and differentiation of cancer cells and possible use in cancer therapy. Postepy Hig Med Dosw (Online). 67:817–827. 2013.(In Polish). View Article : Google Scholar : PubMed/NCBI

21 

Zhao H, Guo W, Peng C, Ji T and Lu X: Arsenic trioxide inhibits the growth of adriamycin resistant osteosarcoma cells through inducing apoptosis. Mol Biol Rep. 37:2509–2515. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Li C, Qu X, Xu W, Qu N, Mei L, Liu Y, Wang X, Yu X, Liu Z, Nie D, et al: Arsenic trioxide induces cardiac fibroblast apoptosis in vitro and in vivo by up-regulating TGF-β1 expression. Toxicol Lett. 219:223–230. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Yedjou C, Tchounwou P, Jenkins J and McMurray R: Basic mechanisms of arsenic trioxide (ATO)-induced apoptosis in human leukemia (HL-60) cells. J Hematol Oncol. 3:282010. View Article : Google Scholar : PubMed/NCBI

24 

Han R, Rostami-Yazdi M, Gerdes S and Mrowietz U: Triptolide in the treatment of psoriasis and other immune-mediated inflammatory diseases. Br J Clin Pharmacol. 74:424–436. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Deng J and Jin J: Study of triptolide-induced apoptosis in MUTZ-1 cells and its allied mechanism. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 13:434–439. 2005.(In Chinese). PubMed/NCBI

26 

Mujumdar N, Mackenzie TN, Dudeja V, Chugh R, Antonoff MB, Borja-Cacho D, Sangwan V, Dawra R, Vickers SM and Saluja AK: Triptolide induces cell death in pancreatic cancer cells by apoptotic and autophagic pathways. Gastroenterology. 139:598–608. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Wu PP, Liu KC, Huang WW, Ma CY, Lin H, Yang JS and Chung JG: Triptolide induces apoptosis in human adrenal cancer NCI-H295 cells through a mitochondrial-dependent pathway. Oncol Rep. 25:551–557. 2011.PubMed/NCBI

28 

Meng HT, Zhu L, Ni WM, You LS, Jin J and Qian WB: Triptolide inhibits the proliferation of cells from lymphocytic leukemic cell lines in association with downregulation of NF-κB activity and miR-16-1*. Acta Pharmacol Sin. 32:503–511. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Sekeres MA, Maciejewski JP, Erba HP, Afable M, Englehaupt R, Sobecks R, Advani A, Seel S, Chan J and Kalaycio ME: A Phase 2 study of combination therapy with arsenic trioxide and gemtuzumab ozogamicin in patients with myelodysplastic syndromes or secondary acute myeloid leukemia. Cancer. 117:1253–1261. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

31 

Chou TC and Talalay P: Quantitative analysis of dose-effect relationships: The combined effects of multiple drugs or enzyme inhibitors. Adv Enzym Regul. 22:27–55. 1984. View Article : Google Scholar

32 

Takahashi S, Harigae H, Yokoyama H, Ishikawa I, Abe S, Imaizumi M, Sasaki T and Kaku M: Synergistic effect of arsenic trioxide and flt3 inhibition on cells with flt3 internal tandem duplication. Int J Hematol. 84:256–261. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Li M, Ding Y, Mu Y, Ao J and Chen X: Molecular cloning and characterization of caspase-3 in large yellow croaker (Pseudosciaena crocea). Fish Shellfish Immunol. 30:910–916. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Clawson KA, Borja-Cacho D, Antonoff MB, Saluja AK and Vickers SM: Triptolide and TRAIL combination enhances apoptosis in cholangiocarcinoma. J Surg Res. 163:244–249. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Spampanato C, De Maria S, Sarnataro M, Giordano E, Zanfardino M, Baiano S, Cartenì M and Morelli F: Simvastatin inhibits cancer cell growth by inducing apoptosis correlated to activation of Bax and down-regulation of BCL-2 gene expression. International J Onco. 40:935–941. 2012.

36 

Abdelrahman IY, Helwa R, Elkashef H and Hassan NH: Induction of P3NS1 myeloma cell death and cell cycle arrest by simvastatin and/or γ-radiation. Asian Pac J Cancer Prev. 16:7103–7110. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Park MT, Kang YH, Park IC, Kim CH, Lee YS, Chung HY and Lee SJ: Combination treatment with arsenic trioxide and phytosphingosine enhances apoptotic cell death in arsenic trioxide-resistant cancer cells. Mol Cancer Ther. 6:82–92. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2016
Volume 14 Issue 5

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
Hua HY, Gao HQ, Sun AN, Cen JN and Wu LL: Arsenic trioxide and triptolide synergistically induce apoptosis in the SKM‑1 human myelodysplastic syndrome cell line. Mol Med Rep 14: 4180-4186, 2016
APA
Hua, H., Gao, H., Sun, A., Cen, J., & Wu, L. (2016). Arsenic trioxide and triptolide synergistically induce apoptosis in the SKM‑1 human myelodysplastic syndrome cell line. Molecular Medicine Reports, 14, 4180-4186. https://doi.org/10.3892/mmr.2016.5779
MLA
Hua, H., Gao, H., Sun, A., Cen, J., Wu, L."Arsenic trioxide and triptolide synergistically induce apoptosis in the SKM‑1 human myelodysplastic syndrome cell line". Molecular Medicine Reports 14.5 (2016): 4180-4186.
Chicago
Hua, H., Gao, H., Sun, A., Cen, J., Wu, L."Arsenic trioxide and triptolide synergistically induce apoptosis in the SKM‑1 human myelodysplastic syndrome cell line". Molecular Medicine Reports 14, no. 5 (2016): 4180-4186. https://doi.org/10.3892/mmr.2016.5779