Open Access

Piperlongumine induces apoptosis and autophagy in leukemic cells through targeting the PI3K/Akt/mTOR and p38 signaling pathways

  • Authors:
    • Hongfei Wang
    • Yongqiang Wang
    • Hongmei Gao
    • Bing Wang
    • Lin Dou
    • Yin Li
  • View Affiliations

  • Published online on: November 29, 2017     https://doi.org/10.3892/ol.2017.7498
  • Pages: 1423-1428
  • Copyright: © Wang 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

Piperlongumine is an alkaloid compound extracted from Piper longum L. It is a chemical substance with various pharmacological effects and medicinal value, including anti‑tumor, lipid metabolism regulatory, antiplatelet aggregation and analgesic properties. The present study aimed to understand whether piperlongumine induces the apoptosis and autophagy of leukemic cells, and to identify the mechanism involved. Cell viability and autophagy were detected using MTT, phenazine methyl sulfate and trypan blue exclusion assays. The apoptosis rate was calculated using flow cytometry. The protein expression levels of microtubule‑associated protein 1A/1B‑light chain 3, Akt and mechanistic target of rapamycin (mTOR) were measured using western blotting. The cell growth of leukemic cells was completely inhibited following treatment with piperlongumine, and marked apoptosis was also induced. Dead cells as a result of autophagy were stained using immunofluorescence and observed under a light microscope. Phosphoinositide 3‑kinase (PI3K)/Akt/mTOR signaling was suppressed by treatment with piperlongumine, while p38 signaling and caspase‑3 activity were induced by treatment with piperlongumine. It was concluded that piperlongumine induces apoptosis and autophagy in leukemic cells through targeting the PI3K/Akt/mTOR and p38 signaling pathways.

Introduction

Leukemia is a kind of malignant tumor of the hemopoietic system posing a great threat to human health (1). The mortality rate ranks sixth in males and eighth in females among all malignant tumors in China in all age groups, and first among children and adults under 35 years of age (2). There is a higher number of patients with acute myeloid leukemia (AML) than with acute lymphoblastic leukemia (ALL) (AML:ALL=2.67:1.00); AML is mostly reported in young adults (76.2%) and has a negative effect on health (3). In contrast to solid tumors, leukemia is a kind of ‘liquid tumor’ that cannot be removed by surgery (4). At present, chemotherapy is still the most widely used method in the treatment of patients with leukemia due to the difficulty in matching bone marrow for transplantation and the high price of molecular targeted therapy (5). However, the current research focus is to identify novel chemotherapy drugs with strong activity and low toxicity from natural drugs that are able to overcome chemotherapeutic resistance and adverse side reactions (6).

Following cell apoptosis, autophagy has become the hot research topic as type-II programmed death in recent years (5). Autophagy involves the formation of autophagic vacuoles, their integration with lysosomes and the degradation of the contents of these autophagosomes (7). In contrast to cell apoptosis, autophagy has a dual regulatory function in cell survival, which serves an important role in the occurrence and development of tumors (8). Previous studies have shown that intracellular autophagy in certain tumor cells can kill these cells and maintain the normal internal body environment in the early stages of cancer (9,10). However, upon the formation of tumors, autophagy promotes apoptosis by removing damaged organelles, so as to reduce the growth of tumor cells (11,12). Therefore, researching the mechanism of autophagy development has a good prospect in clinical application for the prevention and treatment of cancer (13).

Piperlongumine (Fig. 1) belongs to the family of alkaloids, and exhibits a variety of pharmacological effects and pharmacological activities, including antiplatelet aggregation, analgesia and antifungal properties (14). In addition, piperlongumine has a marked cytotoxic effect on tumor cells and can regulate blood lipid metabolism in hyperlipidemic rats (14). Previous studies have shown that piperlongumine has a specific cytotoxic effect on a variety of tumor cells, but exhibits low toxicity towards normal cells (14,15). Therefore, piperlongumine is an extract of a herb used in traditional Chinese medicine, which has the potential to selectively kill tumors (14). The pharmacological effects of piperlongumine mainly include antitumor, antiplatelet aggregation, analgesic, antifungal, anti-schistosomiasis, anti-anxiety and anti-depression effects (16). The aim of the present study was to investigate whether piperlongumine induces apoptosis and autophagy in leukemic cells, and to explore the underlying molecular mechanisms.

Materials and methods

Cell lines and mice

Leukemic monocytic lymphoma U937 cells were purchased from the Cell Bank of Type Culture Collection of the Chinese Academy of Sciences (Shanghai, China) and cultured in RPMI-1640 medium (pH 6.8) supplemented with 10% fetal bovine serum (both from Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) and antibiotics (50 IU/ml penicillin G and 50 µg/ml streptomycin), and were incubated at 37°C in a humidified atmosphere containing 5% CO2.

Cell viability assay and immunofluorescence

The effect of piperlongumine on the viability of U937 cells was evaluated. U937 cells (2.5–5.0×104 cells/200 µl of RPMI-1640 medium/well) were seeded in 96-well tissue culture plates and incubated with piperlongumine (0–20 µM) for 48 h at 37°C in the presence of 5% CO2. MTT (2.0 mg/ml) and phenazine methyl sulfate (0.92 mg/ml) were added into every well and incubated at 37°C for 3 h. The absorbance was measured at 490 nm in an ELISA reader (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Cells (at 60–70% confluence) were seeded in 6-well tissue culture plates and treated with piperlongumine (0–20 µM) for 48 h at 37°C in the presence of 5% CO2.

U937 cells treated with piperlongumine (0, 5, 10 and 20 µM) were washed with PBS and fixed using 4% paraformaldehyde for 15 min at room temperature. Cells were incubated with Triton X-100 (0.1%) for 10 min at room temperature and then incubated with anti-LC3 antibodies (cat. no. 3868; 1:3,000; Cell Signaling Technology, Inc., Beverly, MA, USA) at 4°C overnight. Cells were incubated with Alexa Fluor 488-labeled goat anti-rabbit secondary antibody (Thermo Fisher Scientific, Inc.) for 1 h at room temperature in darkness. Cells were observed using a Nikon Eclipse E800 fluorescence microscope and NIS-Elements 4.0 software (all from Nikon; Tokyo, Japan).

Apoptosis assay

U937 cells (2.5×105 cells/ml) were incubated with piperlongumine (0–20 µM) for 48 h at 37°C in the presence of 5% CO2. Cells were washed twice with PBS and resuspended in 100 µl of Annexin V binding buffer (pH 7.4) (BD Biosciences, Franklin Lakes, NJ, USA). Then, annexin V-fluorescein isothiocyanate (BD Biosciences) was added and incubated for 15 min under dark conditions. Propidium iodide (0.1 µg/ml; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) was added just prior to signal acquisition. The apoptosis rate was acquired using a FACSAria flow cytometer (BD Biosciences) and analyzed with FACSDiva 7.6.1 software (BD Biosciences).

Western blotting

Total cellular proteins were isolated from U937 cells (2.5×105 cells/ml) incubated with piperlongumine (0–20 µM) for 48 h at 37°C in the presence of 5% CO2. Cells were lysed using radioimmunoprecipitation assay (RIPA) buffer (Beyotime Institute of Biotechnology, Jiangsu, China) and centrifuged for 10 min at 4°C at 10,000 × g. Protein concentration was estimated using the bicinchoninic acid method (Beyotime Institute of Biotechnology). Electrophoretic separations were carried out on 10% SDS-PAGE, and the proteins were then electrotransferred onto a polyvinylidene fluoride membrane. Blots were blocked for 1 h at 37°C in TBS containing 0.01% Tween-20 (TBST) and 5% skimmed milk, and probed overnight at 4°C with appropriate primary antibodies: Anti-microtubule-associated protein 1A/1B-light chain 3 (LC3-I; cat. no. 3868; 1:3,000), anti-phosphorylated (p)-Akt (cat. no. 4228; 1:3,000), anti-Akt (cat. no. 6211; 1:3,000), anti-p-mechanistic target of rapamycin (mTOR; cat. no. 2974; 1:3,000), anti-p-p38 (cat. no. 4511, 1:2,000) and anti-β-actin (cat. no. 4970, 1:2,000) (all from Cell Signaling Technology, Inc., Beverly, MA, USA) antibodies. Next, the membranes were washed with TBST and incubated with anti-rabbit IgG, HRP-linked antibody (cat. no. 7074, 1:5,000, Cell Signaling Technology, Inc.) at 37°C for 1 h. Proteins were visualized using BeyoECL Plus (Beyotime Institute of Biotechnology) and analyzed uisng Bio-Rad Laboratories Quantity One software 3.0 (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Measurement of caspase activity

U937 cells (2.5–5.0×104 cells/200 µl of RPMI-1640 medium/well) were seeded in 96-well tissue culture plates and incubated with piperlongumine (0–20 µM) for 48 h at 37°C in the presence of 5% CO2. Cells were lysed using RIPA buffer (Beyotime Institute of Biotechnology) and centrifuged for 10 min at 4°C at 10,000 × g. Protein concentration was estimated using the BCA Protein Assay kit (Beyotime Institute of Biotechnology). U937 cell lysates (5 µg) were incubated in 50 µl of reaction buffer and acetyl-Asp-Glu-Val-Asp p-nitroanilide (p-NA) (Beyotime Institute of Biotechnology) at 37°C for 5 h for determination of caspase-3 activity. The emission of pNA was measured at 405 nm in an ELISA reader (Bio-Rad Laboratories, Inc.).

Statistical analysis

All experiments were repeated independently at least three times using SPSS 17.0 (SPSS, Inc., Chicago, IL, USA). The values were expressed as mean ± standard error of the mean, and statistical analyses were performed with a two-way analysis of variance followed by the Student-Newman-Keuls test.

Results

Piperlongumine suppresses cell proliferation in leukemic cells

The anticancer effect of piperlongumine in terms of suppressing the proliferation of leukemic cells was evaluated. An MTT assay was used to analyze the change in cell proliferation in U937 cells. As shown in Fig. 2, 0–20 µM of piperlongumine could suppress the proliferation of U937 cells in a time- and dose-dependent manner. At 20 µM, piperlongumine significantly suppressed the proliferation of U937 cells at 24 h, while 10 or 20 µM of piperlongumine significantly suppressed the proliferation of U937 cells at 48 h (Fig. 2).

Piperlongumine induces apoptosis in leukemic cells

To further confirm the role of apoptosis in the effect of piperlongumine on leukemic cells, the apoptosis rate was evaluated using flow cytometry. The results revealed that 0–20 µM of piperlongumine induced the apoptosis of U937 cells in a dose-dependent manner (Fig. 3). Treatment with 10 or 20 µM of piperlongumine significantly induced the apoptosis of U937 cells, compared with that of U937 cells incubated with 0 µM of piperlongumine (Fig. 3).

Piperlongumine induces autophagy in leukemic cells

The autophagy of U937 cells was measured by fluorescence microscopy targeted at LC3. An increase in the autophagy of U937 cells incubated with 0–20 µM of piperlongumine was observed (Fig. 4). Treatment with 10 or 20 µM of piperlongumine activated the autophagy of U937 cells, compared with that of U937 cells incubated with 0 µM of piperlongumine (Fig. 4).

Piperlongumine induces LC3-I expression in leukemic cells

Upon co-incubation of U937 cells with 0–20 µM of piperlongumine for 48 h, the protein expression of LC3-I was explored using western blotting. The protein expression of LC3-I was induced by 0–20 µM of piperlongumine in a dose-dependent manner (Fig. 5). In particular, 10 or 20 µM of piperlongumine significantly increased LC3-I protein expression in U937 cells, compared with that of U937 cells incubated with 0 µM of piperlongumine (Fig. 5).

Piperlongumine reduces Akt levels in leukemic cells

To analyze the mechanism of piperlongumine on autophagy, the phosphorylation of Akt in U937 cells was determined by western blotting. As shown in Fig. 6, a dose-dependent decrease in the ratio of p-Akt/Akt protein expression was detected in U937 cells following treatment with 0–20 µM of piperlongumine. Particularly, 10 or 20 µM of piperlongumine significantly reduced the p-Akt/Akt ratio in U937 cells, compared with that of U937 cells incubated with 0 µM of piperlongumine (Fig. 6A and B).

Piperlongumine reduces mTOR levels in leukemic cells

To further analyze the mechanism of piperlongumine on autophagy, the protein expression of p-mTOR was measured. Fig. 6 shows that 0–20 µM of piperlongumine inhibited p-mTOR protein expression in U937 cells in a dose-dependent manner. Treatment with 10 or 20 µM of piperlongumine significantly inhibited p-mTOR protein expression in U937 cells, compared with that of the 0 µM of piperlongumine group (Fig. 6A and C).

Piperlongumine induces the phosphorylation of p38 protein in leukemic cells

To investigate the protein expression of p38, U937 cells were incubated with piperlongumine (0–20 µM) for 48 h. Treatment with 0–20 µM of piperlongumine induced the protein expression of p-p38 in U937 cells (Fig. 6A and D). Treatment with 10 or 20 µM of piperlongumine significantly induced p-p38 protein expression in U937 cells, compared with that of U937 cells incubated with 0 µM of piperlongumine (Fig. 6A and D).

Piperlongumine induces caspase-3 activity in leukemic cells

In order to assess the role of caspase-3 activity in U937 cells, the cells were co-incubated with 0–20 µM of piperlongumine for 48 h. As shown in Fig. 7, 0–20 µM of piperlongumine increased the caspase-3 activity of U937 cells in a dose-dependent manner. Particularly, treatment with 10 or 20 µM of piperlongumine significantly increased the caspase-3 activity of U937 cells, compared with that of U937 cells incubated with 0 µM of piperlongumine (Fig. 7).

Discussion

Leukemia is a type of malignant tumor of the hemopoietic system posing a severe threat to human health, and its mortality rate ranks first among all malignant tumors among children and adults under 35 years of age (17). Its morbidity and mortality are increasing, and it is urgently required to identify novel therapeutics against leukemic cells (18). Dhillon et al reported that piperlongumine induces pancreatic cancer cell death (19). The present data clearly demonstrated that piperlongumine significantly suppresses cell proliferation and induces apoptosis in U937 cells.

Autophagy is a conserved self-degradation system in eukaryotic cells, which is involved in numerous physiological and pathological processes (13). The autophagosome is the typical characteristic of autophagy, and the regulation of the formation and degradation of the autophagosome is the main regulation of autophagy (8). Due to the dual characteristics of autophagy in promoting both cell growth and death, autophagy determines cell survival to certain extent (20). Autophagy is closely associated with tumors, and is involved in tumor development, metastasis and drug resistance (12). Targeted autophagy may become a new strategy for the treatment of cancer and drug resistance (21). Further study of autophagy in leukemic cells will clarify the mechanism of induction and regulation of autophagy in leukemic cells, and may provide a new treatment target and strategy for leukemic cells (22). In the present study, it was demonstrated that piperlongumine induces autophagy and induces expression of LC3-I protein in leukemic cells. Makhov et al demonstrated that piperlongumine promotes autophagy in a xenograft mouse model through inhibition of Akt/mTOR signaling, and mediates cancer cell death (23). Wang et al (24) showed that Piperlongumine induces autophagy of cancer cell by targeting p38 signaling.

The main function of mTOR is to inhibit the occurrence of self-autophagy via two mechanisms: i) mTOR regulates the transcription and translation of autophagy-associated genes through the activation of downstream effectors, which affects the signal transduction pathway; and ii) inhibition of mTOR can induce the occurrence of autophagy (25). PI3K/Akt is the upstream signaling pathway that activates mTOR (26). PI3K is important in cancer development, while the serine-threonine protein kinase Akt is the downstream effector of PI3K and is involved in the regulation of various biological processes, including cell metastasis, growth, development, apoptosis, and regulation of gene transcription, protein synthesis and nutritional metabolism (27). Akt may be key to the inhibition and survival signal pathway and inhibit autophagy by phosphorylating mTOR and contacting PI3K/Akt (27). The activated mTOR signal transduction pathway can inhibit the apoptosis and autophagy induced by various factors, which leads to cell cycle progression, cell growth and proliferation. It is also associated with angiogenesis, thus serving an important role in the formation, invasion and metastasis of tumors (28,29). Numerous tumors exhibit mutated genes coding for proteins involved in mTOR signaling, and the resulting over-activated mTOR signaling pathway is mainly caused by abnormal expression of these proteins (28,29). Previous studies have demonstrated that breast cancer, leukemia, small cell lung cancer, urinary system tumors and other diseases progress through PI3K/Akt/mTOR signaling (22,27). The present study has demonstrated that piperlongumine significantly reduces Akt/mTOR signaling in U937 cells. Wang et al previously demonstrated that piperlongumine induces apoptosis and autophagy through inhibition of the PI3K/Akt/mTOR signaling pathway in human lung cancer cells (16).

The p38/mitogen-activated protein kinase (MAPK) signaling pathway is involved in the activation of autophagy in macrophages (30). MAPK p38 mainly inhibits autophagy, and the effect of the p38/MAPK signaling pathway is markedly complex in the development of cells (31). The activation of this pathway leads to the inhibition of cell proliferation. p38 can also induce the arrest of the cell cycle into the stationary phase and promote DNA repair against the DNA damage induced by chemotherapy (32). In particular, p38 has been reported to exhibit anti-apoptotic properties in various cell lines, and may have a direct effect on tumor infiltration and metastasis (33). The present study noticed that piperlongumine significantly induced p-p38 protein expression and increased caspase-3 activity in U937 cells. Xiong et al reported that piperlongumine induces the autophagic death of primary myeloid leukemia cells through the p38/c-Jun N-terminal kinase signaling pathway (34).

In summary, the present study has demonstrated that piperlongumine significantly suppresses cell proliferation and induces apoptosis in U937 cells. Preferentially, piperlongumine significantly induced the autophagy of U937 cells through targeting the PI3K/Akt/mTOR and p38 signaling pathways. The present data suggest that piperlongumine could be applied in the treatment of leukemic cells.

References

1 

Hefazi M, Siddiqui M, Patnaik M, Wolanskyj A, Alkhateeb H, Zblewski D, Elliott M, Hogan W, Litzow M and Al-Kali A: Prognostic impact of combined NPM1+/FLT3-genotype in patients with acute myeloid leukemia with intermediate risk cytogenetics stratified by age and treatment modalities. Leuk Res: Sep. 3:2015.(Epub ahead of print).

2 

Ong E, Szedlak A, Kang Y, Smith P, Smith N, McBride M, Finlay D, Vuori K, Mason J, Ball ED, et al: A scalable method for molecular network reconstruction identifies properties of targets and mutations in acute myeloid leukemia. J Comput Biol. 22:266–288. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Urtishak KA, Edwards AY, Wang LS, Hudome A, Robinson BW, Barrett JS, Cao K, Cory L, Moore JS, Bantly AD, et al: Potent obatoclax cytotoxicity and activation of triple death mode killing across infant acute lymphoblastic leukemia. Blood. 121:2689–2703. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Wang F, Liu Z, Zeng J, Zhu H, Li J, Cheng X, Jiang T, Zhang L, Zhang C, Chen T, et al: Metformin synergistically sensitizes FLT3-ITD-positive acute myeloid leukemia to sorafenib by promoting mTOR-mediated apoptosis and autophagy. Leuk Res. 39:1421–1427. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Kim Y, Eom JI, Jeung HK, Jang JE, Kim JS, Cheong JW, Kim YS and Min YH: Induction of cytosine arabinoside-resistant human myeloid leukemia cell death through autophagy regulation by hydroxychloroquine. Biomed Pharmacother. 73:87–96. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Zauli G, Celeghini C, Melloni E, Voltan R, Ongari M, Tiribelli M, di Iasio MG, Lanza F and Secchiero P: The sorafenib plus nutlin-3 combination promotes synergistic cytotoxicity in acute myeloid leukemic cells irrespectively of FLT3 and p53 status. Haematologica. 97:1722–1730. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Martelli AM, Lonetti A, Amadori S, McCubrey JA and Chiarini F: Enhancing the effectiveness of nucleoside analogs with mTORC1 blockers to treat acute myeloid leukemia patients. Cell Cycle. 12:1815–1816. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Liu C, Xu P, Chen D, Fan X, Xu Y, Li M, Yang X and Wang C: Roles of autophagy-related genes Beclin-1 and LC3 in the development and progression of prostate cancer and benign prostatic hyperplasia. Biomed Rep. 1:855–860. 2013.PubMed/NCBI

9 

Liu L, He J, Wei X, Wan G, Lao Y, Xu W, Li Z, Hu H, Hu Z, Luo X, et al: MicroRNA-20a-mediated loss of autophagy contributes to breast tumorigenesis by promoting genomic damage and instability. Oncogene. Jun 19–2017.(Epub ahead of print). View Article : Google Scholar

10 

Qiu S, Sun L, Jin Y, An Q, Weng C and Zheng J: Silencing of BAG3 promotes the sensitivity of ovarian cancer cells to cisplatin via inhibition of autophagy. Oncol Rep. 38:309–316. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Brigger D, Proikas-Cezanne T and Tschan MP: WIPI-dependent autophagy during neutrophil differentiation of NB4 acute promyelocytic leukemia cells. Cell Death Dis. 5:e13152014. View Article : Google Scholar : PubMed/NCBI

12 

Xie N, Zhong L, Liu L, Fang Y, Qi X, Cao J, Yang B, He Q and Ying M: Autophagy contributes to dasatinib-induced myeloid differentiation of human acute myeloid leukemia cells. Biochem Pharmacol. 89:74–85. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Zhang SP, Niu YN, Yuan N, Zhang AH, Chao D, Xu QP, Wang LJ, Zhang XG, Zhao WL, Zhao Y and Wang JR: Role of autophagy in acute myeloid leukemia therapy. Chin J Cancer. 32:130–135. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Jin HO, Park JA, Kim HA, Chang YH, Hong YJ, Park IC and Lee JK: Piperlongumine downregulates the expression of HER family in breast cancer cells. Biochem Biophys Res Commun. 486:1083–1089. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Bullova P, Cougnoux A, Abunimer L, Kopacek J, Pastorekova S and Pacak K: Hypoxia potentiates the cytotoxic effect of piperlongumine in pheochromocytoma models. Oncotarget. 7:40531–40545. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Wang F, Mao Y, You Q, Hua D and Cai D: Piperlongumine induces apoptosis and autophagy in human lung cancer cells through inhibition of PI3K/Akt/mTOR pathway. Int J Immunopathol Pharmacol. 28:362–373. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Malkan UY, Gunes G, Isik A, Eliacik E, Etgul S, Aslan T, Balaban MS, Haznedaroglu IC, Demiroglu H, Goker H, et al: Rebound thrombocytosis following induction chemotherapy is an independent predictor of a good prognosis in acute myeloid leukemia patients attaining first complete remission. Acta Haematol. 134:32–37. 2015. View Article : Google Scholar : PubMed/NCBI

18 

O'Hear C, Inaba H, Pounds S, Shi L, Dahl G, Bowman WP, Taub JW, Pui CH, Ribeiro RC, Coustan-Smith E, et al: Gemtuzumab ozogamicin can reduce minimal residual disease in patients with childhood acute myeloid leukemia. Cancer. 119:4036–4043. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Dhillon H, Chikara S and Reindl KM: Piperlongumine induces pancreatic cancer cell death by enhancing reactive oxygen species and DNA damage. Toxicol Rep. 1:309–318. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Yu J, Lan L, Lewin SJ, Rogers SA, Roy A, Wu X, Gao P, Karanicolas J, Aubé J, Sun B and Xu L: Identification of novel small molecule Beclin 1 mimetics activating autophagy. Oncotarget. May 18–2017.(Epub ahead of print).

21 

Tanios R, Bekdash A, Kassab E, Stone E, Georgiou G, Frankel AE and Abi-Habib RJ: Human recombinant arginase I(Co)-PEG5000 [HuArgI(Co)-PEG5000]-induced arginine depletion is selectively cytotoxic to human acute myeloid leukemia cells. Leuk Res. 37:1565–1571. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Liu LL, Long ZJ, Wang LX, Zheng FM, Fang ZG, Yan M, Xu DF, Chen JJ, Wang SW, Lin DJ and Liu Q: Inhibition of mTOR pathway sensitizes acute myeloid leukemia cells to aurora inhibitors by suppression of glycolytic metabolism. Mol Cancer Res. 11:1326–1336. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Makhov P, Golovine K, Teper E, Kutikov A, Mehrazin R, Corcoran A, Tulin A, Uzzo RG and Kolenko VM: Piperlongumine promotes autophagy via inhibition of Akt/mTOR signalling and mediates cancer cell death. Br J Cancer. 110:899–907. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Wang Y, Wang JW, Xiao X, Shan Y, Xue B, Jiang G, He Q, Chen J, Xu HG, Zhao RX, et al: Piperlongumine induces autophagy by targeting p38 signaling. Cell Death Dis. 4:e8242013. View Article : Google Scholar : PubMed/NCBI

25 

Wang KF, Yang H, Jiang WQ, Li S and Cai YC: Puquitinib mesylate (XC-302) induces autophagy via inhibiting the PI3K/AKT/mTOR signaling pathway in nasopharyngeal cancer cells. Int J Mol Med. 36:1556–1562. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Huang WR, Chiu HC, Liao TL, Chuang KP, Shih WL and Liu HJ: Avian reovirus protein p17 functions as a nucleoporin Tpr suppressor leading to activation of p53, p21 and PTEN and inactivation of PI3K/AKT/mTOR and ERK signaling pathways. PLoS One. 10:e01336992015. View Article : Google Scholar : PubMed/NCBI

27 

Zhou ZW, Li XX, He ZX, Pan ST, Yang Y, Zhang X, Chow K, Yang T, Qiu JX, Zhou Q, et al: Induction of apoptosis and autophagy via sirtuin1- and PI3 K/Akt/mTOR-mediated pathways by plumbagin in human prostate cancer cells. Drug Des Devel Ther. 9:1511–1554. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Tsai JP, Lee CH, Ying TH, Lin CL, Lin CL, Hsueh JT and Hsieh YH: Licochalcone A induces autophagy through PI3K/Akt/mTOR inactivation and autophagy suppression enhances licochalcone A-induced apoptosis of human cervical cancer cells. Oncotarget. 6:28851–28866. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Sun H, Wang Z and Yakisich JS: Natural products targeting autophagy via the PI3K/Akt/mTOR pathway as anticancer agents. Anticancer Agents Med Chem. 13:1048–1056. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Li JP, Yang YX, Liu QL, Pan ST, He ZX, Zhang X, Yang T, Chen XW, Wang D, Qiu JX and Zhou SF: The investigational Aurora kinase A inhibitor alisertib (MLN8237) induces cell cycle G2/M arrest, apoptosis, and autophagy via p38 MAPK and Akt/mTOR signaling pathways in human breast cancer cells. Drug Des Devel Ther. 9:1627–1652. 2015.PubMed/NCBI

31 

Henson SM, Lanna A, Riddell NE, Franzese O, Macaulay R, Griffiths SJ, Puleston DJ, Watson AS, Simon AK, Tooze SA and Akbar AN: p38 signaling inhibits mTORC1-independent autophagy in senescent human CD8+ T cells. J Clin Invest. 124:4004–4016. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Liu J, Chang F, Li F, Fu H, Wang J, Zhang S, Zhao J and Yin D: Palmitate promotes autophagy and apoptosis through ROS-dependent JNK and p38 MAPK. Biochem Biophys Res Commun. 463:262–267. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Qin Y, Zhou ZW, Pan ST, He ZX, Zhang X, Qiu JX, Duan W, Yang T and Zhou SF: Graphene quantum dots induce apoptosis, autophagy, and inflammatory response via p38 mitogen-activated protein kinase and nuclear factor-κB mediated signaling pathways in activated THP-1 macrophages. Toxicology. 327:62–76. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Xiong XX, Liu JM, Qiu XY, Pan F, Yu SB and Chen XQ: Piperlongumine induces apoptotic and autophagic death of the primary myeloid leukemia cells from patients via activation of ROS-p38/JNK pathways. Acta Pharmacol Sin. 36:362–374. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2018
Volume 15 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang H, Wang Y, Gao H, Wang B, Dou L and Li Y: Piperlongumine induces apoptosis and autophagy in leukemic cells through targeting the PI3K/Akt/mTOR and p38 signaling pathways. Oncol Lett 15: 1423-1428, 2018
APA
Wang, H., Wang, Y., Gao, H., Wang, B., Dou, L., & Li, Y. (2018). Piperlongumine induces apoptosis and autophagy in leukemic cells through targeting the PI3K/Akt/mTOR and p38 signaling pathways. Oncology Letters, 15, 1423-1428. https://doi.org/10.3892/ol.2017.7498
MLA
Wang, H., Wang, Y., Gao, H., Wang, B., Dou, L., Li, Y."Piperlongumine induces apoptosis and autophagy in leukemic cells through targeting the PI3K/Akt/mTOR and p38 signaling pathways". Oncology Letters 15.2 (2018): 1423-1428.
Chicago
Wang, H., Wang, Y., Gao, H., Wang, B., Dou, L., Li, Y."Piperlongumine induces apoptosis and autophagy in leukemic cells through targeting the PI3K/Akt/mTOR and p38 signaling pathways". Oncology Letters 15, no. 2 (2018): 1423-1428. https://doi.org/10.3892/ol.2017.7498