Abstract
Pseudolaric acid B (PAB), a diterpene acid isolated from the root bark of Pseudolarix kaempferi Gordon, exerts anti-tumor effects in several cancer cell lines. Our previous study showed that PAB mainly induced senescence via p53-p21 activation rather than apoptosis in suppression of the growth of human lung cancer A549 cells (p53 wild-type). In p53-null human lung cancer H1299 cells, however, PAB caused apoptosis without senescence. In this study we investigated what mechanism was responsible for the switch from senescence to apoptosis in PAB-treated human lung cancer cell lines. Senescent cells were examined by SA-β-gal staining. Glucose uptake and the apoptosis ratio were assessed using a FACScan flow cytometer. Commercial assay kits were used to measure the levels of ATP and lactate. Transfection of siRNA was used to knockdown the expression of p53 or p21. Western blot analysis was applied to measure the protein expression levels. In p53 wild-type A549 cells, PAB (20 μmol/L) caused senescence, and time-dependently increased glucose utilization; knockdown of p53 or p21 significantly decreased the uptake and metabolism of glucose but elevated PAB-induced apoptosis. Inhibition of glucose utilization using a glycolytic inhibitor 2-DG (1 mmol/L) significantly enhanced apoptosis induction. Similar results were observed in another p53 wild-type H460 cells treated with PAB. Opposite results were found in p53-null H1299 cells, where PAB time-dependently decreased glucose utilization, and induced only apoptosis. Our results demonstrate that PAB-induced senescence is associated with enhanced glucose utilization, and lower glucose utilization might contribute to apoptosis induction. Thus, blocking glucose utilization contributes to the switch from senescence to apoptosis, and p53 plays an important role in this process.
Similar content being viewed by others
Introduction
Many cancer cells consume glucose avidly and produce lactic acid rather than catabolizing glucose via the tricarboxylic acid cycle (TCA) cycle. Cancer cells preferentially utilize glycolysis instead of oxidative phosphorylation in the presence of oxygen, despite an enormous demand for ATP to supply their growth. Glycolysis is much better suited to meeting the demand for the survival of cancer cells1,2. The advantage of the glycolytic metabolism is the production of glycolytic intermediates that can be used in anabolic pathways, allowing for the synthesis of the proteins, nucleic acids and lipids required for proliferation3. Therefore, the poor energetic pay-off of glycolysis by a high glucose supply is compensated.
This aerobic glycolysis is known as the “Warburg effect” and is considered a hallmark of cancer4,5. The transmembrane glucose transporter (Glut) protein mediates glucose uptake, which is required for the rapid proliferation of cancer cells6. Hexokinase is the first and rate-limiting enzyme during glycolysis and catalyzes the conversion of glucose to glucose-6-phosphate (G-6-P)7. The production of ATP or lactate by glycolysis might be an advantage for tumor survival and growth7,8. Most tumor cells primarily utilize glucose via aerobic glycolysis for their energy needs, suggesting that targeting the metabolic changes could be an effective strategy for cancer treatment9.
Pseudolaric acid B (PAB) exerts multiple biological and pharmacological activities, including anti-fungal, -tumor, -fertility and -microtubule properties10. Relationships between the molecular mechanisms of apoptosis and G2/M cell cycle arrest induced by PAB have been extensively studied11,12,13. However, our previous studies demonstrated that PAB partially induced senescence in human breast cancer MCF-7 and murine fibrosarcoma L929 cells11,14. In addition, PAB mainly induced senescence rather than apoptosis in p53 wild-type A549 cells15. However, PAB caused apoptosis without senescence in p53-null H1299 cells15.
Although the induction of cellular senescence is a well-established therapeutic strategy for tumor suppression, adverse actions are reported in some cases; for example, the secretion of tumor cell growth-promoting factors has been reported to cause chemoresistance16,17. Apoptosis is still one of the most effective anti-cancer strategies in cancer therapy, but the side effects on normal cells remain an important issue18. Understanding the advantages and drawbacks between senescence and apoptosis might provide a novel strategy for cancer therapy.
Here, we apply p53 wild-type and null cell lines to investigate the relationship between senescence and apoptosis induced by PAB. Additionally, the role of glucose utilization is also evaluated.
Materials and methods
Reagents
PAB with 99% purity was obtained from the National Institutes for Food and Drug Control (Beijing, China). PAB was dissolved in dimethylsulfoxide (DMSO) as a stock solution. The concentration of DMSO was maintained below 0.1% in all the cell cultures to prevent any detectable effect on cell growth. 2-Deoxy-D-glucose (2-DG) was purchased from Sigma Chemical Co (St Louis, MO, USA). Primary antibodies against p-p53 (Ser 15), p53, p21, AMPK, p-AMPK (Thr 172), caspase 3 and β-actin as well as horseradish peroxidase-conjugated secondary antibodies were purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA). Primary antibodies against Glut-1 and hexokinase-2 (HK-2) were purchased from Proteintech Group (Chicago, IL, USA). The SuperSignal® West Pico Chemiluminescent Substrate for horseradish peroxidase (HRP) was obtained from Thermo Scientific (Rockford, IL, USA).
Cell culture
A549, H460 and H1299 cells were obtained from the American Type Culture Collection (ATCC, Manassas, VA, USA). A549 cells were cultured in DMEM (Gibco, Grand Island, NY, USA) supplemented with 10% fetal bovine serum (FBS), 100 U/mL penicillin and 100 μg/mL streptomycin. H460 and H1299 cells were cultured in RPMI-1640 (Gibco, Grand Island, NY, USA) supplemented with 10% fetal bovine serum (FBS), 100 U/mL penicillin and 100 μg/mL streptomycin. Cells were incubated at 37°C with 5% CO2 in a humidified atmosphere. Logarithmically growing cells were used in all the experiments.
Cell viability assay
To determine the effect of glycolysis inhibition on cell viability, the cells were treated with or without 2-DG at the given concentrations for 1 h and were subsequently treated with PAB for the indicated time periods. The effect was measured by the MTT assay. The percentage of cell viability was calculated as follows:
Cell viability (%)=(A492,sample–A492,blank)/(A492,control–A492,blank)×100
SA-β-Gal staining
To detect senescent cells, the cellular senescence assay kit (Beyotime, Shanghai, China) was used according to the manufacturer's instructions. The cells were washed twice with PBS and were incubated in 1 mL of fixing solution at room temperature for 15 min. After removing the fixing solution followed by washing the fixed cells three times with PBS, the cells were stained with 1 mL of the working solution for freshly prepared cell staining at 37 °C for at least 12 h under protection from light. After staining, the cells were washed, and senescent cells were identified as blue-stained cells by light microscope (Olympus, Tokyo, Japan). At least 200 cells per field of vision were counted for each sample in three random fields to determine the percentage of SA-β-Gal-positive cells.
Apoptosis detection
Quantitative analysis of apoptotic cell death caused by PAB with or without 2-DG was analyzed using the Annexin V-FITC Apoptosis Detection Kit (KeyGEN BioTECH, Nanjing, Jiangsu, China) following the manufacturer's protocol. The cells were analyzed with a FACScan flow cytometer.
Glucose uptake assay
The cells were seeded and treated with drugs for the indicated period of time. Subsequently, the cells were washed twice with PBS and were incubated with 10 μmol/L of the fluorescent glucose analog 2-(N-(7-nitrobenz-2-oxa-1, 3-diazol-4-yl) amino)-2-deoxyglucose (2-NBDG; Invitrogen, Carlsbad, CA, USA)) in the dark at 37 °C for 2 h. The cells were collected, and glucose uptake was assessed by FACScan flow cytometry (Becton Dickinson, Franklin Lakes, NJ, USA).
Intracellular ATP assay
The intracellular ATP concentration was measured using the ATP Assay Kit (Beyotime, Shanghai, China). Briefly, cells were cultured in DMEM or 1640 medium for 24 h, followed by subsequent experiments. The ATP levels were measured according to the manufacturer's instructions. The ATP levels were expressed in terms of the protein content for signal normalization.
Lactate production assay
The cells were seeded into 6-well cell culture clusters (Corning, NY, USA) at a density of 1.5×104. After 24 h of incubation, the cells were treated with various drugs. For the assessment of lactate production, the media were collected and estimated using the Lactate Assay Kit (Jiancheng Bioengineering Institute, Nanjing, Jiangsu, China) according to the manufacturer's instructions. The cells were counted and used to normalize the relative lactate levels.
Western blot analysis
Cells were harvested, washed twice with PBS, and then were lysed in whole-cell RIPA lysis buffer (Beyotime, Shanghai, China) supplemented with PMSF (1 mmol/L). After 15 000×g centrifugation at 4°C for 15 min, the protein concentration was determined using the Bio-Rad protein assay reagent (Bio-Rad, Hercules, CA, USA). After denaturation with boiling water for 5 min, the lysates containing the same amount of protein were separated by 12% SDS-PAGE and were transferred onto a Millipore Immobilon®-P Transfer Membrane (Millipore, Billerica, MA, USA). The membranes were soaked in 5% skim milk, and then they were incubated with primary polyclonal antibodies overnight, followed by incubation with the corresponding HRP-conjugated secondary antibodies. The blots were visualized using the SuperSignal® West Pico Chemiluminescent Substrate.
Transfection of siRNA
Human p53-targeted (p53-siRNA), p21-targeted (p21-siRNA) and negative control (control-siRNA) siRNAs were purchased from GenePharma (Suzhou, Jiangsu, China). The cells were transfected with 5 nmol/L p53-, p21- or control-siRNA using siRNA-Mate™ (GenePharma, Suzhou, Jiangsu, China) according to the manufacturer's protocols. The transfected cells were used 24 h later for subsequent experiments.
Statistical analysis
The data from at least three independent experiments were presented as the mean±SD. Statistical comparisons were analyzed by one-way ANOVA using Statistics Package for Social Science software (version 13.0; SPSS, Chicago, IL, USA), and LSD-post hoc test was employed to assess the statistical significance of the difference between the control and treated groups. P<0.05 were considered to be statistically significant.
Results
PAB causes senescence and increases the utilization of glucose in A549 cells
In our previous study, we demonstrated that PAB primarily caused senescence rather than apoptosis in A549 cells15. To determine glucose utilization in senescence-capable cells after PAB treatment, flow cytometry was applied to detect glucose uptake. The results showed that PAB induced senescence accompanied by the increase in glucose uptake (Figure 1A, 1B). Next, we assessed whether PAB showed any effect on glucose transport in A549 cells. Western blot analysis indicated that PAB increased the expression of glucose transport protein 1 (Glut-1), and hexokinase 2 (HK-2), a rate-limiting enzyme during glycolysis, was enhanced by PAB treatment (Figure 1E, 1F). Further study also showed that PAB increased the level of ATP (Figure 1C) and lactate production (Figure 1D). Moreover, PAB enhanced the activation of AMPK (adenosine mono-phosphate (AMP)-activated protein kinase), a central energy sensor of the cell, suggesting that senescence-capable cells demanded much more bioenergy (Figure 1E, 1F). This result was consistent with a higher ATP level in PAB-induced senescent cells (Figure 1C). Collectively, these results exploited that PAB increased senescence-capable cells and enhanced the utilization of glucose.
P53 inactivation decreases PAB-induced high glucose utilization in A549 cells
Since p53 inhibition switched PAB-induced senescence to apoptosis15, the role of p53 in the regulation of glucose utilization was studied in subsequent experiments. Flow cytometric assay showed that the knockdown of p53 or p21 decreased the glucose uptake in PAB-treated A549 cells (Figure 2A). Accordingly, the ATP level and lactate production were attenuated by p53 or p21 inhibition in senescent A549 cells with PAB treatment (Figure 2B, 2C). Inactivation of p53 or p21 decreased the expression of senescent proteins but increased the cleavage of caspase 3 (a classical apoptotic protein) (Figure 2D, 2E). At the same time, the levels of glucose transport protein, a rate-limiting glycolytic enzyme, and phosphorylated AMPK were all inhibited by p53 or p21 interference in PAB-treated A549 cells (Figure 2D, 2E). These results demonstrated that p53 and p21 activation played important roles in PAB-induced higher utilization of glucose in A549 cells.
Inhibition of glucose utilization enhances apoptosis induction in PAB-treated A549 cells
The present study showed that the downregulation of senescence by p53-p21 inhibition decreases glucose utilization. Meanwhile, apoptosis induction is enhanced. Subsequently, a glycolytic inhibitor 2-deoxy-D-glucose (2-DG), targeting hexokinase, which is the entry-point enzyme for glycolysis, was applied to further investigate the relationships among glucose utilization, senescence and apoptosis. The MTT assay showed that PAB attenuated cell viability in the presence of 2-DG in A549 cells (Figure 3A). Consistently, 2-DG deceased senescence-capable cells but enhanced the apoptotic ratio (Figure 3B, 3C, 3G, 3H) in PAB-treated A549 cells. More generally, 2-DG attenuated the increased glucose utilization by treatment with PAB (Figure 3D-3F). These results indicated that senescence induction was accompanied by higher glucose utilization; however, apoptotic cells were elevated when blocking glucose metabolism.
Glucose utilization regulates the switch between senescence and apoptosis
To investigate whether the regulation of glucose utilization in senescence and apoptosis was specific for A549 cells, we further assessed the effect of PAB on H460 cells, which is another p53 wild-type cell line. The glucose uptake assay suggested that PAB might enhance the utilization of glucose in H460 cells (Figure 4A). Similar results to those of A549 cells regarding the ATP and lactate levels were shown following PAB treatment (Figure 4B, 4C). PAB caused senescence rather than apoptosis in H460 cells (Figure 4D, 4E). Meanwhile, the expression levels of Glut-1, HK-2 and p-AMPK were elevated in PAB-treated H460 cells (Figure 4D, 4E). Knockdown of p53 and p21 also decreased glucose utilization and increased apoptosis induction (Figure 4F, 4G). Subsequently, PAB-induced senescence was switched to apoptosis accompanied by poorer cell viability after treatment with a glycolytic inhibitor (Figure 4H-4L). These results indicated that glucose utilization regulated apoptosis, and senescence might require functional p53.
To further evaluate the role of p53 in this effect, we investigated whether H1299 cells (p53-null) had an opposite result because PAB induced only apoptosis in this cell line15. As expected, PAB decreased glucose uptake, and the levels of ATP and lactate were also attenuated in H1299 cells (Figure 5A-5C). The protein expression levels of glucose and energy metabolism were downregulated in PAB-treated H1299 cells, while the level of cleaved caspase 3 was elevated (Figure 5D, 5E). These results demonstrated that PAB-induced senescence was associated with enhanced glucose utilization, and lower glucose utilization might contribute to apoptosis induction. Furthermore, glucose utilization regulated the switch between senescence and apoptosis, and p53 played an important role in this process (Figure 5F).
Discussion
In this study, our results showed that PAB-induced senescent cells exhibit higher glucose utilization in p53 wild-type A549 and H460 cells. However, the utilization of glucose was decreased in PAB-induced apoptotic H1299 cells (p53-null). These data suggest that cells undergoing mitotic catastrophe enter into different fates (senescence or apoptosis) because of the status of p53 to regulate higher or lower glucose utilization. Further studies also demonstrate that blocking the utilization of glucose could elevate the apoptosis ratio in PAB-treated p53 wild-type cells.
Cellular senescence is a state of irreversible cell cycle arrest with active metabolism19. Drug-induced senescence might be a desirable therapeutic strategy, especially for tumors with apoptosis resistance, but there are some potentially harmful properties in viable senescent cells20. After chemotherapy, senescence-competent lymphomas show increased glucose utilization and much higher ATP production20. Similar effects are observed in PAB-caused senescent cells.
Autophagy, a lysosome-dependent degradation pathway, promoted glucose uptake and glycolytic flux21. In some cases, autophagy inhibition could switch camptothecin-induced senescence to apoptosis in cancer cells22. Moreover, emerging evidence also demonstrates that apoptosis induction in tumor cells may be closely associated with the inhibition of glycolysis23. Our previous study indicated that the knockdown of p53 or p21 increased apoptotic cell death in PAB-treated A549 cells. Therefore, we speculated that the knockdown of p53 (or p21) might impair the autophagy process and lead to lower glucose utilization with the decrease in Glut-1, HK-2 and p-AMPKα levels. Meanwhile, the decreased energy demand was accompanied with AMPK inactivation24. The detailed mechanism should be further studied.
Under some conditions, glucose withdrawal combined with antitumor drugs induces much more cancer cell death25. By contrast, cells cultured in high-glucose medium sometimes promote the senescence process26,27. Although metabolism is involved directly or indirectly in any cell, the examination of how metabolism is regulated in proliferating cells would provide new insight. The metabolism of glutamine, another major nutrient consumed by tumor cells besides glucose, has also been considered as a target for cancer therapy. Deficiency in glutamine, but not in glucose, induces proto-oncogene Myc-dependent apoptosis in human cells28.
AMPK, which is found in all eukaryotes, is an evolutionarily conserved sensor of the cellular energy status. AMPK activity is regulated by the cellular AMP/ATP ratio and upstream kinases29. It couples cellular bioenergetics to metabolic control and cell growth30. The activation of AMPK indicates that cells exhibit enhanced energetic demands24. Several bodies of evidence have suggested that the regulation of AMPK has been considered as a target for cancer therapy31. Senescent cells were susceptible to the inhibition of the energy sensor AMPK20.
Cells deficient in ATP often undergo apoptosis32. The inhibition of glycolysis may severely abolish ATP generation in cancer cells and thus may preferentially kill the tumor cells33. However, a potential problem is that the glycolytic inhibitors currently available are not very potent; hence, high doses are required7. 2-DG is a relatively specific blocker of glycolysis without altering other nutrients or metabolic pathways34. 2-DG alone cannot induce obvious cytotoxicity between 0.5 and 5 mmol/L in non-small cell lung cancer cells, while it enhances other antitumor drug-induced cell death35,36. Our investigation revealed that the combination of PAB with a glycolytic inhibitor (2-DG) converts senescence into apoptosis. Thus, PAB can induce apoptosis in A549 and H460 cells with glycolysis inhibition. Therefore, the elimination of senescent tumor cells can enhance the efficiency of cancer therapy. However, there is still much to learn about how tumor cell metabolism is regulated.
P53, a well-known tumor suppressor, plays critical roles in the regulation of cellular processes, including apoptosis and senescence37,38. However, PAB induced p53-dependent senescence but p53-independent apoptosis in human lung cancer cells15. Recent studies have also revealed that p53 possesses glucose metabolic function39. In some cases, p53 downregulates the activity of glucose-6-phosphate dehydrogenase (G6PD)40 or represses glucose transport genes41 in tumor cells. In this study, PAB caused enhanced glucose utilization, which resists the induction of apoptosis. This effect might be associated with the drawbacks of p53-dependent senescence. A growing body of evidence has suggested that p53 provides mechanisms to overcome metabolic stress and might contribute to tumor survival42,43.
The role of p53 in senescence induction has been well documented in tumor cells as a response to chemotherapy. In some studies, chemotherapeutic drugs were found to induce senescence in p53 wild-type cell lines, whereas p53 mutation or inactivation would result in the transition from senescence to apoptosis44,45,46,47,48,49. Additionally, several reports have shown that both wild-type and mutant p53 are involved in glucose metabolism, but with opposite effects50. Moreover, the AMPK activity was also inhibited in p53 mutant cell lines51,52. Thus, we speculated that PAB can exhibit similar effects in the H1299 cell line in p53 mutant lung cancer cells. Oridonin, an activated diterpenoid isolated from Rabdosia rubescens, caused cell death as well as glucose metabolism inhibition in p53-mutated colorectal cancer cells53.
The present study has identified a new underlying mechanism by which PAB induces a switch from senescence to apoptosis by blocking the utilization of glucose. The combination of glycolytic inhibition and other anticancer agents could provide a preferential method to induce tumor cell death with fewer side effects.
Author contribution
Guo-dong YAO and Takashi IKEJIMA designed the research; Guo-dong YAO, Jing YANG, Xiu-xiu LI and Xiao-yu SONG performed the research; Shin-Ichi TASHIRO and Satoshi ONODERA contributed some reagents; Guo-dong YAO analyzed the data and wrote the paper; Toshihiko HAYASHI, Shao-jiang SONG and Takashi IKEJIMA revised the paper.
References
Warburg O . On the origin of cancer cells. Science 1956; 123: 309–14.
Pfeiffer T, Schuster S, Bonhoeffer S . Cooperation and competition in the evolution of ATP-producing pathways. Science 2001; 292: 504–7.
Garcia-Jimenez C, Garcia-Martinez JM, Chocarro-Calvo A, De la Vieja A . A new link between diabetes and cancer: enhanced WNT/beta-catenin signaling by high glucose. J Mol Endocrinol 2014; 52: R51–66.
Upadhyay M, Samal J, Kandpal M, Singh OV, Vivekanandan P . The Warburg effect: insights from the past decade. Pharmacol Ther 2013; 137: 318–30.
Hanahan D, Weinberg RA . The hallmarks of cancer. Cell 2000; 100: 57–70.
Flier JS, Mueckler MM, Usher P, Lodish HF . Elevated levels of glucose transport and transporter messenger RNA are induced by ras or src oncogenes. Science 1987; 235: 1492–5.
Pelicano H, Martin DS, Xu RH, Huang P . Glycolysis inhibition for anticancer treatment. Oncogene 2006; 25: 4633–46.
Liao EC, Hsu YT, Chuah QY, Lee YJ, Hu JY, Huang TC, et al. Radiation induces senescence and a bystander effect through metabolic alterations. Cell Death Dis 2014; 5: e1255.
Christofk HR, Vander Heiden MG, Harris MH, Ramanathan A, Gerszten RE, Wei R, et al. The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature 2008; 452: 230–3.
Yan Z, Hua H, Xu Y, Samaranayake LP . Potent antifungal activity of pure compounds from traditional Chinese medicine extracts against six oral Candida species and the synergy with fluconazole against azole-resistant Candida albicans. Evid Based Complement Alternat Med 2012; 2012: 106583.
Yu JH, Cui Q, Jiang YY, Yang W, Tashiro S, Onodera S, et al. Pseudolaric acid B induces apoptosis, senescence, and mitotic arrest in human breast cancer MCF-7. Acta Pharmacol Sin 2007; 28: 1975–83.
Meng AG, Jiang LL . Induction of G2/M arrest by pseudolaric acid B is mediated by activation of the ATM signaling pathway. Acta Pharmacol Sin 2009; 30: 442–50.
Yao G, Qi M, Ji X, Fan S, Xu L, Hayashi T, et al. ATM-p53 pathway causes G2/M arrest, but represses apoptosis in pseudolaric acid B-treated HeLa cells. Arch Biochem Biophys 2014; 558: 51–60.
Qi M, Fan S, Yao G, Li Z, Zhou H, Tashiro S, et al. Pseudolaric acid B-induced autophagy contributes to senescence via enhancement of ROS generation and mitochondrial dysfunction in murine fibrosarcoma L929 cells. J Pharmacol Sci 2013; 121: 200–11.
Yao GD, Yang J, Li Q, Zhang Y, Qi M, Fan SM, et al. Activation of p53 contributes to pseudolaric acid B-induced senescence in human lung cancer cells in vitro. Acta Pharmacol Sin 2016; 37: 919–29.
Cahu J, Bustany S, Sola B . Senescence-associated secretory phenotype favors the emergence of cancer stem-like cells. Cell Death Dis 2012; 3: e446.
Coppe JP, Desprez PY, Krtolica A, Campisi J . The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol 2010; 5: 99–118.
Ghobrial IM, Witzig TE, Adjei AA . Targeting apoptosis pathways in cancer therapy. CA Cancer J Clin 2005; 55: 178–94.
Campisi J . Aging, cellular senescence, and cancer. Annu Rev Physiol 2013; 75: 685–705.
Dorr JR, Yu Y, Milanovic M, Beuster G, Zasada C, Dabritz JH, et al. Synthetic lethal metabolic targeting of cellular senescence in cancer therapy. Nature 2013; 501: 421–5.
Goldsmith J, Levine B, Debnath J . Autophagy and cancer metabolism. Methods Enzymol 2014; 542: 25–57.
Zhang JW, Zhang SS, Song JR, Sun K, Zong C, Zhao QD, et al. Autophagy inhibition switches low-dose camptothecin-induced premature senescence to apoptosis in human colorectal cancer cells. Biochem Pharmacol 2014; 90: 265–75.
Gao C, Yan X, Wang B, Yu L, Han J, Li D, et al. Jolkinolide B induces apoptosis and inhibits tumor growth in mouse melanoma B16F10 cells by altering glycolysis. Sci Rep 2016; 6: 36114.
Jones RG, Plas DR, Kubek S, Buzzai M, Mu J, Xu Y, et al. AMP-activated protein kinase induces a p53-dependent metabolic checkpoint. Mol Cell 2005; 18: 283–93.
Menendez JA, Oliveras-Ferraros C, Cufi S, Corominas-Faja B, Joven J, Martin-Castillo B, et al. Metformin is synthetically lethal with glucose withdrawal in cancer cells. Cell Cycle 2012; 11: 2782–92.
del Nogal M, Troyano N, Calleros L, Griera M, Rodriguez-Puyol M, Rodriguez-Puyol D, et al. Hyperosmolarity induced by high glucose promotes senescence in human glomerular mesangial cells. Int J Biochem Cell Biol 2014; 54: 98–110.
E S, Kijima R, Honma T, Yamamoto K, Hatakeyama Y, Kitano Y, et al. 1-Deoxynojirimycin attenuates high glucose-accelerated senescence in human umbilical vein endothelial cells. Exp Gerontol 2014; 55: 63–9.
Yuneva M, Zamboni N, Oefner P, Sachidanandam R, Lazebnik Y . Deficiency in glutamine but not glucose induces MYC-dependent apoptosis in human cells. J Cell Biol 2007; 178: 93–105.
Hardie DG, Scott JW, Pan DA, Hudson ER . Management of cellular energy by the AMP-activated protein kinase system. FEBS Lett 2003; 546: 113–20.
Blagih J, Krawczyk CM, Jones RG . LKB1 and AMPK: central regulators of lymphocyte metabolism and function. Immunol Rev 2012; 249: 59–71.
Rehman G, Shehzad A, Khan AL, Hamayun M . Role of AMP-activated protein kinase in cancer therapy. Arch Pharm (Weinheim) 2014; 347: 457–68.
Izyumov DS, Avetisyan AV, Pletjushkina OY, Sakharov DV, Wirtz KW, Chernyak BV, et al. "Wages of fear": transient threefold decrease in intracellular ATP level imposes apoptosis. Biochim Biophys Acta 2004; 1658: 141–7.
Xu RH, Pelicano H, Zhou Y, Carew JS, Feng L, Bhalla KN, et al. Inhibition of glycolysis in cancer cells: a novel strategy to overcome drug resistance associated with mitochondrial respiratory defect and hypoxia. Cancer Res 2005; 65: 613–21.
Sols A, Crane RK . The inhibition of brain hexokinase by adenosinediphosphate and sulfhydryl reagents. J Biol Chem 1954; 206: 925–36.
Vuyyuri SB, Rinkinen J, Worden E, Shim H, Lee S, Davis KR . Ascorbic acid and a cytostatic inhibitor of glycolysis synergistically induce apoptosis in non-small cell lung cancer cells. PLoS One 2013; 8: e67081.
Mi YJ, Geng GJ, Zou ZZ, Gao J, Luo XY, Liu Y, et al. Dihydroartemisinin inhibits glucose uptake and cooperates with glycolysis inhibitor to induce apoptosis in non-small cell lung carcinoma cells. PLoS One 2015; 10: e0120426.
Schmitt CA, Fridman JS, Yang M, Lee S, Baranov E, Hoffman RM, et al. A senescence program controlled by p53 and p16INK4a contributes to the outcome of cancer therapy. Cell 2002; 109: 335–46.
Vogelstein B, Lane D, Levine AJ . Surfing the p53 network. Nature 2000; 408: 307–10.
Bensaad K, Tsuruta A, Selak MA, Vidal MN, Nakano K, Bartrons R, et al. TIGAR, a p53-inducible regulator of glycolysis and apoptosis. Cell 2006; 126: 107–20.
Jiang P, Du W, Wang X, Mancuso A, Gao X, Wu M, et al. p53 regulates biosynthesis through direct inactivation of glucose-6-phosphate dehydrogenase. Nat Cell Biol 2011; 13: 310–6.
Schwartzenberg-Bar-Yoseph F, Armoni M, Karnieli E . The tumor suppressor p53 down-regulates glucose transporters GLUT1 and GLUT4 gene expression. Cancer Res 2004; 64: 2627–33.
Vousden KH, Ryan KM . p53 and metabolism. Nat Rev Cancer 2009; 9: 691–700.
Maddocks OD, Vousden KH . Metabolic regulation by p53. J Mol Med (Berl) 2011; 89: 237–45.
Chang BD, Broude EV, Dokmanovic M, Zhu H, Ruth A, Xuan Y, et al. A senescence-like phenotype distinguishes tumor cells that undergo terminal proliferation arrest after exposure to anticancer agents. Cancer Res 1999; 59: 3761–7.
te Poele RH, Okorokov AL, Jardine L, Cummings J, Joel SP . DNA damage is able to induce senescence in tumor cells in vitro and in vivo. Cancer Res 2002; 62: 1876–83.
Ewald JA, Desotelle JA, Wilding G, Jarrard DF . Therapy-induced senescence in cancer. J Natl Cancer Inst 2010; 102: 1536–46.
Roninson IB . Tumor senescence as a determinant of drug response in vivo. Drug Resist Updat 2002; 5: 204–8.
Jackson JG, Pant V, Li Q, Chang LL, Quintas-Cardama A, Garza D, et al. p53-mediated senescence impairs the apoptotic response to chemotherapy and clinical outcome in breast cancer. Cancer Cell 2012; 21: 793–806.
Tonnessen-Murray CA, Lozano G, Jackson JG . The regulation of cellular functions by the p53 protein: cellular senescence. Cold Spring Harb Perspect Med 2017; 7.
Liu J, Zhang C, Hu W, Feng Z . Tumor suppressor p53 and its mutants in cancer metabolism. Cancer Lett 2015; 356: 197–203.
Zhou G, Wang J, Zhao M, Xie TX, Tanaka N, Sano D, et al. Gain-of-function mutant p53 promotes cell growth and cancer cell metabolism via inhibition of AMPK activation. Mol Cell 2014; 54: 960–74.
Parrales A, Iwakuma T . p53 as a regulator of lipid metabolism in cancer. Int J Mol Sci 2016; 17.
Yao Z, Xie F, Li M, Liang Z, Xu W, Yang J, et al. Oridonin induces autophagy via inhibition of glucose metabolism in p53-mutated colorectal cancer cells. Cell Death Dis 2017; 8: e2633.
Author information
Authors and Affiliations
Corresponding author
Rights and permissions
About this article
Cite this article
Yao, Gd., Yang, J., Li, Xx. et al. Blocking the utilization of glucose induces the switch from senescence to apoptosis in pseudolaric acid B-treated human lung cancer cells in vitro. Acta Pharmacol Sin 38, 1401–1411 (2017). https://doi.org/10.1038/aps.2017.39
Received:
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/aps.2017.39