Skip to main content
Top
Published in: Journal of Hematology & Oncology 1/2016

Open Access 01-12-2016 | Research

TIGAR cooperated with glycolysis to inhibit the apoptosis of leukemia cells and associated with poor prognosis in patients with cytogenetically normal acute myeloid leukemia

Authors: Sixuan Qian, Jianyong Li, Ming Hong, Yu Zhu, Huihui Zhao, Yue Xie, Jiayu Huang, Yun Lian, Yanru Li, Shuai Wang, Jianping Mao, Yaoyu Chen

Published in: Journal of Hematology & Oncology | Issue 1/2016

Login to get access

Abstract

Background

Cancer cells show increased glycolysis and take advantage of this metabolic pathway to generate ATP. The TP53-induced glycolysis and apoptosis regulator (TIGAR) inhibits aerobic glycolysis and protects tumor cells from intracellular reactive oxygen species (ROS)-associated apoptosis. However, the function of TIGAR in glycolysis and survival of acute myeloid leukemia cells remains unclear.

Methods

We analyzed TIGAR expression in cytogenetically normal (CN-) AML patients and the correlations with clinical and biological parameters. In vivo and in vitro, we tested whether glycolysis may induce TIGAR expression and evaluated the combination effect of glycolysis inhibitor and TIGAR knockdown on human leukemia cell proliferation.

Results

High TIGAR expression was an independent predictor of poor survival and high incidence of relapse in adult patients with CN-AML. TIGAR also showed high expression in multiple human leukemia cell lines and knockdown of TIGAR activated glycolysis through PFKFB3 upregulation in human leukemia cells. Knockdown of TIGAR inhibited the proliferation of human leukemia cells and sensitized leukemia cells to glycolysis inhibitor both in vitro and in vivo. Furthermore, TIGAR knockdown in combination with glycolysis inhibitor 2-DG led leukemia cells to apoptosis. In addition, the p53 activator Nutlin-3α showed a significant combinational effect with TIGAR knockdown in leukemia cells. However, TIGAR expression and its anti-apoptotic effects were uncoupled from overexpression of exogenous p53 in leukemia cells.

Conclusions

TIGAR might be a predictor of poor survival and high incidence of relapse in AML patients, and the combination of TIGAR inhibitors with anti-glycolytic agents may be novel therapies for the future clinical use in AML patients.
Appendix
Available only for authorised users
Literature
1.
go back to reference A J, Qian S, Wang G, Yan B, Zhang S, Huang Q, et al. Chronic myeloid leukemia patients sensitive and resistant to imatinib treatment show different metabolic responses. PloS one. 2010;5:e13186. A J, Qian S, Wang G, Yan B, Zhang S, Huang Q, et al. Chronic myeloid leukemia patients sensitive and resistant to imatinib treatment show different metabolic responses. PloS one. 2010;5:e13186.
2.
go back to reference Herst PM, Howman RA, Neeson PJ, Berridge MV, Ritchie DS. The level of glycolytic metabolism in acute myeloid leukemia blasts at diagnosis is prognostic for clinical outcome. J Leukoc Biol. 2011;89:51–5.CrossRefPubMed Herst PM, Howman RA, Neeson PJ, Berridge MV, Ritchie DS. The level of glycolytic metabolism in acute myeloid leukemia blasts at diagnosis is prognostic for clinical outcome. J Leukoc Biol. 2011;89:51–5.CrossRefPubMed
3.
go back to reference Zhao K, Zhou Y, Qiao C, Ni T, Li Z, Wang X, et al. Oroxylin A promotes PTEN-mediated negative regulation of MDM2 transcription via SIRT3-mediated deacetylation to stabilize p53 and inhibit glycolysis in wt-p53 cancer cells. J Hematol Oncol. 2015;8:41.CrossRefPubMedPubMedCentral Zhao K, Zhou Y, Qiao C, Ni T, Li Z, Wang X, et al. Oroxylin A promotes PTEN-mediated negative regulation of MDM2 transcription via SIRT3-mediated deacetylation to stabilize p53 and inhibit glycolysis in wt-p53 cancer cells. J Hematol Oncol. 2015;8:41.CrossRefPubMedPubMedCentral
4.
go back to reference Cheung EC, Athineos D, Lee P, Ridgway RA, Lambie W, Nixon C, et al. TIGAR is required for efficient intestinal regeneration and tumorigenesis. Dev Cell. 2013;25:463–77.CrossRefPubMedPubMedCentral Cheung EC, Athineos D, Lee P, Ridgway RA, Lambie W, Nixon C, et al. TIGAR is required for efficient intestinal regeneration and tumorigenesis. Dev Cell. 2013;25:463–77.CrossRefPubMedPubMedCentral
5.
go back to reference Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324:1029–33.CrossRefPubMedPubMedCentral Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324:1029–33.CrossRefPubMedPubMedCentral
8.
go back to reference 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.CrossRefPubMed 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.CrossRefPubMed
10.
go back to reference Ros S, Schulze A. Balancing glycolytic flux: the role of 6-phosphofructo-2-kinase/fructose 2,6-bisphosphatases in cancer metabolism. Cancer Metab. 2013;1:8.CrossRefPubMedPubMedCentral Ros S, Schulze A. Balancing glycolytic flux: the role of 6-phosphofructo-2-kinase/fructose 2,6-bisphosphatases in cancer metabolism. Cancer Metab. 2013;1:8.CrossRefPubMedPubMedCentral
11.
go back to reference Won KY, Lim SJ, Kim GY, Kim YW, Han SA, Song JY, et al. Regulatory role of p53 in cancer metabolism via SCO2 and TIGAR in human breast cancer. Hum Pathol. 2012;43:221–8.CrossRefPubMed Won KY, Lim SJ, Kim GY, Kim YW, Han SA, Song JY, et al. Regulatory role of p53 in cancer metabolism via SCO2 and TIGAR in human breast cancer. Hum Pathol. 2012;43:221–8.CrossRefPubMed
12.
go back to reference Ko YH, Lin Z, Flomenberg N, Pestell RG, Howell A, Sotgia F, et al. Glutamine fuels a vicious cycle of autophagy in the tumor stroma and oxidative mitochondrial metabolism in epithelial cancer cells: implications for preventing chemotherapy resistance. Cancer Biol Ther. 2011;12:1085–97.CrossRefPubMedPubMedCentral Ko YH, Lin Z, Flomenberg N, Pestell RG, Howell A, Sotgia F, et al. Glutamine fuels a vicious cycle of autophagy in the tumor stroma and oxidative mitochondrial metabolism in epithelial cancer cells: implications for preventing chemotherapy resistance. Cancer Biol Ther. 2011;12:1085–97.CrossRefPubMedPubMedCentral
13.
go back to reference Sinha S, Ghildiyal R, Mehta VS, Sen E. ATM-NFkappaB axis-driven TIGAR regulates sensitivity of glioma cells to radiomimetics in the presence of TNFalpha. Cell Death Dis. 2013;4:e615.CrossRefPubMedPubMedCentral Sinha S, Ghildiyal R, Mehta VS, Sen E. ATM-NFkappaB axis-driven TIGAR regulates sensitivity of glioma cells to radiomimetics in the presence of TNFalpha. Cell Death Dis. 2013;4:e615.CrossRefPubMedPubMedCentral
14.
go back to reference Pena-Rico MA, Calvo-Vidal MN, Villalonga-Planells R, Martinez-Soler F, Gimenez-Bonafe P, Navarro-Sabate A, et al. TP53 induced glycolysis and apoptosis regulator (TIGAR) knockdown results in radiosensitization of glioma cells. Radiother Oncol. 2011;101:132–9.CrossRefPubMed Pena-Rico MA, Calvo-Vidal MN, Villalonga-Planells R, Martinez-Soler F, Gimenez-Bonafe P, Navarro-Sabate A, et al. TP53 induced glycolysis and apoptosis regulator (TIGAR) knockdown results in radiosensitization of glioma cells. Radiother Oncol. 2011;101:132–9.CrossRefPubMed
15.
go back to reference Wanka C, Steinbach JP, Rieger J. Tp53-induced glycolysis and apoptosis regulator (TIGAR) protects glioma cells from starvation-induced cell death by up-regulating respiration and improving cellular redox homeostasis. J Biol Chem. 2012;287:33436–46.CrossRefPubMedPubMedCentral Wanka C, Steinbach JP, Rieger J. Tp53-induced glycolysis and apoptosis regulator (TIGAR) protects glioma cells from starvation-induced cell death by up-regulating respiration and improving cellular redox homeostasis. J Biol Chem. 2012;287:33436–46.CrossRefPubMedPubMedCentral
16.
go back to reference Lui VW, Lau CP, Cheung CS, Ho K, Ng MH, Cheng SH, et al. An RNA-directed nucleoside anti-metabolite, 1-(3-C-ethynyl-beta-d-ribo-pentofuranosyl)cytosine (ECyd), elicits antitumor effect via TP53-induced glycolysis and apoptosis regulator (TIGAR) downregulation. Biochem Pharmacol. 2010;79:1772–80.CrossRefPubMed Lui VW, Lau CP, Cheung CS, Ho K, Ng MH, Cheng SH, et al. An RNA-directed nucleoside anti-metabolite, 1-(3-C-ethynyl-beta-d-ribo-pentofuranosyl)cytosine (ECyd), elicits antitumor effect via TP53-induced glycolysis and apoptosis regulator (TIGAR) downregulation. Biochem Pharmacol. 2010;79:1772–80.CrossRefPubMed
17.
go back to reference Lui VW, Wong EY, Ho K, Ng PK, Lau CP, Tsui SK, et al. Inhibition of c-Met downregulates TIGAR expression and reduces NADPH production leading to cell death. Oncogene. 2011;30:1127–34.CrossRefPubMed Lui VW, Wong EY, Ho K, Ng PK, Lau CP, Tsui SK, et al. Inhibition of c-Met downregulates TIGAR expression and reduces NADPH production leading to cell death. Oncogene. 2011;30:1127–34.CrossRefPubMed
18.
go back to reference Yin L, Kosugi M, Kufe D. Inhibition of the MUC1-C oncoprotein induces multiple myeloma cell death by down-regulating TIGAR expression and depleting NADPH. Blood. 2012;119:810–6.CrossRefPubMedPubMedCentral Yin L, Kosugi M, Kufe D. Inhibition of the MUC1-C oncoprotein induces multiple myeloma cell death by down-regulating TIGAR expression and depleting NADPH. Blood. 2012;119:810–6.CrossRefPubMedPubMedCentral
19.
go back to reference Xie JM, Li B, Yu HP, Gao QG, Li W, Wu HR, et al. TIGAR has a dual role in cancer cell survival through regulating apoptosis and autophagy. Cancer Res. 2014;74:5127–38.CrossRefPubMed Xie JM, Li B, Yu HP, Gao QG, Li W, Wu HR, et al. TIGAR has a dual role in cancer cell survival through regulating apoptosis and autophagy. Cancer Res. 2014;74:5127–38.CrossRefPubMed
20.
go back to reference Fu Y, Zhu H, Wu W, Xu J, Chen T, Xu B, et al. Clinical significance of lymphoid enhancer-binding factor 1 expression in acute myeloid leukemia. Leuk Lymphoma. 2014;55:371–7.CrossRefPubMed Fu Y, Zhu H, Wu W, Xu J, Chen T, Xu B, et al. Clinical significance of lymphoid enhancer-binding factor 1 expression in acute myeloid leukemia. Leuk Lymphoma. 2014;55:371–7.CrossRefPubMed
21.
go back to reference Qiao C, Zhang R, Hong M, Wang L, Zhang JF, Wu YJ, et al. Heterogeneous leukemic clones identified by NPM1 mutation analysis in patient with acute monocytic leukemia. Leuk Lymphoma. 2012;53:886–90.CrossRefPubMed Qiao C, Zhang R, Hong M, Wang L, Zhang JF, Wu YJ, et al. Heterogeneous leukemic clones identified by NPM1 mutation analysis in patient with acute monocytic leukemia. Leuk Lymphoma. 2012;53:886–90.CrossRefPubMed
22.
go back to reference Heaney NB, Pellicano F, Zhang B, Crawford L, Chu S, Kazmi SM, et al. Bortezomib induces apoptosis in primitive chronic myeloid leukemia cells including LTC-IC and NOD/SCID repopulating cells. Blood. 2010;115:2241–50.CrossRefPubMedPubMedCentral Heaney NB, Pellicano F, Zhang B, Crawford L, Chu S, Kazmi SM, et al. Bortezomib induces apoptosis in primitive chronic myeloid leukemia cells including LTC-IC and NOD/SCID repopulating cells. Blood. 2010;115:2241–50.CrossRefPubMedPubMedCentral
23.
go back to reference Rastogi RP, Singh SP, Hader DP, Sinha RP. Detection of reactive oxygen species (ROS) by the oxidant-sensing probe 2′,7′-dichlorodihydrofluorescein diacetate in the cyanobacterium Anabaena variabilis PCC 7937. Biochem Biophys Res Commun. 2010;397:603–7.CrossRefPubMed Rastogi RP, Singh SP, Hader DP, Sinha RP. Detection of reactive oxygen species (ROS) by the oxidant-sensing probe 2′,7′-dichlorodihydrofluorescein diacetate in the cyanobacterium Anabaena variabilis PCC 7937. Biochem Biophys Res Commun. 2010;397:603–7.CrossRefPubMed
24.
go back to reference Hulleman E, Kazemier KM, Holleman A, VanderWeele DJ, Rudin CM, Broekhuis MJ, et al. Inhibition of glycolysis modulates prednisolone resistance in acute lymphoblastic leukemia cells. Blood. 2009;113:2014–21.CrossRefPubMedPubMedCentral Hulleman E, Kazemier KM, Holleman A, VanderWeele DJ, Rudin CM, Broekhuis MJ, et al. Inhibition of glycolysis modulates prednisolone resistance in acute lymphoblastic leukemia cells. Blood. 2009;113:2014–21.CrossRefPubMedPubMedCentral
25.
go back to reference Huang Y, Du KM, Xue ZH, Yan H, Li D, Liu W, et al. Cobalt chloride and low oxygen tension trigger differentiation of acute myeloid leukemic cells: possible mediation of hypoxia-inducible factor-1alpha. Leukemia. 2003;17:2065–73.CrossRefPubMed Huang Y, Du KM, Xue ZH, Yan H, Li D, Liu W, et al. Cobalt chloride and low oxygen tension trigger differentiation of acute myeloid leukemic cells: possible mediation of hypoxia-inducible factor-1alpha. Leukemia. 2003;17:2065–73.CrossRefPubMed
26.
28.
go back to reference Kwee JK. A paradoxical chemoresistance and tumor suppressive role of antioxidant in solid cancer cells: a strange case of Dr Jekyll and Mr Hyde. BioMed Res Int. 2014;2014:209845.CrossRefPubMedPubMedCentral Kwee JK. A paradoxical chemoresistance and tumor suppressive role of antioxidant in solid cancer cells: a strange case of Dr Jekyll and Mr Hyde. BioMed Res Int. 2014;2014:209845.CrossRefPubMedPubMedCentral
29.
go back to reference Telang S, Clem BF, Klarer AC, Clem AL, Trent JO, Bucala R, et al. Small molecule inhibition of 6-phosphofructo-2-kinase suppresses t cell activation. J Transl Med. 2012;10:95.CrossRefPubMedPubMedCentral Telang S, Clem BF, Klarer AC, Clem AL, Trent JO, Bucala R, et al. Small molecule inhibition of 6-phosphofructo-2-kinase suppresses t cell activation. J Transl Med. 2012;10:95.CrossRefPubMedPubMedCentral
30.
go back to reference Beesley AH, Firth MJ, Ford J, Weller RE, Freitas JR, Perera KU, et al. Glucocorticoid resistance in T-lineage acute lymphoblastic leukaemia is associated with a proliferative metabolism. Br J Cancer. 2009;100:1926–36.CrossRefPubMedPubMedCentral Beesley AH, Firth MJ, Ford J, Weller RE, Freitas JR, Perera KU, et al. Glucocorticoid resistance in T-lineage acute lymphoblastic leukaemia is associated with a proliferative metabolism. Br J Cancer. 2009;100:1926–36.CrossRefPubMedPubMedCentral
31.
go back to reference Herst PM, Hesketh EL, Ritchie DS, Berridge MV. Glycolytic metabolism confers resistance to combined all-trans retinoic acid and arsenic trioxide-induced apoptosis in HL60rho0 cells. Leuk Res. 2008;32:327–33.CrossRefPubMed Herst PM, Hesketh EL, Ritchie DS, Berridge MV. Glycolytic metabolism confers resistance to combined all-trans retinoic acid and arsenic trioxide-induced apoptosis in HL60rho0 cells. Leuk Res. 2008;32:327–33.CrossRefPubMed
32.
go back to reference 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.CrossRefPubMedPubMedCentral 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.CrossRefPubMedPubMedCentral
33.
go back to reference Madan E, Gogna R, Bhatt M, Pati U, Kuppusamy P, Mahdi AA. Regulation of glucose metabolism by p53: emerging new roles for the tumor suppressor. Oncotarget. 2011;2:948–57.CrossRefPubMedPubMedCentral Madan E, Gogna R, Bhatt M, Pati U, Kuppusamy P, Mahdi AA. Regulation of glucose metabolism by p53: emerging new roles for the tumor suppressor. Oncotarget. 2011;2:948–57.CrossRefPubMedPubMedCentral
34.
go back to reference Ok CY, Patel KP, Garcia-Manero G, Routbort MJ, Peng J, Tang G, Goswami M, et al. TP53 mutation characteristics in therapy-related myelodysplastic syndromes and acute myeloid leukemia is similar to de novo diseases. J Hematol Oncol. 2015;8:45.CrossRefPubMedPubMedCentral Ok CY, Patel KP, Garcia-Manero G, Routbort MJ, Peng J, Tang G, Goswami M, et al. TP53 mutation characteristics in therapy-related myelodysplastic syndromes and acute myeloid leukemia is similar to de novo diseases. J Hematol Oncol. 2015;8:45.CrossRefPubMedPubMedCentral
Metadata
Title
TIGAR cooperated with glycolysis to inhibit the apoptosis of leukemia cells and associated with poor prognosis in patients with cytogenetically normal acute myeloid leukemia
Authors
Sixuan Qian
Jianyong Li
Ming Hong
Yu Zhu
Huihui Zhao
Yue Xie
Jiayu Huang
Yun Lian
Yanru Li
Shuai Wang
Jianping Mao
Yaoyu Chen
Publication date
01-12-2016
Publisher
BioMed Central
Published in
Journal of Hematology & Oncology / Issue 1/2016
Electronic ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-016-0360-4

Other articles of this Issue 1/2016

Journal of Hematology & Oncology 1/2016 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine