DOI QR코드

DOI QR Code

Alkylglyceronephosphate Synthase (AGPS) Alters Lipid Signaling Pathways and Supports Chemotherapy Resistance of Glioma and Hepatic Carcinoma Cell Lines

  • Zhu, Yu (Department of Clinical Laboratory, Tianjin Huan Hu Hospital, Tianjin Key Laboratory of Cerebral Vessels and Neural Degeneration) ;
  • Liu, Xing-Jun (Department of Neurosurgery, Tianjin Haihe Hospital) ;
  • Yang, Ping (Department of Clinical Laboratory, Tianjin Huan Hu Hospital, Tianjin Key Laboratory of Cerebral Vessels and Neural Degeneration) ;
  • Zhao, Meng (Department of Immunology and Department of Biochemistry, School of Basic Medical Sciences, Tianjin Medical University) ;
  • Lv, Li-Xia (Department of Clinical Laboratory, Tianjin Huan Hu Hospital, Tianjin Key Laboratory of Cerebral Vessels and Neural Degeneration) ;
  • Zhang, Guo-Dong (Department of Clinical Laboratory, Tianjin Huan Hu Hospital, Tianjin Key Laboratory of Cerebral Vessels and Neural Degeneration) ;
  • Wang, Qin (Department of Clinical Laboratory, Tianjin Huan Hu Hospital, Tianjin Key Laboratory of Cerebral Vessels and Neural Degeneration) ;
  • Zhang, Ling (Department of Clinical Laboratory, Tianjin Huan Hu Hospital, Tianjin Key Laboratory of Cerebral Vessels and Neural Degeneration)
  • Published : 2014.04.01

Abstract

Chemotherapy continues to be a mainstay of cancer treatment, although drug resistance is a major obstacle. Lipid metabolism plays a critical role in cancer pathology, with elevated ether lipid levels. Recently, alkylglyceronephosphate synthase (AGPS), an enzyme that catalyzes the critical step in ether lipid synthesis, was shown to be up-regulated in multiple types of cancer cells and primary tumors. Here, we demonstrated that silencing of AGPS in chemotherapy resistance glioma U87MG/DDP and hepatic carcinoma HepG2/ADM cell lines resulted in reduced cell proliferation, increased drug sensitivity, cell cycle arrest and cell apoptosis through reducing the intracellular concentration of lysophosphatidic acid (LPA), lysophosphatidic acid-ether (LPAe) and prostaglandin E2 (PGE2), resulting in reduction of LPA receptor and EP receptors mediated PI3K/AKT signaling pathways and the expression of several multi-drug resistance genes, like MDR1, MRP1 and ABCG2. ${\beta}$-catenin, caspase-3/8, Bcl-2 and survivin were also found to be involved. In summary, our studies indicate that AGPS plays a role in cancer chemotherapy resistance by mediating signaling lipid metabolism in cancer cells.

Keywords

References

  1. Aust S, Pils S, Polterauer S, et al (2013). Expression of Bcl-2 and the antiapoptotic BAG family proteins in ovarian cancer. Appl Immunohistochem Mol Morphol, 21, 518-24. https://doi.org/10.1097/PAI.0b013e318284a053
  2. Benjamin DI, Cozzo A, Ji X, et al (2013). Ether lipid generating enzyme AGPS alters the balance of structural and signaling lipids to fuel cancer pathogenicity. Proc Natl Acad Sci USA, 110, 14912-7. https://doi.org/10.1073/pnas.1310894110
  3. Blackburn J, Mansell JP (2012). The emerging role of lysophosphatidic acid (LPA) in skeletal biology. Bone, 50, 756-62. https://doi.org/10.1016/j.bone.2011.12.002
  4. Brindley DN, Lin FT, Tigyi GJ (2013). Role of the autotaxin-lysophosphatidate axis in cancer resistance to chemotherapy and radiotherapy. Biochim Biophys Acta, 1831, 74-85. https://doi.org/10.1016/j.bbalip.2012.08.015
  5. Castellone MD, Teramoto H, Williams BO, et al (2005). Prostaglandin E2 promotes colon cancer cell growth through a Gs-axin-beta-catenin signaling axis. Science, 310, 1504-10. https://doi.org/10.1126/science.1116221
  6. Gillibert-Duplantier J, Neaud V, Blanc JF, et al (2007). Thrombin inhibits migration of human hepatic myofibroblasts. Am J Physiol Gastrointest Liver Physiol, 293, 128-36. https://doi.org/10.1152/ajpgi.00031.2007
  7. Greenhough A, Smartt HJ, Moore AE, et al (2009). The COX-2/PGE2 pathway: key roles in the hallmarks of cancer and adaptation to the tumour microenvironment. Carcinogenesis, 30, 377-86. https://doi.org/10.1093/carcin/bgp014
  8. Han C, Michalopoulos GK, Wu T (2006). Prostaglandin E2 receptor EP1 transactivates EGFR/MET receptor tyrosine kinases and enhances invasiveness in human hepatocellular carcinoma cells. J Cell Physiol, 207, 261-70. https://doi.org/10.1002/jcp.20560
  9. Henderson MJ, Haber M, Porro A, et al (2011). ABCC multidrug transporters in childhood neuroblastoma: clinical and biological effects independent of cytotoxic drug efflux. J Natl Cancer Inst, 103, 1236-51. https://doi.org/10.1093/jnci/djr256
  10. Hu S, Sun W, Wei W, et al (2013). Involvement of the prostaglandin E receptor EP2 in paeoniflorin-induced human hepatoma cell apoptosis. Anticancer Drugs, 24, 140-9. https://doi.org/10.1097/CAD.0b013e32835a4dac
  11. Hull MA, Ko SC, Hawcroft G (2004). Prostaglandin EP receptors: targets for treatment and prevention of colorectal cancer? Mol Cancer Ther, 3, 1031-9. https://doi.org/10.4161/cbt.3.10.1227
  12. Kambe A, Iguchi G, Moon Y, et al (2008). Regulation of EP4 expression via the Sp-1 transcription factor: inhibition of expression by anti-cancer agents. Biochim Biophys Acta, 1783, 1211-9. https://doi.org/10.1016/j.bbamcr.2008.01.032
  13. Kashiwagi E, Shiota M, Yokomizo A, et al (2013). Prostaglandin receptor EP3 mediates growth inhibitory effect of aspirin through androgen receptor and contributes to castration resistance in prostate cancer cells. Endocr Relat Cancer, 20, 431-41. https://doi.org/10.1530/ERC-12-0344
  14. Krysan K, Merchant FH, Zhu L, et al (2004). COX-2-dependent stabilization of survivin in non-small cell lung cancer. FASEB J, 18, 206-8. https://doi.org/10.1096/fj.03-0369fje
  15. Liu F1, Wei WQ, Cormier RT, et al (2014). Association of single nucleotide polymorphisms in the prostaglandinendoperoxide synthase 2 (PTGS2) and phospholipase A2 Group IIA (PLA2G2A) genes with susceptibility to esophageal squamous cell carcinoma. Asian Pac J Cancer Prev, 15, 1797-802. https://doi.org/10.7314/APJCP.2014.15.4.1797
  16. Louisa M, Soediro TM, Suyatna FD (2014). In vitro modulation of P-glycoprotein, MRP-1 and BCRP Expression by Mangiferin in Doxorubicin-Treated MCF-7 Cells. Asian Pac J Cancer Prev, 15, 1639-42. https://doi.org/10.7314/APJCP.2014.15.4.1639
  17. Madan D, Ferguson CG, Lee WY, et al (2013). Non-invasive imaging of tumors by monitoring autotaxin activity using an enzyme-activated near-infrared fluorogenic substrate. PLoS One, 8, 79065. https://doi.org/10.1371/journal.pone.0079065
  18. Marquardt JU, Andersen JB (2012). Next-generation sequencing: application in liver cancer-past, present and future? Biology, 1, 383-94. https://doi.org/10.3390/biology1020383
  19. Mita AC, Mita MM, Nawrocki ST, et al (2008). Survivin: key regulator of mitosis and apoptosis and novel target for cancer therapeutics. Clin Cancer Res, 14, 5000-5. https://doi.org/10.1158/1078-0432.CCR-08-0746
  20. Morandi A, Chiarugi P (2014). Metabolic implication of tumor:stroma crosstalk in breast cancer. J Mol Med, 92, 117-26. https://doi.org/10.1007/s00109-014-1124-7
  21. Nomura DK, Long JZ, Niessen S, et al (2010). Monoacylglycerol lipase regulates a fatty acid network that promotes cancer pathogenesis. Cell, 140, 49-61. https://doi.org/10.1016/j.cell.2009.11.027
  22. Pommier Y, Leo E, Zhang H, et al (2010). DNA topoisomerases and their poisoning by anticancer and antibacterial drugs. Chem Biol, 17, 421-33. https://doi.org/10.1016/j.chembiol.2010.04.012
  23. Prabhu AV, Krycer JR, Brown AJ (2013). Overexpression of a key regulator of lipid homeostasis, Scap, promotes respiration in prostate cancer cells. FEBS Lett, 587, 983-8. https://doi.org/10.1016/j.febslet.2013.02.040
  24. Srivastava AN, Gupta A, Srivastava S, et al (2010). Cisplatin combination chemotherapy induces oxidative stress in advance non small cell lung cancer patients. Asian Pac J Cancer Prev, 11, 465-71.
  25. Takaoka K, Kishimoto H, Segawa E, et al (2008). In vitro susceptibility to anticancer agents of the human KB carcinoma cell line transfected with COX-2 cDNA. Oncol Rep, 20, 645-9.
  26. Tani K, Naganawa A, Ishida A, et al (2001). Design and synthesis of a highly selective EP2-receptor agonist. Bioorg Med Chem Lett, 11, 2025-8. https://doi.org/10.1016/S0960-894X(01)00359-6
  27. Vander Heiden MG, Cantley LC, Thompson CB (2009). Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science, 324, 1029-33. https://doi.org/10.1126/science.1160809
  28. Weinstein D, Sarfstein R, Laron Z, et al (2014). Insulin receptor compensates for IGF1R inhibition and directly induces mitogenic activity in prostate cancer cells. Endocr Connect, 3, 24-35. https://doi.org/10.1530/EC-13-0086
  29. Woodward DF, Protzman CE, Krauss AH, et al (1993). Identification of 19 (R)-OH prostaglandin E2 as a selective prostanoid EP2-receptor agonist. Prostaglandins, 46, 371-83. https://doi.org/10.1016/0090-6980(93)90102-D

Cited by

  1. Effect of microRNA-21 on multidrug resistance reversal in A549/DDP human lung cancer cells vol.11, pp.1, 2014, https://doi.org/10.3892/mmr.2014.2662
  2. The Effects and Molecular Mechanisms of MiR-106a in Multidrug Resistance Reversal in Human Glioma U87/DDP and U251/G Cell Lines vol.10, pp.5, 2015, https://doi.org/10.1371/journal.pone.0125473
  3. Effect and mechanism of peroxisome proliferator-activated receptor-γ on the drug resistance of the U-87 MG/CDDP human malignant glioma cell line vol.12, pp.2, 2015, https://doi.org/10.3892/mmr.2015.3625
  4. Cytokine-induced killer cells induce apoptosis and inhibit the Akt/nuclear factor-κB signaling pathway in cisplatin-resistant human glioma U87MG cells vol.12, pp.5, 2015, https://doi.org/10.3892/mmr.2015.4236
  5. The role and mechanism of WEE1 on the cisplatin resistance reversal of the HepG2/DDP human hepatic cancer cell line pp.1792-1082, 2015, https://doi.org/10.3892/ol.2015.3647
  6. Effect of miR-155 knockdown on the reversal of doxorubicin resistance in human lung cancer A549/dox cells vol.11, pp.2, 2015, https://doi.org/10.3892/ol.2015.3995