DOI QR코드

DOI QR Code

Exogenous p53 Upregulated Modulator of Apoptosis (PUMA) Decreases Growth of Lung Cancer A549 Cells

  • Liu, Chun-Ju (Department of Biochemistry and Molecular Biology, Basic Medical College, Nan Chang University) ;
  • Zhang, Xia-Li (Department of Biochemistry and Molecular Biology, Basic Medical College, Nan Chang University) ;
  • Luo, Da-Ya (Department of Biochemistry and Molecular Biology, Basic Medical College, Nan Chang University) ;
  • Zhu, Wei-Feng (Department of Biochemistry and Molecular Biology, Basic Medical College, Nan Chang University) ;
  • Wan, Hui-Fang (Department of Biochemistry and Molecular Biology, Basic Medical College, Nan Chang University) ;
  • Yang, Jun-Ping (Clinical Laboratory of Jiangxi University of Traditional Chinese Medicine) ;
  • Yang, Xiao-Jun (Clinical Laboratory of Jiangxi University of Traditional Chinese Medicine) ;
  • Wan, Fu-Sheng (Department of Biochemistry and Molecular Biology, Basic Medical College, Nan Chang University)
  • Published : 2015.02.25

Abstract

Purpose: To investigate the influence of exogenous p53 upregulated modulator of apoptosis (PUMA) expression on cell proliferation and apoptosis in human non-small cell lung cancer A549 cells and transplanted tumor cell growth in nude mice. Materials and Methods: A549 cells were divided into the following groups: control, non-carrier (NC), PUMA (transfected with pCEP4-(HA) 2-PUMA plasmid), DDP ($10{\mu}g/mL$ cisplatin treatment) and PUMA+DDP (transfected with pCEP4-(HA)2-PUMA plasmid and $10{\mu}g/mL$ cisplatin treatment). The MTT method was used to detect the cell survival rate. Cell apoptosis rates were measured by flow cytometry, and PUMA, Bax and Bcl-2 protein expression levels were measured by Western blotting. Results: Compared to the control group, the PUMA, DDP and PUMA+DDP groups all had significantly decreased A549 cell proliferation (p<0.01), with the largest reduction in the PUMA+DDP group. Conversely, the apoptosis rates of the three groups were significantly increased (P<0.01), and the PUMA and DDP treatments were synergistic. Moreover, Bax protein levels significantly increased (p<0.01), while Bcl-2 protein levels significantly decreased (p<0.01). Finally, both the volume and the weights of transplanted tumors were significantly reduced (p<0.01), and the inhibition ratio of the PUMA+DDP group was significantly higher than in the single DDP or PUMA groups. Conclusions: Exogenous PUMA effectively inhibited lung cancer A549 cell proliferation and transplanted tumor growth by increasing Bax protein levels and reducing Bcl-2 protein levels.

Keywords

References

  1. Avila JL, Grundmann O, Burd R, et al (2009). Radiation-induced salivary gland dysfunction results from p53-dependent apoptosis. Int J Radiat Oncol Biol Phys, 73, 523-9. https://doi.org/10.1016/j.ijrobp.2008.09.036
  2. Berna MJ, Igarashi H, Jensen RT, et al (2012). Multiple endocrine neoplasia type 1. Indian J Endocrinol Metab, 12, 272-295.
  3. Chen Y, Qian H, Wang H, et al (2007). Ad-PUMA sensitizes drug-resistant choriocarcinoma cells to chemotherapeuticagents. Gynecol Oncol, 107, 505-12. https://doi.org/10.1016/j.ygyno.2007.08.007
  4. Gao R, Yu Y, Inoue A, et al (2013). Heterogeneous nuclear ribonucleo protein K (hnRNP-K) promotes tumor metastasis by induction of genes involved in extracellular matrix, cell movement and angiogenesis. J Biol Chem, 4, 380-405.
  5. Han JW (2001). Expression of bbc3, apro-apoptotic BH3-only gene, is regulated by diverse cell death and survival signals. Proc Natl Acad Sci USA, 98, 113-8. https://doi.org/10.1073/pnas.98.1.113
  6. Jeffers JR, Parganas E, Lee Y, et al (2003). Puma is an essential mediator of p53-dependent and p53-independent apoptotic pathways. Cancer Cell, 4, 321-8. https://doi.org/10.1016/S1535-6108(03)00244-7
  7. Kuo L, Thengchaisri N, Hein TW, et al (2012). Regulation of coronaryvasomotor function by reactive oxygen species. Mol Med Ther, 8, 473-86.
  8. Lovet JM, Burroughs A, Bruix J (2003). Hepatocellular carcinoma. Lancet, 362, 1907-17. https://doi.org/10.1016/S0140-6736(03)14964-1
  9. Meng J, Fang B, Liao Y, et al (2010). Apoptosis induction by MEK inhibition in human lung cancer cells is mediated by Bim. Plos One, 5, 13026-052. https://doi.org/10.1371/journal.pone.0013026
  10. Niizuma K, Endo H, Nito C et al (2009). Potential role of PUMA in delayed death of hippocampal CA1 neurons after transient global cerebral ischemia. Stroke, 40, 618-25. https://doi.org/10.1161/STROKEAHA.108.524447
  11. Nakano K, Vousden KH (2001). PUMA, a novel proapoptotic gene, is induced by p53. Mol Cell, 7, 683-94. https://doi.org/10.1016/S1097-2765(01)00214-3
  12. Perez BA, Ghafoori AP, Lee CL, et al (2013). Assessing the radiation response of lung cancer with different gene mutations using genetically engineered mice. Front Oncol, 3, 533-50.
  13. Rouibaa F, Bakkar M, Seddik H, et al (2013). Interest of expandable metallic stents in the management of colonic tumor occlusion: experience of a Moroccan hospital. Pan Afr Med J, 2, 245-69.
  14. Roberts CG, Millar EK, O'Toole SA, et al (2011). Identification of PUMA as an estrogen target gene that mediates the apoptotic response to tamoxifen in human breast cancer cells and predicts patient outcome and tamoxifen responsiveness in breast cancer. Oncogene, 30, 3186-97. https://doi.org/10.1038/onc.2011.36
  15. Singh S, Chhipa RR, Vijayakumar MV, et al (2006). DNA damaging drugs-induced down-regulation of Bcl-2 is essential for nduction of apoptosis in high-risk HPV-positive A549 and KB cells. Cancer lett, 36, 213-21.
  16. Sun Q, Sakaida T, Yue W, et al (2007). Chemosensitization of head and neck cancer cells by PUMA. Mol Cancer Ther, 6, 3180-88. https://doi.org/10.1158/1535-7163.MCT-07-0265
  17. Wang H, Pei W, Luan Q, et al (2012). A feasibility study on gene therapy of pancreatic carcinoma with Ad-PUMA. Cancer Biol Ther, 13, 712-9. https://doi.org/10.4161/cbt.20552
  18. Wang H, Qian H, Yu J, et al (2006). Administration of PUMA adenovirus increases the sensitivity of esophageal cancer cells to anticancer drugs. Cancer Biol Ther, 5, 380-5. https://doi.org/10.4161/cbt.5.4.2477
  19. Watanabe A, Taniguchi F , Izawa M, et al (2009). The role of survivin in the resistance of endom etriotic stromal cells to drug - induced apop osis. Hum Reprod, 5, 1109-31.
  20. Wan HF, Yu LH, Wu JL, et al (2013). Effect of diallyl trisulfide on SKOV-3/DDP cells apoptosis in human ovarian cancer. Asian Pac J Cancer Prev, 14, 7197-201. https://doi.org/10.7314/APJCP.2013.14.12.7197
  21. Wang P, Yu J, Zhang L, et al (2007). The nuclear function of p53 is required for PUMA-mediated apoptosis induced by DNA damage. Proc Natl Acad Sci USA, 104, 4054-9. https://doi.org/10.1073/pnas.0700020104
  22. Yu J. PUMA, a potent killer with or without p53. Oncogene, 27, 71-83.
  23. Yee KS, Vousden KH (2008). Contribution of membrane localization to the apoptotic activity of PUMA. Apoptosis, 13, 87-95. https://doi.org/10.1007/s10495-007-0140-2
  24. Yu J, Yue W, Wu B, et al (2006). PUMA sensitizes lung cancer cells to chemotherapeutic agents and irradiation. Clin Cancer Res, 12, 2928-36. https://doi.org/10.1158/1078-0432.CCR-05-2429
  25. Yu J, Zhang L, Hwang PM, et al (2001). PUMA induces the rapid apoptosis of colorectal cancer cells. Mol Cell, 7, 673-82. https://doi.org/10.1016/S1097-2765(01)00213-1
  26. Zaffaroni N, Costa A, Pennati M, et al (2007). Survivin is highly expressed and promotes cell surv ival in malignant peritoneal mesothelioma. Cell Oncol, 29, 453-66.
  27. Zhang XL, Tu S, Wang YB, et al (2014). The mechanism of taurine-induced apoptosis in human colon cancer cells. Acta Biochim Biophys Sin, 46, 261-72. https://doi.org/10.1093/abbs/gmu004

Cited by

  1. Treatment with a Small Synthetic Compound, KMU-193, induces Apoptosis in A549 Human Lung Carcinoma Cells through p53 Up-Regulation vol.16, pp.14, 2015, https://doi.org/10.7314/APJCP.2015.16.14.5883
  2. Growth inhibitory and apoptosis-inducing effects of allergen-free Rhus verniciflua Stokes extract on A549 human lung cancer cells vol.36, pp.5, 2016, https://doi.org/10.3892/or.2016.5131
  3. Purified vitexin compound 1, a new neolignan isolated compound, promotes PUMA-dependent apoptosis in colorectal cancer pp.20457634, 2018, https://doi.org/10.1002/cam4.1769
  4. A novel benzamine lead compound of histone deacetylase inhibitor ZINC24469384 can suppresses HepG2 cells proliferation by upregulating NR1H4 vol.9, pp.1, 2019, https://doi.org/10.1038/s41598-019-39487-6