DOI QR코드

DOI QR Code

In vitro Study of Nucleostemin as a Potential Therapeutic Target in Human Breast Carcinoma SKBR-3 Cells

  • Guo, Yu (Bioscience Department, Department of laboratory medicine, Bengbu Medical College) ;
  • Liao, Ya-Ping (Bioscience Department, Department of laboratory medicine, Bengbu Medical College) ;
  • Zhang, Ding (Bioscience Department, Department of laboratory medicine, Bengbu Medical College) ;
  • Xu, Li-Sha (Bioscience Department, Department of laboratory medicine, Bengbu Medical College) ;
  • Li, Na (Bioscience Department, Department of laboratory medicine, Bengbu Medical College) ;
  • Guan, Wei-Jun (Institute of Animal Science, Chinese Academy of Agricultural Science) ;
  • Liu, Chang-Qing (Bioscience Department, Department of laboratory medicine, Bengbu Medical College)
  • Published : 2014.03.01

Abstract

Although nucleolar protein nucleostemin (NS) is essential for cell proliferation and early embryogenesis and expression has been observed in some types of human cancer and stem cells, the molecular mechanisms involved in mediation of cell proliferation and cell cycling remains largely elusive. The aim of the present study was to evaluate NS as a potential target for gene therapy of human breast carcinoma by investigating NS gene expression and its effects on SKBR-3 cell proliferation and apoptosis. NS mRNA and protein were both found to be highly expressed in all detected cancer cell lines. The apoptotic rate of the pcDNA3.1-NS-Silencer group ($12.1-15.4{\pm}3.8%$) was significantly higher than those of pcDNA3.1-NS ($7.2-12.0{\pm}1.7%$) and non-transfection groups ($4.1-6.5{\pm}1.8%$, P<0.01). MTT assays showed the knockdown of NS expression reduced the proliferation rate of SKBR-3 cells significantly. Matrigel invasion and wound healing assays indicated that the number of invading cells was significantly decreased in the pcDNA3.1-NS-siRNA group (P<0.01), but there were no significant difference between non-transfected and over-expression groups (P>0.05). Moreover, RNAi-mediated NS down-regulation induced SKBR-3 cell G1 phase arrest, inhibited cell proliferation, and promoted p53 pathway-mediated cell apoptosis in SKBR-3 cells. NS might thus be an important regulator in the G2/M check point of cell cycle, blocking SKBR-3 cell progression through the G1/S phase. On the whole, these results suggest NS might be a tumor suppressor and important therapeutic target in human cancers.

Keywords

References

  1. Fan Y, Liu Z, Zhao S, et al (2006). Nucleostemin mRNA is expressed in both normal and malignant renal tissues. Br J Cancer, 94, 1658-62.
  2. Gao HX, Gao XF, Wang GQ, et al (2012). In vitro study of Nucleostemin gene as a potential therapeutic target for human lung carcinoma. Biomed Environ Sci, 25, 91-7.
  3. Gil-Ranedo J, Mendiburu-Elicabe M, Garcia-Villanueva M, et al (2011). An off-target nucleostemin RNAi inhibits growth in human glioblastoma-derived cancer stem cells. PLoS One, 6, e28753. https://doi.org/10.1371/journal.pone.0028753
  4. Gu Y, Li H, Miki J, et al (2006). Phenotypic characterization of telomerase-immortalized primary non-malignant and malignant tumor-derived human prostate epithelial cell lines. Exp Cell Res, 312, 831-43. https://doi.org/10.1016/j.yexcr.2005.11.029
  5. Jafarnejad SM, Mowla SJ, and Matin MM (2008). Knockingdown the expression of nucleostemin significantly decreases rate of proliferation of rat bone marrow stromal stem cells in an apparently p53-independent manner. Cell Prolif, 41, 28-35. https://doi.org/10.1111/j.1365-2184.2007.00505.x
  6. Kafienah W, Mistry S, Williams C, and Hollander AP (2006). Nucleostemin is a marker of proliferating stromal stem cells in adult human bone marrow. Stem Cells, 24, 1113-20. https://doi.org/10.1634/stemcells.2005-0416
  7. Kano M, Seki N, Kikkawa N, et al (2010). miR-145, miR-133a and miR-133b: Tumor-suppressive miRNAs target FSCN1 in esophageal squamous cell carcinoma. Int J Cancer, 127, 2804-14. https://doi.org/10.1002/ijc.25284
  8. Ma J, Zhang Q, Chen S, et al (2013). Mitochondrial dysfunction promotes breast cancer cell migration and invasion through HIF1alpha accumulation via increased production of reactive oxygen species. PLoS One, 8, e69485. https://doi.org/10.1371/journal.pone.0069485
  9. Meng L, Lin T, and Tsai RY (2008). Nucleoplasmic mobilization of nucleostemin stabilizes MDM2 and promotes G2-M progression and cell survival. J Cell Sci, 121, 4037-46. https://doi.org/10.1242/jcs.037952
  10. Nomura J, Maruyama M, Katano M, et al (2009). Differential requirement for nucleostemin in embryonic stem cell and neural stem cell viability. Stem Cells, 27, 1066-76. https://doi.org/10.1002/stem.44
  11. Romanova L, Kellner S, Katoku-Kikyo N, and Kikyo N (2009). Novel role of nucleostemin in the maintenance of nucleolar architecture and integrity of small nucleolar ribonucleoproteins and the telomerase complex. J Biol Chem, 284, 26685-94. https://doi.org/10.1074/jbc.M109.013342
  12. Sakamoto Y, Terashita N, Muraguchi T, et al (2013). Effects of epigallocatechin-3-gallate (EGCG) on A549 lung cancer tumor growth and angiogenesis. Biosci Biotechnol Biochem, 77, 1799-803. https://doi.org/10.1271/bbb.120882
  13. Schon O, Friedler A, Bycroft M, et al (2002). Molecular mechanism of the interaction between MDM2 and p53. J Mol Biol, 323, 491-501. https://doi.org/10.1016/S0022-2836(02)00852-5
  14. Tamase A, Muraguchi T, Naka K, et al (2009). Identification of tumor-initiating cells in a highly aggressive brain tumor using promoter activity of nucleostemin. Proc Natl Acad Sci U S A, 106, 17163-8. https://doi.org/10.1073/pnas.0905016106
  15. Tsai RY (2009). Nucleolar modulation of TRF1: a dynamic way to regulate telomere and cell cycle by nucleostemin and GNL3L. Cell Cycle, 8, 2912-6.
  16. Tsai RY, and McKay RD (2002). A nucleolar mechanism controlling cell proliferation in stem cells and cancer cells. Genes Dev, 16, 2991-3003. https://doi.org/10.1101/gad.55671
  17. Zhong LR, Chen X, and Wei KM (2013). Radix tetrastigma hemsleyani flavone induces apoptosis in human lung carcinoma a549 cells by modulating the MAPK pathway. Asian Pac J Cancer Prev, 14, 5983-7. https://doi.org/10.7314/APJCP.2013.14.10.5983
  18. Zhu Q, Meng L, Hsu JK, et al (2009). GNL3L stabilizes the TRF1 complex and promotes mitotic transition. J Cell Biol, 185, 827-39. https://doi.org/10.1083/jcb.200812121
  19. Zhu Q, Yasumoto H, and Tsai RY (2006). Nucleostemin delays cellular senescence and negatively regulates TRF1 protein stability. Mol Cell Biol, 26, 9279-90. https://doi.org/10.1128/MCB.00724-06

Cited by

  1. Betaine Effects on Morphology, Proliferation, and p53-induced Apoptosis of HeLa Cervical Carcinoma Cells in Vitro vol.16, pp.8, 2015, https://doi.org/10.7314/APJCP.2015.16.8.3195
  2. Nucleostemin exerts anti-apoptotic function via p53 signaling pathway in cardiomyocytes vol.51, pp.10, 2015, https://doi.org/10.1007/s11626-015-9934-7
  3. Gene expression profiling of NB4 cells following knockdown of nucleostemin using DNA microarrays vol.14, pp.1, 2016, https://doi.org/10.3892/mmr.2016.5213