Overexpression of stathmin1 in the diffuse type of gastric cancer and its roles in proliferation and migration of gastric cancer cells

T-Y Jeon, M-E Han, Y-W Lee, Y-S Lee, G-H Kim, G-A Song, G-Y Hur, J-Y Kim, H-J Kim, S Yoon, S-Y Baek, B-S Kim, J-B Kim, S-O Oh, T-Y Jeon, M-E Han, Y-W Lee, Y-S Lee, G-H Kim, G-A Song, G-Y Hur, J-Y Kim, H-J Kim, S Yoon, S-Y Baek, B-S Kim, J-B Kim, S-O Oh

Abstract

Background: Stathmin1 is a microtubule-regulating protein that has an important role in the assembly and disassembly of the mitotic spindle. The roles of stathmin1 in carcinogenesis of various cancers, including prostate and breast cancer, have been explored. However, its expression and roles in gastric cancer have not yet been described.

Methods: Stathmin1 expression in paraffin-embedded tissue sections from 226 patients was analysed by immunohistochemistry. Roles of stathmin1 were studied using a specific small interfering RNA (siRNA).

Results: The expression of stathmin1 was positively correlated with lymph node metastasis, TNM stages and vascular invasion, and negatively with recurrence-free survival, in the diffuse type of gastric cancer. The median recurrence-free survival in patients with a negative and positive expression of stathmin1 was 17.0 and 7.0 months, respectively (P=0.009). When the expression of stathmin1 was knocked down using siRNA, the proliferation, migration and invasion of poorly differentiated gastric cancer cells in vitro were significantly inhibited. Moreover, stathmin1 siRNA transfection significantly slowed the growth of xenografts in nude mice.

Conclusion: These results suggest that stathmin1 can be a good prognostic factor for recurrence-free survival rate and is a therapeutic target in diffuse-type gastric cancer.

Figures

Figure 1
Figure 1
Immunohistochemical staining of stathmin1 in oral and gastric cancer sections. Anti-stathmin1 antibody was used for immunohistochemical staining in human oral and gastric cancer tissues as described in ‘Materials and Methods’ section. The invading oral cancer cells were positive for stathmin1 protein in the cytoplasm. The normal gastric mucosal cells were either negative or very weakly positive for stathmin1 protein in the cytoplasm. The invading gastric cancer cells were positive for stathmin1 protein. Scale bar, 200 μm.
Figure 2
Figure 2
Correlationship between the stathmin1 expression level and clinicopathological characteristics. Data of stathmin-positive patients with diffuse-type gastric cancer are presented. (A) The state of stathmin1 protein expression in patients with lymph node metastasis in diffuse type gastric cancer (n=17) and in patients without metastasis (n=7). Stathmin1 protein expression in patients with lymph node metastasis is significantly higher than in patients without lymph node metastasis (*P<0.05, Mann–Whitney U-test). (B) The state of stathmin1 protein expression in patients with early stages (I and II) of diffuse type gastric cancer (n=9) and patients with advanced stages (III and IV) of diffuse-type gastric cancer (n=15). Stathmin1 protein expression in advanced stages is significantly higher than that in early stages (**P<0.01, Mann–Whitney U-test). (C) The state of stathmin1 protein expression in patients with vascular invasion in diffuse-type gastric cancer (n=14) and in patients without vascular invasion (n=10). Stathmin1 expression in patients with vascular invasion is significantly higher than in patients without vascular invasion (**P<0.01, Mann–Whitney U-test).
Figure 3
Figure 3
Kaplan–Meier curves for recurrence-free survival according to stathmin1 overexpression in patients with gastric cancer ((A) diffuse type+intestinal type, P<0.05; (B) diffuse type only, P<0.01). When stathmin1 overexpression was observed, the recurrence-free survival rate decreased significantly, especially in patients with diffuse-type gastric cancer.
Figure 4
Figure 4
Stathmin1 was specifically downregulated by stathmin1siRNA. Cells were transfected with scrambled siRNA or stathmin1 siRNA. (A) Two days later, SNU638 and SNU16 cells were collected and stathmin1 protein levels were detected by western blotting. β-actin expression was monitored for normalisation. (B) SNU638 cells were collected and stathmin1 mRNA levels were examined by real-time PCR. Values are expressed as the percentage of control, which was defined as 100% (n=6). Results were analysed by one-way ANOVA, followed by Tukey's multiple comparison test. *P<0.01 vs control. mRNA, messenger RNA; SCR, scrambled; siRNA, small interfering RNA.
Figure 5
Figure 5
Effect of stathmin1 silencing on proliferation of gastric cancer cells. SNU638 (A) or SNU16 (B) cells were transfected with SCR siRNA or stathmin1 (STMN1) siRNA. After 5 days of incubation, cell proliferation was evaluated in WST assay. Data are expressed as percentage change (means±s.d.) compared with controls and represent four independent experiments. (*P<0.05, **P<0.01 vs SCR siRNA, one-way ANOVA followed by Tukey's multiple comparison). ANOVA, one-way analysis of variance; SCR, scrambled; siRNA, small interfering RNA.
Figure 6
Figure 6
Effect of stathmin1 silencing on the migration and invasion of gastric cancer cells. (A) Cell migration was evaluated in the Boyden migration assay two days after SNU638 cells were transfected with scrambled (SCR) small interfering RNA (siRNA) or stathmin1 siRNA. (B) Cell invasion was evaluated in the Matrigel invasion assay as described in the ‘Materials and Methods’ section. Data are expressed as percentage change (means±s.d.) compared with controls and represent four independent experiments. (*P<0.01 vs SCR siRNA, one-way analysis of variance (ANOVA) followed by Tukey's multiple comparison). Representative microscopic images were presented in the upper panel of each assay graph. ANOVA, one-way analysis of variance; SCR, scrambled; siRNA, small interfering RNA.
Figure 7
Figure 7
Effect of stathmin1 silencing on in vivo tumour growth. (A) SNU638 cells were transfected with scrambled (SCR) small interfering RNA (siRNA) or stathmin1 (STMN1) siRNA, and nude mice were inoculated subcutaneously with 2 × 106 cells at two sites per mouse. The tumour mass (xenograft) volume was measured every week from week 3 to week 7. Data are expressed as the means±s.d. and represent four independent experiments. (*P<0.05, **P<0.01 vs SCR siRNA, one-way analysis of variance (ANOVA) followed by Tukey's multiple comparison) (B) The tumour masses (xenograft) were dissected out 7 weeks later and presented. ANOVA, one-way analysis of variance; SCR, scrambled; siRNA, small interfering RNA.

References

    1. Belmont LD, Mitchison TJ (1996) Identification of a protein that interacts with tubulin dimers and increases the catastrophe rate of microtubules. Cell 84: 623–631
    1. Brattsand G (2000) Correlation of oncoprotein 18/stathmin expression in human breast cancer with established prognostic factors. Br J Cancer 83: 311–318
    1. Chen G, Wang H, Gharib TG, Huang CC, Thomas DG, Shedden KA, Kuick R, Taylor JM, Kardia SL, Misek DE, Giordano TJ, Iannettoni MD, Orringer MB, Hanash SM, Beer DG (2003) Overexpression of oncoprotein 18 correlates with poor differentiation in lung adenocarcinomas. Mol Cell Proteomics 2: 107–116
    1. Friedrich B, Gronberg H, Landstrom M, Gullberg M, Bergh A (1995) Differentiation-stage specific expression of oncoprotein 18 in human and rat prostatic adenocarcinoma. Prostate 27: 102–109
    1. Gavet O, Ozon S, Manceau V, Lawler S, Curmi P, Sobel A (1998) The stathmin phosphoprotein family: intracellular localization and effects on the microtubule network. J Cell Sci 111(Part 22): 3333–3346
    1. Giampietro C, Luzzati F, Gambarotta G, Giacobini P, Boda E, Fasolo A, Perroteau I (2005) Stathmin expression modulates migratory properties of GN-11 neurons in vitro. Endocrinology 146: 1825–1834
    1. Horwitz SB, Shen HJ, He L, Dittmar P, Neef R, Chen J, Schubart UK (1997) The microtubule-destabilizing activity of metablastin (p19) is controlled by phosphorylation. J Biol Chem 272: 8129–8132
    1. Jin K, Mao XO, Cottrell B, Schilling B, Xie L, Row RH, Sun Y, Peel A, Childs J, Gendeh G, Gibson BW, Greenberg DA (2004) Proteomic and immunochemical characterization of a role for stathmin in adult neurogenesis. FASEB J 18: 287–299
    1. Kouzu Y, Uzawa K, Koike H, Saito K, Nakashima D, Higo M, Endo Y, Kasamatsu A, Shiiba M, Bukawa H, Yokoe H, Tanzawa H (2006) Overexpression of stathmin in oral squamous-cell carcinoma: correlation with tumour progression and poor prognosis. Br J Cancer 94: 717–723
    1. Larsson N, Melander H, Marklund U, Osterman O, Gullberg M (1995) G2/M transition requires multisite phosphorylation of oncoprotein 18 by two distinct protein kinase systems. J Biol Chem 270: 14175–14183
    1. Lin WC, Li AF, Chi CW, Chung WW, Huang CL, Lui WY, Kung HJ, Wu CW (1999) tie-1 protein tyrosine kinase: a novel independent prognostic marker for gastric cancer. Clin Cancer Res 5: 1745–1751
    1. Luo XN, Mookerjee B, Ferrari A, Mistry S, Atweh GF (1994) Regulation of phosphoprotein p18 in leukemic cells. Cell cycle regulated phosphorylation by p34cdc2 kinase. J Biol Chem 269: 10312–10318
    1. Marklund U, Brattsand G, Shingler V, Gullberg M (1993) Serine 25 of oncoprotein 18 is a major cytosolic target for the mitogen-activated protein kinase. J Biol Chem 268: 15039–15047
    1. Marklund U, Larsson N, Gradin HM, Brattsand G, Gullberg M (1996) Oncoprotein 18 is a phosphorylation-responsive regulator of microtubule dynamics. EMBO J 15: 5290–5298
    1. Melhem RF, Strahler JR, Hailat N, Zhu XX, Hanash SM (1991) Involvement of OP18 in cell proliferation. Biochem Biophys Res Commun 179: 1649–1655
    1. Mistry SJ, Atweh GF (2001) Stathmin inhibition enhances okadaic acid-induced mitotic arrest: a potential role for stathmin in mitotic exit. J Biol Chem 276: 31209–31215
    1. Mistry SJ, Atweh GF (2006) Therapeutic interactions between stathmin inhibition and chemotherapeutic agents in prostate cancer. Mol Cancer Ther 5: 3248–3257
    1. Mistry SJ, Li HC, Atweh GF (1998) Role for protein phosphatases in the cell-cycle-regulated phosphorylation of stathmin. Biochem J 334(Part 1): 23–29
    1. Ozon S, Guichet A, Gavet O, Roth S, Sobel A (2002) Drosophila stathmin: a microtubule-destabilizing factor involved in nervous system formation. Mol Biol Cell 13: 698–710
    1. Qian F, Li YP, Sheng X, Zhang ZC, Song R, Dong W, Cao SX, Hua ZC, Xu Q (2007) PRL-3 siRNA inhibits the metastasis of B16-BL6 mouse melanoma cells in vitro and in vivo. Mol Med 13: 151–159
    1. Rubin CI, Atweh GF (2004) The role of stathmin in the regulation of the cell cycle. J Cell Biochem 93: 242–250
    1. Singer S, Ehemann V, Brauckhoff A, Keith M, Vreden S, Schirmacher P, Breuhahn K (2007) Protumorigenic overexpression of stathmin/Op18 by gain-of-function mutation in p53 in human hepatocarcinogenesis. Hepatology 46: 759–768
    1. Vogiatzi P, Vindigni C, Roviello F, Renieri A, Giordano A (2007) Deciphering the underlying genetic and epigenetic events leading to gastric carcinogenesis. J Cell Physiol 211: 287–295
    1. Wang R, Dong K, Lin F, Wang X, Gao P, Wei SH, Cheng SY, Zhang HZ (2007) Inhibiting proliferation and enhancing chemosensitivity to taxanes in osteosarcoma cells by RNA interference-mediated downregulation of stathmin expression. Mol Med 13: 567–575
    1. Workman P, Twentyman P, Balkwill F, Balmain A, Chaplin D, Double J, Embleton J, Newell D, Raymond R, Stables J, Stephens T, Wallace J (1998) United Kingdom Co-ordinating Committee on Cancer Research (UKCCCR) Guidelines for the welfare of animals in experimental neoplasia (second edition). Br J Cancer 77: 1–10
    1. Wu CW, Hsieh MC, Lo SS, Lui WY, P’Eng FK (1996) Results of curative gastrectomy for carcinoma of the distal third of the stomach. J Am Coll Surg 183: 201–207
    1. Wu CW, Hsieh MC, Lo SS, Tsay SH, Li AF, Lui WY, P’Eng FK (1997) Prognostic indicators for survival after curative resection for patients with carcinoma of the stomach. Dig Dis Sci 42: 1265–1269
    1. Yuasa Y (2003) Control of gut differentiation and intestinal-type gastric carcinogenesis. Nat Rev 3: 592–600

Source: PubMed

3
구독하다