Stanniocalcin1 (STC1) Inhibits Cell Proliferation and Invasion of Cervical Cancer Cells

Fengjie Guo, Yalin Li, Jiajia Wang, Yandong Li, Yuehui Li, Guancheng Li, Fengjie Guo, Yalin Li, Jiajia Wang, Yandong Li, Yuehui Li, Guancheng Li

Abstract

STC1 is a glycoprotein hormone involved in calcium/phosphate (Pi) homeostasis. There is mounting evidence that STC1 is tightly associated with the development of cancer. But the function of STC1 in cancer is not fully understood. Here, we found that STC1 is down-regulated in Clinical tissues of cervical cancer compared to the adjacent normal cervical tissues (15 cases). Subsequently, the expression of STC1 was knocked down by RNA interference in cervical cancer CaSki cells and the low expression promoted cell growth, migration and invasion. We also found that STC1 overexpression inhibited cell proliferation and invasion of cervical cancer cells. Moreover, STC1 overexpression sensitized CaSki cells to drugs. Further, we showed that NF-κB p65 protein directly bound to STC1 promoter and activated the expression of STC1 in cervical cancer cells. Thus, these results provided evidence that STC1 inhibited cell proliferation and invasion through NF-κB p65 activation in cervical cancer.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. The expression of STC1 was…
Figure 1. The expression of STC1 was down-regulated in cervical cancer tissues (15 cases).
(A) The expression level of STC1 mRNA in cancerous (T) and adjacent normal (N) matched cervical tissues was examined by RT-PCR. GAPDH served as a loading control. (B) Box plot graph showed the quantitative results of STC1 mRNA expression in all pairs of samples (p<0.05). (C) Protein level of STC1 in tumor and normal tissues were examined by immunohistochemistry analysis. Bar size: 100 µm. (D) Quantity analysis of protein level of STC1 expression. The intensity of reactivity was scored using a three-tier system (p<0.05). Data was expressed as mean ± SEM of three separated experiments. A value of P<0.05 was considered as statistical significance.
Figure 2. Down-regulation of STC1 promoted CaSki…
Figure 2. Down-regulation of STC1 promoted CaSki cells growth and invasion.
(A) Knock down of STC1 in CaSki cells. CaSki cells were transfected by STC1 targeting siRNA, and knockdown efficiency was shown by RT-PCR and western blotting. (B) MTT assays showed that the effect of decreased STC1 on CaSki cell growth. Following a 7-day period, the growth of CaSki/siRNA cells was much faster than CaSki/NC cells (*p<0.05). (C) Colony formation assay demonstrated the large number of cell colonies from CaSki/siRNA cells compared to CaSki/NC cells (p<0.05). (D) Wound healing assays showed the effect of STC1 on the migration of CaSki cells. CaSki/siRNA cells migrated faster compared to CaSki/NC cells (left panel). The relative migration distance of CaSki cells was calculated (right panel) (p<0.05). Bar size: 100 µm. (E) Matrigel invasion assays showed the effect of STC1 on the invasion of CaSki cells. The number of CaSki/siRNA cells on the filter surface was larger than CaSki/NC cells (left panel) (p<0.05). The mean value of invaded cells was shown in right panel. Bar size: 100 µm. (F) The growth curves of the xenografts were determined by tumor volume (left panel) (*p<0.05). The growth rates of the xenografts were valuated by tumor volume/days (right panel) (*p<0.05). CaSki/siRNA or CaSki/NC cells were injected subcutaneously into nude mice. (G) At end of experimental period, the final xenograft tumors were shown. (H) RT-PCR analyzed the expression of STC1 in representative xenograft tumors. (I) Representative images of histological inspection of xenograft tumors. The sections of xenograft tumors were stained with H&E. Bar size: 20 µm. Data was expressed as mean ± SEM of three separated experiments. Data was expressed as mean ± SEM of three separated experiments. A value of P<0.05 was considered as statistical significance.
Figure 3. Overexpression of STC1 inhibited cell…
Figure 3. Overexpression of STC1 inhibited cell proliferation and invasion of CaSki cells.
(A) Overexpression of STC1 in CaSki cells. STC1 expression vector was transfected into CaSki cells, and increased expression of STC1 was shown by RT-PCR and western blotting. (B) MTT assays showed that the growth of CaSki/STC1 cells was much slower than CaSki/NC cells (*p<0.05). (C) Colony formation assay showed that a small amount of cell colonies from CaSki/STC1 cells demonstrated a low activity (p<0.05). (D) Matrigel invasion assay revealed that up-regulation of STC1 mitigated the invasion of cells in vitro. Bar size: 100 µm. (E) STC1 overexpressed tumors emerged later and slowly grew compared to control tumors (*p<0.05). (F) At end of experimental period, the final weights of STC1 overexpressed tumors were found to be lower than controls. (G) RT-PCR of STC1 in xenograft tumors indicated that increased STC1 expression had been maintained throughout experimental time course. (H) H&E staining of STC1 overexpressed tumors showed a low nuclear/cytoplasmic ratio, and limited to the cancer nests compared to control tumors. Bar size: 20 µm. Data was expressed as mean ± SEM of three separated experiments. A value of P<0.05 was considered as statistical significance.
Figure 4. STC1 sensitized CaSki cells to…
Figure 4. STC1 sensitized CaSki cells to drugs.
(A) Effect of cisplatin on CaSki cells growth. CaSki cells were treated with with or without cisplatin (0, 1, 2, and 3 mg/L) for 96 h, removing aliquots every 24 h to evaluate cell viability. (B) Effect of thapsigargin on CaSki cells growth. CaSki cells were treated with with or without thapsigargin (0, 1, 3, 6, and 9 µM) for 72 h, removing aliquots every 24 h to evaluate cell viability. (C) Effect of rapamycin on CaSki cells growth. CaSki cells were treated with with or without rapamycin (0, 0.01, 0.1, 0.5, and 1 mg/L) for 72 h, removing aliquots every 24 h to evaluate cell viability. (D) STC1 sensitized CaSki cells to cisplatin. CaSki/STC1 or CaSki/NC cells were treated with cisplatin (2 mg/L) for 96 h. MTT assays detected the cell growth of CaSki/STC1 or CaSki/NC cells in the face of cisplatin (*p<0.05). (E) STC1 sensitized CaSki cells to thapsigargin. CaSki/STC1 or CaSki/NC cells were treated with thapsigargin (3 µM) for 72 h. MTT assays detected the cell growth of CaSki/STC1 or CaSki/NC cells in the face of thapsigargin (*p<0.05). (F) STC1 sensitized CaSki cells to rapamycin. CaSki/STC1 or CaSki/NC cells were treated with rapamycin (0.5 mg/L) for 72 h. MTT assays detected the cell growth of CaSki/STC1 or CaSki/NC cells in the face of rapamycin (*p<0.05). Data was expressed as mean ± SEM of three separated experiments. A value of P<0.05 was considered as statistical significance.
Figure 5. Direct binding of NF-κB p65…
Figure 5. Direct binding of NF-κB p65 protein to STC1 promoter and regulated the expression of STC1.
(A) The binding sites of STC1 were tested in CaSki cells by chromatin coimmunoprecipitation. The site was found to bind to NFκB p65. (B) Western blotting detected the activity of PARP, caspase-3, STC1, and NF-κB p65 in CaSki cells. CaSki cells were treated with thapsigargin (3 µM) for 12 h. (C) Western blotting detected the activity of p65, STC1, and caspase-3 in CaSki cells. CaSki cells were treated with increasing time of TNFα (10 mg/L) for 2.5 h. (D) Western blotting detected the activity of p65 and STC1 in CaSki cells. CaSki cells were treated with PDTC (10 µM) for 60 min. (E) Subcellular activity of p65 and STC1 in CaSki cells was analyzed by Western blotting. CaSki cells were treated with siRNA knockdown of p65 for 72 h. (F) The expression of p65 and STC1 in CaSki was detected by RT-PCR at mRNA level. CaSki cells were treated with siRNA knockdown of p65 for 48 h. (G) Schematic representation of some findings in this work.

References

    1. Jiang WQ, Chang AC, Satoh M, Furuichi Y, Tam PP, et al. (2000) The distribution of stanniocalcin 1 protein in fetal mouse tissues suggests a role in bone and muscle development. J Endocrinol 165 457–466.
    1. Yoshiko Y, Aubin JE (2004) Stanniocalcin 1 as a pleiotropic factor in mammals. Peptides 25 1663–1669.
    1. Olsen HS, Cepeda MA, Zhang QQ, Rosen CA, Vozzolo BL (1996) Human stanniocalcin: a possible hormonal regulator of mineral metabolism. Proc Natl Acad Sci USA 93: 1792–1796.
    1. Liu G, Yang G, Chang B, Mercado-Uribe I, Huang M, et al. (2010) Stanniocalcin 1 and ovarian tumorigenesis. J Natl Cancer Inst 102: 812–827.
    1. Law AY, Yeung BH, Ching L, Wong CK (2011) Sp1 is a transcription repressor to stanniocalcin-1 expression in TSA-treated human colon cancer cells, HT29. J Cell Biochem 112: 2089–2096.
    1. Wu S, Yoshiko Y, De Luca F (2006) Stanniocalcin 1 acts as a paracrine regulator of growth plate chondrogenesis. J Biol Chem 281: 5120–5127.
    1. Varghese R, Gagliardi AD, Bialek PE, Yee SP, Wagner GF, et al. (2002) Overexpression of human stanniocalcin affects growth and reproduction in transgenic mice. Endocrinology 143: 868–876.
    1. Hasilo CP, McCudden CR, Gillespie JR, James KA, Hirvi ER, et al. (2005) Nuclear targeting of stanniocalcin to mammary gland alveolar cells during pregnancy and lactation. Am J Phsiol Endoerinol Metab 289: 634–642.
    1. Filvaroff EH, Guillet S, Zlot C, Bao M, Ingle G, et al. (2002) Stanniocalcin 1 alter smuscle and bone structure and function in transgenic mice. Endocrinology 143: 3681–3690.
    1. Zaidi D, James KA, Wagner GF (2006) Passive immunization of lactating mice with stanniocalcin-1 antiserum reduces mammary gland development, milk fat content, and postnatal pup growth. Am J Physiol Endocrinol Metab 291: E974–981.
    1. Chang AC, Janosi J, Hulsbeek M, de Jong D, Jeffrey KJ, et al. (1995) A novel human cDNA highly homologous to the fish hormone stanniocalcin. Mol Cell Endocrinol 112: 241–247.
    1. McCudde CR, Majewski A, Chakrabarti S, Wagner GF (2004) Co-localizaztion of stanniocalcin-1 ligand and receptor in human breast carcinomas. Mol Cell Endocrinol 213: 167–172.
    1. Fujiwara Y, Sugita Y, Nakamori S, Miyamoto A, Shiozaki K, et al. (2000) Assessment of Stanniocalcin-1 mRNA as a molecular marker for micrometastases of various human cancers. Int J Oncol 16: 799–804.
    1. Okabe H, Satoh S, Kato T, Kitahara O, Yanagawa R, et al. (2001) Genome-wide analysis of gene expression in human hepatocellular carcinomas using cDNA microarray: identification of genes involved in viral carcinogenesis and tumor progression. Cancer Res 61: 2129–2137.
    1. Macartney-Coxson DP, Hood KA, Shi HJ, Ward T, Wiles A, et al. (2008) Metastatic susceptibility locus, an 8p hot-spot for tumour progression disrupted in colorectal liver metastases: 13 candidate genes examined at the DNA, mRNA and protein level. BMC Cancer 8: 187.
    1. Watanabe T, Ichihara M, Hashimoto M, Shimono K, Shimoyama Y, et al. (2002) Characterization of gene expression induced by RET with MEN2A or MEN2B mutation. Am J Pathol 161: 249–256.
    1. Ismail RS, Baldwin RL, Fang J, Browning D, Karlan BY, et al. (2000) Differential gene expression between normal and tumor-derived ovarian epithelial cells. Cancer Res 60: 6744–6749.
    1. Tohmiya Y, Koide Y, Fujimaki S, Harigae H, Funato T, et al. (2004) Stanniocalcin-1 as a novel marker to detect minimal residual disease of human leukemia. Tohoku J Exp Med 204: 125–133.
    1. Tamura S, Oshima T, Yoshihara K, Kanazawa A, Yamada T, et al. (2011) Clinical significance of STC1 gene expression in patients with colorectal cancer. Anticancer Res 31: 325–329.
    1. Gerritsen ME, Soriano R, Yang S, Ingle G, Zlot C, et al. (2002) In silico data filtering to identify new angiogenesis targets from a large in vitro gene profiling data set. Physiol Genomics 10: 13–20.
    1. Lal A, Peters H, St Croix B, Haroon ZA, Dewhirst MW, et al. (2001) Transcriptional response to hypoxia in human tumors. J Natl Cancer Inst 93: 1337–1343.
    1. Law AY, Lai KP, Lui WC, Wan HT, Wong CK (2008) Histone deacetylase inhibitor-induced cellular apoptosis involves stanniocalcin-1 activation. Exp Cell Res 314: 2975–2984.
    1. del Campo JM, Prat A, Gil-Moreno A, Pérez J, Parera M (2008) Update on novel therapeutic agents for cervical cancer. Gynecol Oncol 110: S72–76.
    1. Christensen SB, Skytte DM, Denmeade SR, Dionne C, Møller JV, et al. (2009) A Trojan horse in drug development: targeting of thapsigargins towards prostate cancer cells. Anticancer Agents Med Chem 9: 276–294.
    1. Fels DR, Ye J, Segan AT, Kridel SJ, Spiotto M, et al. (2008) Preferential cytotoxicity of bortezomib toward hypoxic tumor cells via overactivation of endoplasmic reticulum stress pathways. Cancer Res 68: 9323–9330.
    1. Diaz-Padilla I, Duran I, Clarke BA, Oza AM (2012) Biologic rationale and clinical activity of mTOR inhibitors in gynecological cancer. Cancer Treat Rev 38: 767–775.
    1. Fukasawa M, Tsuchiya T, Takayama E, Shinomiya N, Uyeda K, et al. (2004) Identification and characterization of the hypoxia-responsive element of the human placental 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase gene. J Biochem. 136: 273–277.
    1. Thompson JS, Asmis R, Tapp AA, Nelson B, Chu Y, et al. (. 2010) Pyrrolidine dithiocarbamate (PDTC) blocks apoptosis and promotes ionizing radiation-induced necrosis of freshly-isolated normal mouse spleen cells. Apoptosis 15: 705–714.
    1. Chang AC, Jellinek DA, Reddel RR (2003) Mammalian stanniocalcins and cancer. Endocr Relat Cancer 10: 359–373.
    1. Law AY, Ching LY, Lai KP, Wong CK (2010) Identification and characterization of the hypoxia-responsive element in human stanniocalcin-1 gene. Mol Cell Endocrinol 314: 118–127.
    1. Chen C, Jamaluddin MS, Yan S, Sheikh-Hamad D, Yao Q (2008) Human stanniocalcin-1 blocks TNF-alpha-induced monolayer permeability in human coronary artery endothelial cells. Arterioscler Thromb Vasc Biol 28: 906–912.
    1. Guo F, Li Y, Liu Y, Wang J, Li G (2010) ARL6IP1 mediates cisplatin-induced apoptosis in CaSki cervical cancer cells. Oncol Rep 23: 1449–1455.
    1. Li X, Shen L, Zhang J, Su J, Shen L, et al. (2007) Degradation of HER2 by Cbl-based chimeric ubiquitin ligases. Cancer Res 67: 8716–8724.

Source: PubMed

3
S'abonner