Yonsei Med J.  2016 May;57(3):557-564. 10.3349/ymj.2016.57.3.557.

Isoprenaline Induces Periostin Expression in Gastric Cancer

Affiliations
  • 1Department of General Surgery, Chinese PLA General Hospital, Beijing, China. weibo@vip.163.com, 13381326128@163.com
  • 2Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Trauma Center of Postgraduate Medical School, Chinese PLA General Hospital, Beijing, China.
  • 3Department of Pathology, Chengde Medical College, Chengde, Hebei Province, China.

Abstract

PURPOSE
Periostin mediates critical steps in gastric cancer and is involved in various signaling pathways. However, the roles of periostin in promoting gastric cancer metastasis are not clear. The aim of this study was to investigate the relevance between periostin expression and gastric cancer progression and the role of stress-related hormones in the regulation of cancer development and progression.
MATERIALS AND METHODS
Normal, cancerous and metastatic gastric tissues were collected from patients diagnosed with advanced gastric cancer. The in vivo expression of periostin was evaluated by in situ hybridization and immunofluorescent staining. Meanwhile, human gastric adenocarcinoma cell lines MKN-45 and BGC-803 were used to detect the in vitro expression of periostin by using quantitative real-time polymerase chain reaction (PCR) and western blotting.
RESULTS
Periostin is expressed in the stroma of the primary gastric tumors and metastases, but not in normal gastric tissue. In addition, we observed that periostin is located mainly in pericryptal fibroblasts, but not in the tumor cells, and strongly correlated to the expression of α-smooth muscle actin (SMA). Furthermore, the distribution patterns of periostin were broader as the clinical staging of tumors progressed. We also identified a role of stress-related signaling in promoting cancer development and progression, and found that isoprenaline upregulated expression levels of periostin in gastric cancer cells.
CONCLUSION
These findings suggest that the distribution pattern of periostin was broader as the clinical staging of the tumor progressed and found that isoprenaline upregulated expression levels of periostin in gastric cancer cells.

Keyword

Gastric cancer; stress; metastasis; periostin; extracellular matrix; nich

MeSH Terms

Adenocarcinoma/*metabolism/pathology
Adrenergic beta-Agonists/pharmacology
Aged
Blotting, Western
Cell Adhesion Molecules/drug effects/*metabolism
Cell Line, Tumor
Fibroblasts/*metabolism
Gene Expression Regulation, Neoplastic/*drug effects
Humans
Isoproterenol/*pharmacology
Male
Neoplasm Staging
RNA, Messenger/genetics/metabolism
Real-Time Polymerase Chain Reaction
Signal Transduction
Stomach/metabolism/pathology
Stomach Neoplasms/*metabolism/pathology
Up-Regulation
Adrenergic beta-Agonists
Cell Adhesion Molecules
Isoproterenol
RNA, Messenger

Figure

  • Fig. 1 Representative fluorescence images showing different expression patterns of periostin in peri-cancerous (A), cancerous (B), and metastatic gastric cancer tissue samples (C). In normal gastric tissues, periostin expression was absent in gastric epithelial cells and the stroma of gastric tissues (A). In cancerous tissues, periostin was upregulated and present in the stroma of primary tumors, but not in gastric epithelial cells (B). In metastatic gastric cancer tissues, periostin was detected in the areas of lymphatic metastasis (C).

  • Fig. 2 Periostin expression correlated to the expression of fibroblast-associated protein α-smooth muscle actin (SMA) in gastric cancer tissues. Periostin was located in the pericryptal fibroblasts. (A and B) Representative fluorescence images show different expression patterns of periostin in gastric cancer tissue samples. In cancerous tissue where periostin was expressed at low levels, the expression of α-SMA strongly correlated with the distribution patterns of periostin (A). In cancerous tissues where periostin showed high expression, α-SMA was also expressed at high levels in the membrane of cancerous gastric epithelial cells (B).

  • Fig. 3 The distribution patterns of periostin show a much broader distribution while the clinical staging of the tumor progressed. Periostin was distributed at low levels in early gastric cancer (A). Periostin in intermediate gastric carcinoma shows a much broader distribution than in early gastric cancer (B). The distribution of periostin was highest in late gastric cancer (C).

  • Fig. 4 Isoprenaline upregulated expression levels of periostin in gastric cancer cells. (A) Real-time PCR analyses of periostin expression in MKN-45 and MGC-803 cells after isoprenaline stimulation. Isoprenaline induced periostin expression in gastric cancer cells. (B) After incubation with 10 µM isoprenaline, periostin mRNA levels increased by 4.9-fold in MKN-45 and 1.4-fold in MGC-803 gastric cancer cells compared with control groups. (C and D) MKN-45 and MGC-803 cells were stimulated with 0, 5, or 10 µM isoprenaline after serum starvation. Periostin expression was analyzed by Western blot analysis. (E) Relative expression of periostin in gastric cancer cell lines after isoprenaline treatment. Data represent mean±SD (n=3 for each group, *p<0.05, †p<0.01, ‡p<0.001). POSTN, periostin.

  • Fig. 5 Isoprenaline upregulated periostin expression in MGC-803 gastric cancer cells in a time-dependent manner. (A) MGC-803 cells were starved overnight, and then treated with 5 µM isoprenaline for 0, 1, 2, 3, and 6 hour. The relative expression of periostin in MGC-803 cells was evaluated by real-time PCR. The results show that isoprenaline induced periostin expression in MGC-803 gastric cancer cells in a time-dependent manner. (B) Western blot analysis show that isoprenaline stimulation also upregulated periostin expression in a time-dependent manner in MGC-803 gastric cancer cells. (C) Relative expression of periostin in gastric cancer cells after isoprenaline treatment for indicated time periods. Data represent means±SD (n=3 for each group, *p<0.05, †p<0.01, †p<0.001). POSTN, periostin.


Reference

1. Orditura M, Galizia G, Sforza V, Gambardella V, Fabozzi A, Laterza MM, et al. Treatment of gastric cancer. World J Gastroenterol. 2014; 20:1635–1649.
Article
2. Neri S, Hashimoto H, Kii H, Watanabe H, Masutomi K, Kuwata T, et al. Cancer cell invasion driven by extracellular matrix remodeling is dependent on the properties of cancer-associated fibroblasts. J Cancer Res Clin Oncol. 2016; 142:437–446.
Article
3. Scherzer MT, Waigel S, Donninger H, Arumugam V, Zacharias W, Clark G, et al. Fibroblast-derived extracellular matrices: an alternative cell culture system that increases metastatic cellular properties. PLoS One. 2015; 10:e0138065.
Article
4. Bhakta G, Lim ZX, Rai B, Lin T, Hui JH, Prestwich GD, et al. The influence of collagen and hyaluronan matrices on the delivery and bioactivity of bone morphogenetic protein-2 and ectopic bone formation. Acta Biomater. 2013; 9:9098–9106.
Article
5. Nissinen LM, Kähäri VM. Collagen turnover in wound repair--a macrophage connection. J Invest Dermatol. 2015; 135:2350–2352.
Article
6. Williams PA, Silva EA. The role of synthetic extracellular matrices in endothelial progenitor cell homing for treatment of vascular disease. Ann Biomed Eng. 2015; 43:2301–2313.
Article
7. Miles FL, Sikes RA. Insidious changes in stromal matrix fuel cancer progression. Mol Cancer Res. 2014; 12:297–312.
Article
8. Liu Y, Liu BA. Enhanced proliferation, invasion, and epithelial-mesenchymal transition of nicotine-promoted gastric cancer by periostin. World J Gastroenterol. 2011; 17:2674–2680.
Article
9. Li JS, Sun GW, Wei XY, Tang WH. Expression of periostin and its clinicopathological relevance in gastric cancer. World J Gastroenterol. 2007; 13:5261–5266.
Article
10. Contié S, Voorzanger-Rousselot N, Litvin J, Clézardin P, Garnero P. Increased expression and serum levels of the stromal cell-secreted protein periostin in breast cancer bone metastases. Int J Cancer. 2011; 128:352–360.
Article
11. Liu Y, Shi J, Chen M, Cao YF, Liu YW, Pan J, et al. Periostin: a novel prognostic predictor for meningiomas. J Neurooncol. 2015; 121:505–512.
Article
12. Kikuchi Y, Kunita A, Iwata C, Komura D, Nishiyama T, Shimazu K, et al. The niche component periostin is produced by cancer-associated fibroblasts, supporting growth of gastric cancer through ERK activation. Am J Pathol. 2014; 184:859–870.
Article
13. Sampieri K, Fodde R. Cancer stem cells and metastasis. Semin Cancer Biol. 2012; 22:187–193.
Article
14. Hu Q, Tong S, Zhao X, Ding W, Gou Y, Xu K, et al. Periostin mediates TGF-β-induced epithelial mesenchymal transition in prostate cancer cells. Cell Physiol Biochem. 2015; 36:799–809.
Article
15. Ratajczak-Wielgomas K, Dziegiel P. The role of periostin in neoplastic processes. Folia Histochem Cytobiol. 2015; 53:120–132.
Article
16. Malanchi I, Santamaria-Martínez A, Susanto E, Peng H, Lehr HA, Delaloye JF, et al. Interactions between cancer stem cells and their niche govern metastatic colonization. Nature. 2011; 481:85–89.
Article
17. Morra L, Moch H. Periostin expression and epithelial-mesenchymal transition in cancer: a review and an update. Virchows Arch. 2011; 459:465–475.
Article
18. Kashima TG, Nishiyama T, Shimazu K, Shimazaki M, Kii I, Grigoriadis AE, et al. Periostin, a novel marker of intramembranous ossification, is expressed in fibrous dysplasia and in c-Fos-overexpressing bone lesions. Hum Pathol. 2009; 40:226–237.
Article
19. Kudo Y, Ogawa I, Kitajima S, Kitagawa M, Kawai H, Gaffney PM, et al. Periostin promotes invasion and anchorage-independent growth in the metastatic process of head and neck cancer. Cancer Res. 2006; 66:6928–6935.
Article
20. Zhu S, Belkhiri A, El-Rifai W. DARPP-32 increases interactions between epidermal growth factor receptor and ERBB3 to promote tumor resistance to gefitinib. Gastroenterology. 2011; 141:1738–1748.e1-2.
Article
21. Neverauskiene S, Machtejeviene E, Vaitkiene D, Juodzbaliene EB. [Disseminated ovarian, bone, and bone marrow metastases from gastric cancer]. Medicina (Kaunas). 2006; 42:923–931.
22. Pein M, Oskarsson T. Microenvironment in metastasis: roadblocks and supportive niches. Am J Physiol Cell Physiol. 2015; 309:C627–C638.
Article
23. Liu GX, Xi HQ, Sun XY, Wei B. Role of periostin and its antagonist PNDA-3 in gastric cancer metastasis. World J Gastroenterol. 2015; 21:2605–2613.
Article
24. Valastyan S, Weinberg RA. Tumor metastasis: molecular insights and evolving paradigms. Cell. 2011; 147:275–292.
Article
25. Is¸eri OD, Sahin FI, Terzi YK, Yurtcu E, Erdem SR, Sarialioglu F. beta-Adrenoreceptor antagonists reduce cancer cell proliferation, invasion, and migration. Pharm Biol. 2014; 52:1374–1381.
Article
26. Lu YJ, Geng ZJ, Sun XY, Li YH, Fu XB, Zhao XY, et al. Isoprenaline induces epithelial-mesenchymal transition in gastric cancer cells. Mol Cell Biochem. 2015; 408:1–13.
Article
Full Text Links
  • YMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr