Skip to main content
Log in

Increased Expression of POSTN Predicts Poor Prognosis: a Potential Therapeutic Target for Gastric Cancer

  • Original Article
  • Published:
Journal of Gastrointestinal Surgery Aims and scope

Abstract

Background

Periostin (POSTN) is involved in many biological processes and is associated with the occurrence and development of several cancers, while its role in gastric cancer is not clear. We aimed to demonstrate the relationship between POSTN and gastric cancer based on publicly available data from The Cancer Genome Atlas (TCGA) database.

Methods

POSTN expression data and corresponding clinical information were downloaded from TCGA database. We compared the expression of POSTN in gastric cancer samples and normal samples. POSTN-related differentially expressed genes (DEGs) were identified for further functional enrichment analysis. In addition, the relationships between POSTN expression and clinicopathological features and survival in patients with gastric cancer were also investigated through univariate and multivariate Cox regression analyses. Furthermore, a nomogram was constructed to predict the survival probability of gastric cancer patients.

Results

POSTN expression in gastric cancer was significantly higher than that in normal gastric tissues (p < 0.001). High POSTN expression in gastric cancer was significantly related to poor prognostic features, including greater tumor extent (odds ratio [OR] = 1.638 for T3 and T4 vs. T1 and T2), worse histological type (OR = 0.329 for diffuse type vs. tubular type), and advanced histological grade (OR = 1.646 for grade 3 vs. grades 1 and 2) (all p < 0.05). The 118 identified DEGs were primarily enriched in pathways related to tumorigenesis and tumor progression, including the TGF-β signaling pathway, the WNT signaling pathway, and the signaling by VEGF. POSTN expression was positively correlated with the enrichment of the macrophages (r = 0.599, p < 0.001), helper T2 cells (r = 0.146, p = 0.005), and CD8 + T cells (r = 0.190, p < 0.001), but negatively correlated with the enrichment of Th17 cells (r =  − 0.130, p = 0.012) and NK CD56bright cells. Kaplan–Meier survival analysis showed that high POSTN expression is associated with a short overall survival (hazard ratio [HR] = 1.54; p = 0.011). In the multivariate cox regression analysis, high POSTN expression was confirmed to be an independent predictor of poor overall survival (HR = 1.681; p = 0.017). The constructed nomogram can well predict the 1 and 3 years overall survival probability of patients with GC (0.696 [95% CI, 0.671–0.721]).

Conclusion

POSTN plays an important role in the progression and prognosis of gastric cancer, and it may serve as a useful biomarker to predict survival in gastric cancer patients.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Sung H, Ferlay J, Rebecca L, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: Cancer J Clin  2021;0:1–41. https://doi.org/10.3322/caac.21660.

  2. Allemani C, Matsuda T, Di Carlo V, Harewood R, Matz M, et al. Global surveillance of trends in cancer survival 2000-14 (CONCORD-3): analysis of individual records for 37 513 025 patients diagnosed with one of18 cancers from 322 population-based registries in 71 countries. Lancet. (2018) 391:1023–75. https://doi.org/10.1016/S0140-6736(17)33326-3

    Article  PubMed  PubMed Central  Google Scholar 

  3. Winkler J, Abisoye-Ogunniyan A, Metcalf K, Werb Z. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat Commun. 2020 Oct 9;11(1):5120. https://doi.org/10.1038/s41467-020-18794-x

  4. Malanchi I, Santamaria-Martínez A, Susanto E, Peng H, Lehr H, et al. Interactions between cancer stem cells and their niche govern metastatic colonization. Nature (2011) 481, 85–89. https://doi.org/10.1038/nature10694

    Article  CAS  PubMed  Google Scholar 

  5. Ratajczak-Wielgomas K, Kmiecik A, Dziegiel P. Role of Periostin Expression in Non-Small Cell Lung Cancer: Periostin Silencing Inhibits the Migration and Invasion of Lung Cancer Cells via Regulation of MMP-2 Expression. Int J Mol Sci. 2022 22;23(3):1240. https://doi.org/10.3390/ijms23031240.

  6. Ma Y, He L, Zhao X, Li W, Lv X, et al. Protease activated receptor 2 signaling promotes self-renewal and metastasis in colorectal cancer through β-catenin and periostin. Cancer Lett. 2021 27;521:130-141. https://doi.org/10.1016/j.canlet.2021.08.032

    Article  CAS  Google Scholar 

  7. Yu B, Wu K, Wang X, Zhang J, Wang L, et al. Periostin secreted by cancer-associated fibroblasts promotes cancer stemness in head and neck cancer by activating protein tyrosine kinase 7. Cell Death Dis. 2018 22;9(11):1082. https://doi.org/10.1038/s41419-018-1116-6.

  8. Underwood T, Hayden A, Derouet M, Garcia E, Noble F, et al. Cancer-associated fibroblasts predict poor outcome and promote periostin-dependent invasion in oesophageal adenocarcinoma. J Pathol. 2015; 235(3):466-77. https://doi.org/10.1002/path.4467

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lv Y, Wang W, Jia W, Sun Q, Huang M, et al. High preoparative levels of serum periostin are associated with poor prognosis in patients with hepatocellular carcinoma after hepatectomy. Eur J Surg Oncol. 2013 Oct;39(10):1129-35. https://doi.org/10.1016/j.ejso.2013.06.023

  10. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. (2014) 15:550. https://doi.org/10.1186/s13059-014-0550-8

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. Omics. (2012) 16:284–7. https://doi.org/10.1089/omi.2011.0118

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Subramanian A, Tamayo P, Mootha V K, Mukherjee S, Ebert B, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A, 2005, 102(43): 15545-15550. https://doi.org/10.1073/pnas.0506580102

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Hänzelmann S, Castelo R, Guinney J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC bioinformatics 14.1 (2013): 1-15. https://doi.org/10.1186/1471-2105-14-7

    Article  Google Scholar 

  14. Bindea G, Mlecnik B, Tosolini M, Kirilovsky A, Waldner M, et al. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity 39.4 (2013): 782-795. https://doi.org/10.1016/j.immuni.2013.10.003

    Article  CAS  PubMed  Google Scholar 

  15. Szklarczyk D, Gable A, Lyon D, Junge A, Wyder S, et al. STRING v11: protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic acids research, 2019, 47(D1): D607-D613. https://doi.org/10.1093/nar/gky1131

    Article  CAS  PubMed  Google Scholar 

  16. Gabor C, Nepusz T. The igraph software package for complex network research. Inter Journal, complex systems 1695.5 (2006): 1–9. https://www.researchgate.net/publication/221995787

  17. Robin X, Turck N, Hainard A, Tiberti N, Lisacek F, et al. pROC: an open-source package for R and S+ to analyze and compare ROC curves. BMC Bioinformatics. (2011) 12:77. https://doi.org/10.1186/1471-2105-12-77

    Article  PubMed  PubMed Central  Google Scholar 

  18. Vivian J, Rao AA, Nothaft FA, Ketchum C, Armstrong J, et al. Toil enables reproducible, open source, big biomedical data analyses. Nat Biotechnol. (2017) 35:314-6. https://doi.org/10.1038/nbt.3772

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Wong M, Huang J, Chan P, Choi P, Lao X, et al. Global Incidence and Mortality of Gastric Cancer, 1980-2018. JAMA Netw Open. 2021 Jul 1;4(7):e2118457.

  20. Allemani C, Matsuda T, Carlo V, Harewood R, Matz M, et al. Global surveillance of trends in cancer survival 2000-14 (CONCORD-3): analysis of individual records for 37 513 025 patients diagnosed with one of 18 cancers from 322 population-based registries in 71 countries. Lancet. 2018 Mar 17;391(10125):1023-1075. https://doi.org/10.1016/S0140-6736(17)33326-3

  21. Yue H, Li W, Chen R, et al. Stromal POSTN induced by TGF-β1 facilitates the migration and invasion of ovarian cancer. Gynecol Oncol. 2021 Feb;160(2):530-538. https://doi.org/10.1016/j.ygyno.2020.11.026

  22. Ouyang G, Liu M, Ruan K, Song G, Mao Y, et al. Upregulated expression of periostin by hypoxia in non-small-cell lung cancer cells promotes cell survival via the Akt/PKB pathway. Cancer Lett. 2009 28;281(2):213–9. https://doi.org/10.1016/j.canlet.2009.02.030

  23. Kim G, Lee J, Park M, Yoon J. Epithelial periostin expression is correlated with poor survival in patients with invasive breast carcinoma. PLoS One. 2017 Nov 21;12(11):e0187635. https://doi.org/10.1371/journal.pone.0187635

  24. Labrèche C, Cook D, Abou-Hamad J, et al. Periostin gene expression in neu-positive breast cancer cells is regulated by a FGFR signaling cross talk with TGFβ/PI3K/AKT pathways. Breast Cancer Res. 2021 Nov 22;23(1):107. https://doi.org/10.1186/s13058-021-01487-8

  25. Bowen D, Joong S. Targeting epithelial-mesenchymal transition (EMT) to overcome drug resistance in cancer. Molecules, 2016, 21(7):965. https://doi.org/10.3390/molecules21070965

    Article  CAS  Google Scholar 

  26. Massague J. TGFβ in cancer. Cell. 2008;134(2):215-230. https://doi.org/10.1016/j.cell.2008.07.001

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Batlle E, Massague J. Transforming growth factor-beta signaling in immunity and cancer. Immunity. 2019;50(4):924-940. https://doi.org/10.1016/j.immuni.2019.03.024

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Yan Y, Zhang J, Li JH, et al. High tumor-associated macrophages infiltration is associated with poor prognosis and may contribute to the phenomenon of epithelial-mesenchymal transition in gastric cancer. OncoTargets and Therapy, 2016(9): 3975-3983. https://doi.org/10.2147/OTT.S103112

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Herr P, Hausmann G, Basler K. WNT secretion and signalling in human disease. Trends Mol Med, 2012;18(8):483-93. https://doi.org/10.1016/j.molmed.2012.06.008

    Article  CAS  PubMed  Google Scholar 

  30. Nusse R, Clevers H. Wnt/β-catenin signaling, disease, and emerging therapeutic modalities. Cell. 2017;169:985–99. https://doi.org/10.1016/j.cell.2017.05.016

    Article  CAS  PubMed  Google Scholar 

  31. Wang Y, Zheng L, Shang W, Yang Z, Li T, et al. Wnt/beta-catenin signaling confers ferroptosis resistance by targeting GPX4 in gastric cancer. Cell Death Differ. 2022. https://doi.org/10.1038/s41418-022-01008-w

    Article  PubMed  PubMed Central  Google Scholar 

  32. Pang L, Wang J, Fan Y, Xu R, Bai Y, et al. Correlations of TNM staging and lymph node metastasis of gastric cancer with MRI features and VEGF expression. Cancer Biomark. 2018;23(1):53-59. https://doi.org/10.3233/CBM-181287

    Article  CAS  PubMed  Google Scholar 

  33. Yan R, Peng X, Yuan X, et al. Suppression of growth and migration by blocking the Hedgehog signaling pathway in gastric cancer cells. Cell Oncol, 2013, 36(5):421-435. https://doi.org/10.1007/s13402-013-0149-1

    Article  CAS  Google Scholar 

  34. Murakami D, Tsujitani S, Osaki T, Saito H, Katano K, et al. Expression of phosphorylated Akt (pAkt) in gastric carcinoma predicts prognosis and efficacy of chemotherapy. Gastric Cancer. 2007;10(1):45-51. https://doi.org/10.1007/s10120-006-0410-7

    Article  CAS  PubMed  Google Scholar 

  35. Hu Y, Zheng M, Zhang R, Liang Y, Han H. Notch Signaling Pathway and Cancer Metastasis. Adv Exp Med Biol. 2012, 727, 186–198. https://doi.org/10.1007/978-1-4614-0899-4_14

    Article  CAS  PubMed  Google Scholar 

  36. Mantovani A, Sozzani S, Locati M, Allavena P, Sica A. Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 23, 549–555 (2002). https://doi.org/10.1016/s1471-4906(02)02302-5

    Article  CAS  PubMed  Google Scholar 

  37. Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature 454, 436–444. https://doi.org/10.1038/nature07205

  38. Qian BZ, Pollard JW. Macrophage diversity enhances tumor progression and metastasis. Cell 2010; 141(1):39-51. https://doi.org/10.1016/j.cell.2010.03.014.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity 2014; 41(1):49-61. https://doi.org/10.1016/j.immuni.2014.06.010.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Ubukata H, Motohashi G, Tabuchi T, Nagata H, Konishi S, et al. Evaluations of interferon-γ/interleuki‑4 ratio and neutrophil/lymphocyte ratio as prognostic indicators in gastric cancer patients. J Surg Oncol. 2010 1;102(7):742–7. https://doi.org/10.1002/jso.21725

  41. Chen J, Xia J, Liang X, Pan K, Wang W, et al. Intratumoral expression of IL‑17 and its prognostic role in gastric adenocarcinoma patients. Int. J. Biol. Sci. 7, 53–60 (2011). Int J Biol Sci. 2011 Jan 11;7(1):53–60. https://doi.org/10.7150/ijbs.7.53

Download references

Acknowledgements

The authors would like to thank Dr. Wei Yang for his assistance in data acquisition and guidance on the design of this study.

Funding

This study was supported by the Tackle Key Problems in Medicine of Henan Province (201003124, LHGJ20200188).

Author information

Authors and Affiliations

Authors

Contributions

Concept and design: SL. Acquisition, analysis, or interpretation of data: SL, WY, LP, and FM. Drafting of the manuscript: SL and WY. Critical revision of the manuscript for important intellectual content: ZZ and YH. Administrative, technical, or material support, and study supervision: ZZ and YH. All authors contributed to the article and approved the submitted version.

Corresponding authors

Correspondence to Yawei Hua, Wei Yang or Zhandong Zhang.

Ethics declarations

Consent to Participate

All data used in this research were obtained from TCGA, a publicly available cancer database, so according to Swedish law, this study was exempt from the need to obtain signed informed consent.

Conflict of Interest

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary Data Sheet 1

(XLSX 24.1 KB)

Supplementary Data Sheet 2

(XLSX 17.8 KB)

Supplementary Data Sheet 3

(XLSX 781 KB)

Supplementary Table 1

(DOCX 18 KB)

Supplementary Table 2

(DOCX 18 KB)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lu, S., Peng, L., Ma, F. et al. Increased Expression of POSTN Predicts Poor Prognosis: a Potential Therapeutic Target for Gastric Cancer. J Gastrointest Surg 27, 233–249 (2023). https://doi.org/10.1007/s11605-022-05517-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11605-022-05517-4

Keywords

Navigation