Decreased ST2 expression is associated with gastric cancer progression and pathogenesis

  • Authors:
    • Feihu Bai
    • Fangyun Ba
    • Yanjie You
    • Yaning Feng
    • Wei Tao
    • Chuanxia Wu
    • Mengna Jiu
    • Yongzhan Nie
  • View Affiliations

  • Published online on: April 5, 2019     https://doi.org/10.3892/ol.2019.10223
  • Pages: 5761-5767
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gastric cancer is a type of cancer with increasing incidence and high mortality rates, but molecular biomarkers of diagnostic and therapeutic value are currently lacking. The aim of the present study was to examine the expression pattern of the interleukin 1 receptor‑like 1 (ST2) protein and assess its clinicopathological significance in gastric cancer. Western blot analysis of 12 gastric cancer specimens and paired adjacent tissues demonstrated that the protein levels of 2 isoforms of ST2, soluble secreted ST2 and the ST2 variant without the third immunoglobulin motif and splicing in the C‑terminal, were markedly decreased in cancer tissues compared with non‑cancerous tissues. Immunohistochemical analysis demonstrated that ST2 protein expression was markedly decreased in primary gastric cancer tissues (39.1%, 90/230) compared with adjacent non‑cancerous tissues (60.7%, 54/89) (P<0.05). Statistical analysis demonstrated that decreased ST2 expression was significantly associated with advanced tumor node metastasis stage (P<0.001) and tumor differentiation (P<0.001). These data suggest that ST2 protein may be a valuable biomarker of gastric cancer progression and pathogenesis.

Introduction

Gastric cancer has a poor prognosis that is largely attributable to early and frequent metastasis (13). Surgery and combined radio-chemotherapeutic regimens are associated with modest survival benefits in advanced gastric cancer, with an overall 5-year survival rate of <24% (3). At present, the mechanisms underlying the initiation and progression of gastric cancer remain unclear and molecular markers for gastric cancer remain to be identified. It is imperative to identify the biological and molecular changes that frequently occur during gastric carcinogenesis, in order to elucidate cancer pathology and identify new diagnostic markers, in order to individualize treatment for patients with gastric cancer.

Interleukin 1 receptor-like 1 (ST2/IL1RL1), also known as T1, DER4 or FIT-1, which was originally identified as a primary responsive gene, is a member of the interleukin (IL)-1 receptor family and is highly induced by growth stimulation and oncogenic Ras-induced signaling (4). Based on alternative splicing and processing of mRNA, ST2 exists as 4 isoforms, including a transmembrane full-length form (ST2L), also known as IL1RL1-b, a soluble secreted form (sST2), also known as IL1RL1-a, a variant without the third immunoglobulin motif and splicing in the C-terminal (ST2v) and a fourth that has yet to be fully characterized (46). ST2 is produced by various types of immune cells produce ST2 including mast cells, macrophages and dendritic cells, and non-immune cells, including endothelial, epithelial, smooth muscle and fibroblast cells (7). The widespread expression of the ST2 gene in different cell types indicates that it may have important functions across a broad spectrum of biological systems. A recent functional study demonstrated that ST2 is the ligand-binding component of the interleukin-33 (IL-33) receptor, whereas sST2 is considered as a decoy receptor preventing IL-33 signaling (8). ST2L stimulated by IL-33 was demonstrated to activate the transcription factor through common signaling molecules to interleukin-1 receptor (IL-1R), including myeloid differentiation primary response protein MyD88 (MyD88), MyD88-adapter-like, the Toll/IL-1R domain of the cytoplasmic region of IL1RL1, IL-1R-associated kinase, tumor necrosis factor receptor-associated factor 6 and NF-κB, and the mitogen-activated protein kinase family (8,9). ST2/IL-33 signaling has also been identified to be involved in T-cell mediated immune responses, particularly Th2 cells and Th2-associated cytokines (9). IL-33 binds to ST2 and induces the expression of interferon, IL-4, IL-5 and IL-13, thereby leading to severe pathological changes in mucosal organs. ST2L has previously been used as a marker for Th2 cells. In addition, sST2 may also serve a key role in downregulating the inflammatory response (10). It has also been suggested that ST2L/IL-33 participates in a number of inflammatory immune response processes associated with asthma, allergic diseases, autoimmune diseases, cardiovascular diseases and other conditions associated with acute heart failure, myocardial infarction, respiratory failure and acute trauma (11,12). Previous studies revealed that ST2 was involved in the pathogenesis and prognosis of multiple types of cancer, including glioblastoma, breast and pancreatic cancer, and leukemia (1316). However, to the best of our knowledge, the role of ST2 in gastric cancer remains to be elucidated. The aim of the present study was to investigate the expression status of the 2 primary splice variants, sST2 and ST2v, by western blot analysis, and to evaluate ST2 protein expression in gastric cancer using immunohistochemistry and determine its clinicopathological significance.

Patients and methods

Patient information and tissue specimens

A microarray chip containing 178 samples of gastric cancer tissues from Beijing Hua Nuo Aomei Biotechnology, Inc. was utilized. A second cohort of 52 formalin-fixed paraffin-embedded tumor specimens was collected from patients who underwent surgery at the Xijing Hospital Affiliated to The Fourth Military Medical University (Xi'an, China) between September 2006 and January 2012. There were 230 patients in total, including 69 females and 161 males (median age, 54 years; range, 31–75 years) included in the present study. The patients were diagnosed as follows: 104 cases of gastric antrum carcinoma; 76 cases of carcinoma of the gastric cardia; and 50 cases of gastric body carcinoma. A total of 116 adjacent non-cancerous tissue specimens were also collected as controls. Tumor grade and stage were classified in accordance with the International Union against Cancer/American Joint Committee on Cancer pathological Tumor-Node-Metastasis (TNM) classification, 7th edition (2010) (17). In addition, 12 independent primary gastric cancer tissues and matched adjacent non-cancerous samples were frozen and stored in liquid nitrogen for western blot analysis. Signed informed consent was obtained from the patients prior to tissue sample collection. The study protocol conformed to the ethical guidelines outlined in the Declaration of Helsinki and was approved by The Institutional Review Board (approval no. 07-170) of Ningxia Hui Autonomous Region People's Hospital (Ningxia Hui Autonomous Region, China).

Western blot analysis

This procedure was performed as described previously (1821). Frozen tissue samples, including gastric cancer tissues and paired adjacent non-cancerous tissues obtained from 12 patients were prepared in radioimmunoprecipitation assay lysis buffer (Beyotime Institute of Biotechnology). A total of 20 µg proteins were separated by 8% SDS-PAGE and transferred to a polyvinylidene fluoride membrane (EMD Millipore). The membranes were incubated in blocking buffer (TBS with 0.1% Tween and 5% non-fat dry milk) for 1 h at 37°C and then incubated with a rabbit polyclonal anti-ST2 antibody (cat. no. 11920-1-AP; dilution, 1:500; ProteinTech Group, Inc.) in blocking buffer overnight at 4°C, followed by a horseradish peroxidase-conjugated secondary antibody against rabbit IgG (cat. no. sc-2004; dilution, 1:2,000; Santa Cruz Biotechnology, Inc.) for 1 h at 37°C. Signals were visualized with the enhanced chemiluminescence system according to the manufacturer's protocol (Amersham; GE Healthcare). The blots were probed with an anti-β-actin monoclonal antibody (cat. no. ab8226; dilution, 1:2,000; Abcam) as the control. Densitometric analyses of protein expression levels were performed using Bio-Rad Quantity One software (version 4.5.2; Bio-Rad Laboratories, Inc.). Expression was considered to be decreased when the ratio of expression in tumor and paired non-cancerous tissue was <2.

Immunofluorescence microscopy

Immunofluorescence microscopy was performed as described previously (18,19). Sections (4 µm) cut from tissue specimens fixed in 10% buffered formalin and embedded in paraffin were deparaffinized, rehydrated (anhydrous ethanol gradient, 100, 95, 80 and 70%) and incubated with 0.3% H2O2 for 20 min at 37°C. Non-specific blocking was performed by using 10% goat serum (Sigma-Aldrich; Merck KGaA) containing 0.05% Tweeen-20 for 30 min at 37°C. Glass slides were coated with a rabbit polyclonal anti-ST2 antibody (cat. no. 11920-1-AP; dilution, 1:50; ProteinTech Group, Inc.) overnight at 4°C. The tissue sections were washed with PBS containing 0.05% Tween-20 and then incubated with a fluorescein isothiocyanate-conjugated goat anti-rabbit secondary antibody (cat. no. sc-2012; dilution, 1:2,000; Santa Cruz Biotechnology, Inc.) for 1 h at room temperature. The sections were counterstained with propidium iodide (PI; Sigma-Aldrich; Merck KGaA) for 5 min at 37°C in the dark (final concentration of PI, 50 µg/ml). Finally, the slides were washed twice with PBS and were examined under an Olympus Flouview FV1000 confocal laser scanning microscope (Olympus Corporation) at ×400 magnification.

Immunohistochemistry and evaluation

Immunohistochemical analysis was performed as described previously (20,21). Sections (4 µm thick) were cut from tissue specimens fixed in 10% buffered formalin and embedded in paraffin. After deparaffinization (xylene, 2 times and 10 min each at 37°C) and rehydration (alcohol gradient, 100, 95, 80 and 70%), endogenous peroxidase activity was blocked with 0.3% hydrogen peroxide for 30 min at room temperature. For antigen retrieval, tissue sections were autoclaved at 121°C in citrate buffer (10 mM, pH 6.0) for 10 min. The sections were blocked for 1 h in 5% bovine serum albumin (Sigma-Aldrich; Merck KGaA) in PBS at 37°C, and were subsequently incubated with a rabbit polyclonal anti-ST2 antibody (cat. no. 11920-1-AP; dilution, 1:50; ProteinTech Group, Inc.) overnight at 4°C. Staining was visualized using an EnVision antibody complex method. An EnVision kit (OriGene Technologies, Inc.) was used and 3,3′-diaminobenzidine was used as the chromogen. Nuclei were counterstained with 0.5% hematoxylin for 2 min at room temperature. Sections immunostained with the normal rabbit IgG (cat. no. PP501P; dilution, 1:50; OriGene Technologies, Inc.) as the primary antibody were used as negative controls. A total of 10 random microscopic fields/slide, were evaluated by 2 independent observers who were blinded to the clinical information, at a magnification of ×400 using a light microscope (Carl Zeiss AG, Oberkochen, Germany). ST2 staining was assessed using a semi-quantitative approach, which combined the staining intensity and proportion of positive cells. The mean percentage of positively-stained cells was scored as follows: 0, 0–5%; 1, 5–25%; 2, 26–50%; 3, 51–75%; and 4, 76–100%. Staining intensity was categorized as follows: 0, Absent; 1, weak; 2, moderate; and 3, strong. The multiplication of staining intensity and percentage of positive cells was used as the final staining score. For statistical evaluation, the tumor samples with final staining scores of <3 were classed as negative ST2 expression and those with scores ≥3 as positive ST2 expression.

Statistical analysis

Statistical analysis was performed using the SPSS 17.0 statistical software package (SPSS, Inc., Chicago, IL, USA). The association between the expression levels of ST2 with different clinical variables was assessed using Fisher's exact test or Pearson's χ2 test as appropriate. P<0.05 was considered to indicate a statistically significant difference.

Results

Protein expression levels of ST2 in cancer and non-cancerous tissues

In light of the data indicating that ST2 is a frequently silenced candidate tumor suppressor gene during tumorigenesis (14), the present study first examined whether the expression of ST2 was altered at the protein level in tumors compared with normal tissues. In 12 pairs of gastric cancer tissues and their adjacent non-cancerous tissues, 2 bands were observed with molecular weights corresponding to the size of sST2 (30 kDa) and ST2v (66 kDa) (Fig. 1). Notably, 75% (9/12) of gastric cancer tissues exhibited decreased sST2 expression compared with the adjacent non-cancerous tissues (P<0.001). Similarly, decreased ST2v expression levels were also identified in 66.7% (8/12) of the tumors compared with the adjacent non-cancerous tissues (P<0.01).

The expression profile of ST2 in a cohort of 230 primary gastric cancer tissues and 116 adjacent non-cancerous tissues was additionally investigated by immunohistochemistry. ST2 expression was observed primarily in the cytoplasm of neoplastic cells, although it was also observed to a certain extent in the cell membrane (Fig. 2). Positive expression of the ST2 protein was observed in 39.1% (90/230) of tumor specimens and in 60.7% (54/89) of adjacent non-cancerous tissues (P<0.05). Taken together, these results clearly indicate that ST2 expression is frequently downregulated in gastric cancer tissues compared with normal controls.

Cellular localization of ST2 expression in gastric cancer

To determine the cellular localization of ST2 expression, immunofluorescence analysis was performed. Consistent with the results of the immunohistochemistry analysis, positive immunostaining for ST2 was observed in the cytoplasm and the membrane of cancer cells, whereas marked ST2 immunoreactivity was predominantly identified in the cytoplasm of epithelial cells in non-cancerous tissues (Fig. 3).

Clinicopathological implications of ST2 expression

The potential associations between ST2 protein levels and the clinicopathological characteristics of 230 gastric cancer specimens were next assessed. As demonstrated in Table I, decreased ST2 expression was identified to be significantly associated with decreased differentiation (P<0.001) and advanced TNM stage (P<0.001). No significant association was observed between ST2 expression and the other clinicopathological factors, including patient age and sex. In summary, these data indicated that decreased ST2 expression is closely associated with parameters implicated in gastric cancer progression and pathogenesis.

Table I.

Associations between ST2 expression and clinicopathological variables.

Table I.

Associations between ST2 expression and clinicopathological variables.

ST2 expression

VariablesPatients, nNegative, nPositive, nP-value
Sex
  Male161101600.38
  Female  69  3930
Age, years
  ≤61134  85150.44
  >61  96  5520
Differentiationa
  Well  37     928<0.001
  Moderate  72  3834
  Poor  94  7123
TNM staging
  I/II143  7667<0.001
  III/IV  87  6423

a 27 cases of mucinous adenocarcinoma were censored. ST2, interleukin 1 receptor-like 1; TNM, tumor node metastasis.

Discussion

ST2 has been identified as a regulatory effector for Th2-type responses, and as a negative feedback regulator in pro-inflammatory responses (4). ST2 also has a significant effect on the pathogenesis of several diseases, including inflammation, allergies, fibrillation, cardiac hypertrophy and rheumatoid arthritis (22). Numerous studies have suggested that ST2 expression is associated with carcinogenesis and tumor progression in multiple cancer types (1316,22). ST2 has been shown to regulate innate and acquired immunity in tumors. The biological functions of ST2 in cancer growth and progression are primarily mediated by the IL-33/ST2 signaling pathway, which compromises the integrity of the intestinal barrier and promotes the production of pro-tumorigenic IL-6 by immune cells (22).

However, the exact physiological and pathological functions of ST2 in cancer development and progression remain to be elucidated (23,24). In the present study, the protein expression of the ST2 gene in human gastric cancer tissues and their matched non-cancerous tissues was analyzed by western blot analysis and immunohistochemistry. A total of 2 variant subtypes of the ST2 protein were also identified in gastric cancer tissues, sST2 and ST2v, with corresponding molecular weights of 30 and 66 kDa, respectively. Notably, the expression levels of ST2 among normal tissues adjacent to the tumor were variable across the patient cohort. This discrepancy may be due to the difference between individuals. However, 75% (9/12) of gastric cancer tissues exhibited a significantly decreased sST2 expression compared with the adjacent non-cancerous tissues. This observation suggested that ST2 protein expression was markedly decreased in cancer tissues compared with non-cancerous tissues. The present analysis in gastric cancer indicated an association between negative ST2 expression and tumor progression phenotype, including advanced tumor stage and poor tumor differentiation. These results suggested that ST2 may act as a tumor suppressor gene in gastric cancer, consistent with a previous study in glioblastoma cells (24), but are inconsistent with other studies in breast and colorectal cancer (23,25). Notably, a previous study involving gastric cancer revealed that IL-33 enhanced tumor cell invasion and migration via the ST2-ERK1/2 pathway, and knockdown of the IL-33 receptor ST2 attenuates the IL-33-mediated malignant phenotypes (26). It may be concluded that this disparity may be due to the intrinsic differences among tumor types.

It has been demonstrated that the ST2 protein may be involved in the immunity of the tumor microenvironment. IL-33 combined with IL-1 and other inflammatory cytokines enhance the expression of ST2, resulting in the activation of oncogenes (27). It has been suggested that knockdown of ST2L in mice may lead to the inhibition of mammary tumor growth and metastasis, followed by enhanced circulating levels of pro-inflammatory cytokines and activation of natural killer and CD8+ T cells (28). Accumulating evidence suggests that ST2 may serve as an inflammatory cytokine in promoting cancer development and the production of Th2 cell-associated cytokines, including IL-4, IL-5 and IL-13. These cytokines have been detected in the microenvironment of several tumors (2931). Taking into consideration the complexity of the tumor microenvironment, the physiological and molecular mechanisms of action of the ST2 protein in gastric cancer remain to be elucidated.

There were certain limitations to the present study. Additional validation of the results in a larger series of patients with gastric cancer will strengthen the results of the present study and improve the understanding of the clinical behavior of ST2. In addition, a number of factors are able to contribute to abnormal gene silencing in cancer, and not all aberrant gene silencing is involved in tumor development. Therefore, candidate tumor suppressor genes, the expression levels of which are downregulated in tumors, require additional investigation for their biological roles in cancer development and progression.

In the present study, several methods were used to characterize the ST2 expression profile in gastric cancer. Downregulated ST2 expression is a molecular signature of gastric cancer progression and pathogenesis. Therefore, it may be concluded that ST2 not only appears to be a promising diagnostic biomarker, but also a potential treatment target in gastric cancer.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Natural Science Foundation of Ningxia, China (grant no., 2018AAC02016), the National Natural Science Foundation of China (grant nos. 81760440 and 81860426), the Regional Science and Technology Development Program Conducted by the Central Government of China (grant no. YDZX20176400004650) and the Foundation of Ningxia Medical University (grant no. XM2016077).

Availability of data and materials

The datasets used and/or analyzed in the present study are available from the corresponding authors on reasonable request.

Authors' contributions

FBai, YY and YN conceived, designed the experiments and drafted the manuscript. FBa, YF, WT, CW and MJ performed the experiments. YF and WT analyzed the data.

Ethics approval and consent to participate

Signed informed consent was obtained from the patients prior to tissue sample collection. The study protocol conformed to the ethical guidelines outlined in the Declaration of Helsinki and was approved by the Institutional Review Board (approval no. 07-170) of Ningxia Hui Autonomous Region People's Hospital.

Patient consent for publication

Signed informed consent was obtained from the patients prior to tissue sample collection.

Competing interests

The authors declare that they have no competing interests.

References

1 

Yonemura Y, Endou Y, Sasaki T, Hirano M, Mizumoto A, Matsuda T, Takao N, Ichinose M, Miura M and Li Y: Surgical treatment for peritoneal carcinomatosis from gastric cancer. Eur J Surg Oncol. 36:1131–1138. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Bozzetti F, Yu W, Baratti D, Kusamura S and Deraco M: Locoregional treatment of peritoneal carcinomatosis from gastric cancer. J Surg Oncol. 98:273–276. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Yoo CH, Noh SH, Shin DW, Choi SH and Min JS: Recurrence following curative resection for gastric carcinoma. Br J Surg. 87:236–242. 2000. View Article : Google Scholar : PubMed/NCBI

4 

Iwahana H, Yanagisawa K, Ito-Kosaka A, Kuroiwa K, Tago K, Komatsu N, Katashima R, Itakura M and Tominaga S: Different promoter usage and multiple transcription initiation sites of the interleukin-1 receptor-related human ST2 gene in UT-7 and TM12 cells. Eur J Biochem. 264:397–406. 1999. View Article : Google Scholar : PubMed/NCBI

5 

Bergers G, Reikerstorfer A, Braselmann S, Graninger P and Busslinger M: Alternative promoter usage of the Fos-responsive gene Fit-1 generates mRNA isoforms coding for either secreted or membrane-bound proteins related to the IL-1 receptor. EMBO J. 13:1176–1188. 1994. View Article : Google Scholar : PubMed/NCBI

6 

Hayakawa H, Hayakawa M, Kume A and Tominaga S: Soluble ST2 blocks interleukin-33 signaling in allergic airway inflammation. J Biol Chem. 282:26369–26380. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Trajkovic V, Sweet MJ and Xu D: T1/ST2-an IL-1receptor-like modulator of immune responses. Cytokine Growth Factor Rev. 15:87–95. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Hentschke I, Graser A, Melichar VO, Kiefer A, Zimmermann T, Kroß B, Haag P, Xepapadaki P, Papadopoulos NG, Bogdan C and Finotto S: IL-33/ST2 immune responses to respiratory bacteria in pediatric asthma. Sci Rep 7:43426. 2017. View Article : Google Scholar

9 

Yamamoto M, Sato S, Hemmi H, Hoshino K, Kaisho T, Sanjo H, Takeuchi O, Sugiyama M, Okabe M, Takeda K and Akira S: Role of adaptor TRIF in the MyD88-independent Toll-like receptor signaling pathway. Science. 301:640–643. 2003. View Article : Google Scholar : PubMed/NCBI

10 

Siede J, Fröhlich A, Datsi A, Hegazy AN, Varga DV, Holecska V, Saito H, Nakae S and Löhning M: IL-33 receptor-expressing regulatory T cells are highly activated, Th2 biased and suppress CD4 T cell proliferation through IL-10 and TGFβ release. PLoS One. 11:e01615072016. View Article : Google Scholar : PubMed/NCBI

11 

van Vark LC, Lesman-Leegte I, Baart SJ, Postmus D, Pinto YM, Orsel JG, Westenbrink BD, Brunner-la Rocca HP, van Miltenburg AJM, Boersma E, Hillege HL, Akkerhuis KM; TRIUMPH Investigators, ; et al: Prognostic value of serial ST2 measurements in patients with acute heart failure. J Am Coll Cardiol. 70:2378–2388. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Ko FW, Yan BP, Lam YY, Chu JH, Chan KP and Hui DS: Undiagnosed airflow limitation is common in patients with coronary artery disease and associated with cardiac stress. Respirology. 21:137–142. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Zhang JF, Wang P, Yan YJ, Li Y, Guan MW, Yu JJ and Wang XD: IL-33 enhances glioma cell migration and invasion by upregulation of MMP2 and MMP9 via the ST2-NF-κB pathway. Oncol Rep. 38:2033–2042. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Jovanovic I, Radosavljevic G, Mitrovic M, Juranic VL, McKenzie AN, Arsenijevic N, Jonjic S and Lukic ML: ST2 deletion enhances innate and acquired immunity to murine mammary carcinoma. Eur J Immunol. 41:1902–1912. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Fang Y, Zhao L, Xiao H, Cook KM, Bai Q, Herrick EJ, Chen X, Qin C, Zhu Z, Wakefield MR and Nicholl MB: IL-33 acts as a foe to MIA PaCa-2 pancreatic cancer. Med Oncol. 34:232017. View Article : Google Scholar : PubMed/NCBI

16 

Yoshida K, Arai T, Yokota T, Komatsu N, Miura Y, Yanagisawa K, Tetsuka T and Tominaga S: Studies on natural ST2 gene products in the human leukemic cell line UT-7 using monoclonal antihuman ST2 antibodies. Hybridoma. 14:419–427. 1995. View Article : Google Scholar : PubMed/NCBI

17 

You Y, Bai F, Ye Z, Zhang N, Yao L, Tang Y and Li X: Downregulated CDK10 expression in gastric cancer: Association with tumor progression and poor prognosis. Mol Med Rep. 17:6812–6818. 2018.PubMed/NCBI

18 

Lin C, Xin S, Qin X, Li H, Lin L and You Y: Zoledronic acid suppresses metastasis of esophageal squamous cell carcinoma cells through upregulating the tight junction protein occuludin. Cytotechnology. 68:1233–1241. 2016. View Article : Google Scholar : PubMed/NCBI

19 

You Y, Yang W, Qin X, Wang F, Li H, Lin C, Li W, Gu C, Zhang Y and Ran Y: ECRG4 acts as a tumor suppressor and as a determinant of chemotherapy resistance in human nasopharyngeal carcinoma. Cell Oncol (Dordr). 38:205–214. 2015. View Article : Google Scholar : PubMed/NCBI

20 

You Y, Li H, Qin X, Ran Y and Wang F: Down-regulated ECRG4 expression in breast cancer and its correlation with tumor progression and poor prognosis-a short report. Cell Oncol (Dordr). 39:89–95. 2016. View Article : Google Scholar : PubMed/NCBI

21 

You Y, Li H, Qin X, Zhang Y, Song W, Ran Y and Gao F: Decreased CDK10 expression correlates with lymph node metastasis and predicts poor outcome in breast cancer patients-a short report. Cell Oncol (Dordr). 38:485–491. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Mertz KD, Mager LF, Wasmer MH, Thiesler T, Koelzer VH, Ruzzante G, Joller S, Murdoch JR, Brümmendorf T, Genitsch V, et al: The IL-33/ST2 pathway contributes to intestinal tumorigenesis in humans and mice. Oncoimmunology. 5:e10629662015. View Article : Google Scholar : PubMed/NCBI

23 

Dominguez D, Ye C, Geng Z, Chen S, Fan J, Qin L, Long A, Wang L, Zhang Z, Zhang Y, et al: Exogenous IL-33 restores dendritic cell activation and maturation in established cancer. J Immunol. 198:1365–1375. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Haga Y, Yanagisawa K, Ohto-Ozaki H, Tominaga S, Masuzawa T and Iwahana H: The effect of ST2 gene product on anchorage-independent growth of a glioblastoma cell line, T98G. Eur J Biochem. 270:163–170. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Jovanovic IP, Pejnovic NN, Radosavljevic GD, Pantic JM, Milovanovic MZ, Arsenijevic NN and Lukic ML: Interleukin-33/ST2 axis promotes breast cancer growth and metastases by facilitating intratumoral accumulation of immunosuppressive and innate lymphoid cells. Int J Cancer. 134:1669–1682. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Yu XX, Hu Z, Shen X, Dong LY, Zhou WZ and Hu WH: IL-33 promotes gastric cancer cell invasion and migration Via ST2-ERK1/2 pathway. Dig Dis Sci. 60:1265–1272. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Sun P, Ben Q, Tu S, Dong W, Qi X and Wu Y: Serun interleukin-33 levels in patients with gastric cancer. Dig Dis Sci. 56:3596–3601. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Schmieder A, Multhoff G and Radons J: Interleukin-33 acts as a pro-inflammatory cytokine and modulates its receptor gene expression in highly metastatic human pancreatic carcinoma cells. Cytokine. 60:514–521. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Levescot A, Flamant S, Basbous S, Jacomet F, Féraud O, Anne Bourgeois E, Bonnet ML, Giraud C, Roy L, Barra A, et al: BCR-ABL-induced deregulation of the IL-33/ST2 pathway in CD34+ progenitors from chronic myeloid leukemia patients. Cancer Res. 74:2669–2676. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Qin L, Dominguez D, Chen S, Fan J, Long A, Zhang M, Fang D, Zhang Y, Kuzel TM and Zhang B: Exogenous IL-33 overcomes T cell tolerance in murine acute myeloid leukemia. Oncotarget. 7:61069–61080. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Bergis D, Kassis V, Ranglack A, Koeberle V, Piiper A, Kronenberger B, Zeuzem S, Waidmann O and Radeke HH: High serum levels of the interleukin-33 receptor soluble ST2 as a negative prognostic factor in hepatocellular carcinoma. Transl Oncol. 6:311–318. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2019
Volume 17 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Bai F, Ba F, You Y, Feng Y, Tao W, Wu C, Jiu M and Nie Y: Decreased ST2 expression is associated with gastric cancer progression and pathogenesis. Oncol Lett 17: 5761-5767, 2019
APA
Bai, F., Ba, F., You, Y., Feng, Y., Tao, W., Wu, C. ... Nie, Y. (2019). Decreased ST2 expression is associated with gastric cancer progression and pathogenesis. Oncology Letters, 17, 5761-5767. https://doi.org/10.3892/ol.2019.10223
MLA
Bai, F., Ba, F., You, Y., Feng, Y., Tao, W., Wu, C., Jiu, M., Nie, Y."Decreased ST2 expression is associated with gastric cancer progression and pathogenesis". Oncology Letters 17.6 (2019): 5761-5767.
Chicago
Bai, F., Ba, F., You, Y., Feng, Y., Tao, W., Wu, C., Jiu, M., Nie, Y."Decreased ST2 expression is associated with gastric cancer progression and pathogenesis". Oncology Letters 17, no. 6 (2019): 5761-5767. https://doi.org/10.3892/ol.2019.10223