Next Article in Journal
Moderating Effects of BDNF Genetic Variants and Smoking on Cognition in PTSD Veterans
Next Article in Special Issue
Oximes: Novel Therapeutics with Anticancer and Anti-Inflammatory Potential
Previous Article in Journal
Improving Gemcitabine Sensitivity in Pancreatic Cancer Cells by Restoring miRNA-217 Levels
Previous Article in Special Issue
Cation Transporters of Candida albicans—New Targets to Fight Candidiasis?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Immunohistochemical Analysis of the Expression of Adhesion Proteins: TNS1, TNS2 and TNS3 in Correlation with Clinicopathological Parameters in Gastric Cancer

by
Marcin Nizioł
1,
Justyna Zińczuk
2,
Konrad Zaręba
3,
Katarzyna Guzińska-Ustymowicz
1 and
Anna Pryczynicz
1,*
1
Department of General Pathomorphology, Medical University of Białystok, Kilińskiego 1, 15-089 Białystok, Poland
2
Department of Clinical Laboratory Diagnostics, Medical University of Białystok, Kilińskiego 1, 15-089 Białystok, Poland
3
2nd Clinical Department of General and Gastroenterological Surgery, Medical University of Białystok, Kilińskiego 1, 15-089 Białystok, Poland
*
Author to whom correspondence should be addressed.
Biomolecules 2021, 11(5), 640; https://doi.org/10.3390/biom11050640
Submission received: 26 February 2021 / Revised: 13 April 2021 / Accepted: 22 April 2021 / Published: 26 April 2021
(This article belongs to the Collection Feature Papers in Biochemistry)

Abstract

:
Tensins belong to the group of adhesion proteins that are involved in cell adhesion and migration, actin cytoskeleton maintenance and intercellular communication. TNS1, TNS2 and TNS3 proteins expression was evaluated in 90 patients with gastric cancer by immunohistochemistry method. TNS1 was more frequently present in non-differentiated tumors compared to poorly and moderately differentiated tumors (p = 0.016). TNS1 was also more often observed in metastatic tumors compared to those without distant metastases (p = 0.001). TNS2 was more common in moderately differentiated tumors than in poorly or non-differentiated ones (p = 0.041). TNS2 expression was also more frequently present in tumors with peritumoral inflammation (p = 0.041) and with concomitant H. pylori infection (p = 0.023). In contrast, TNS3 protein was more prevalent in moderately than in poorly and non-differentiated tumors (p = 0.023). No significant relationship was found between tensins’ expression and the overall survival rate of patients. TNS1 protein expression is associated with a poor-prognosis type of GC. Higher expression of TNS2 is accompanied by peritumoral inflammation and H. pylori infection, which favor the development of GC of a better prognosis, similarly to higher TNS3 protein expression.

1. Introduction

Annually, gastric cancer (GC) contributes to the death of nearly 6% of malignant cancer patients worldwide, which makes it the fifth cause of death caused by various cancers [1]. GC affects men more frequently than women and is more and more often diagnosed in young people (<40 years of age) with a history of this type of cancer in their family [2]. For early-stage GC, the five-year survival rate is approximately 90% [3]. It is therefore reasonable to conduct research aimed at identifying changes in the molecular and biochemical profile specific to the early stages of GC progression. In recent years, numerous studies have focused on adhesion proteins as anchor points in anti-cancer therapies. Adhesion proteins may also play an important role in early cancer detection. One of the protein families involved in tumorigenesis are tensins (TNSs). So far, four proteins (TNS1-4) have been recognized and classified as belonging to this family, based on structural similarity. Typical domains for tensins are SH2 (Src homology 2 domain) and PTB (phosphotyrosine-binding domain). A characteristic feature of tensins is the ability of their SH2 domain to interact with tyrosine residues of the kinases: PI3K (phosphoinositide 3-kinase) and FAK (focal adhesion kinase) [4,5] as well as p130Cas [6]. The PTB domain, in turn, can bind to the β1-, 3-, 5- and 7-integrin tails [7]. Furthermore, TNS1–3 has an ABD (actin-binding domain) at the N-terminal fragment. The presence of PTB and ABD domains enables binding to integrin receptors and actin filaments, thus allowing for the contact between the intracellular environment (cytoskeleton) and the extracellular matrix (ECM). Through these domains, TNS1-3 are able to influence many intracellular signaling pathways, e.g., EGFR, c-Met [8], Rho GAP DLC1 (deleted in liver cancer 1) [9], paxillin [10] and c-Cbl [11]. The importance of tensins 1-3 in cell adhesion and migration has been demonstrated through the presence of these proteins in podosomes [12] and invadopodia [13] that condition the phenomenon of epithelial-mesenchymal transition (EMT). The articles presented herein demonstrate the multitude of tensin-dependent processes, including cell adhesion, migration, maintenance of the actin cytoskeleton and intercellular communication [14].
Literature reports indicate that TNS1 expression is strongly present in the myocardium, kidneys, lungs, small and large intestine, ovaries and prostate [15], while no expression of this protein has been observed in the brain, thymus and leukocytes [16]. TNS1 expression is stimulated, for example, by angiotensin [17], oncogenes (v-Src, BCR-ABL) [18], PDGF [19] or thrombin [20], and is hindered by AMP-activated protein kinase (AMPK). At the cellular level, TNS1, in addition to participating in tensin-specific signaling pathways, is involved in apoptosis as a substrate of the active form of caspase-3 [7]. TNS2, unlike TNS1, interacts with Axl tyrosine residue [21]. The overexpression of this protein promotes apoptosis activation and limits cancer cell proliferation [22] by inhibiting the activity of Akt kinase [17]. TNS3 is structurally deficient in the ABD II (present in TNS1) and C1 (present in TNS2) domains [8]. TNS3 is known to be an adaptor protein in podosome formation by interacting with Dock5 [23]. It is overexpressed in cancer cells, which causes their enhanced invasiveness [24]. It has been observed that increased migration capacity of cancer cells is a result of phosphorylation of the SH2 domain by Src [25]. Silencing the expression of this protein inhibits the growth and migration of cancer cells [25]. It has also been demonstrated that TNS3 expression can be regulated by epigenetic mechanisms [26].
The subject of this study was the immunohistochemical evaluation of the expression of tensins 1-3 in gastric cancer (stages I-IV) as well as the assessment of the correlation of tensin expression with selected clinicopathological parameters and patients’ survival.

2. Materials and Methods

2.1. Study Group

The study was conducted on a group of 90 patients diagnosed with gastric cancer, who were surgically treated at the 2nd Clinical Department of General and Gastroenterological Surgery, Medical University of Bialystok in the years 2005–2015. Tissue material was obtained from the archives of the Academic Centre for Pathomorphological Diagnostics and Molecular Genetics in Bialystok. Patients were included in the study group based on a diagnosed adenocarcinoma at any stage. The exclusion criteria were: squamous cell carcinoma and other nonepithelial neoplasms, metastases of other neoplasms to the stomach and lack of complete medical documentation. Normal mucosa from the tumor environment was used as the control tissue. The study was approved by the Bioethics Committee of the Medical University of Bialystok, permission no.: R-I-002/29/2019. The study was conducted in accordance with the World Medical Association Declaration of Helsinki for ethical principles for medical research involving human subjects. The characteristics of the study group are presented in Table 1.

2.2. Tissue Preparations

Tissue sections taken during the surgery were fixed in 4% buffered formalin solution and embedded in paraffin. The paraffin blocks were then sliced with a microtome into approximately 4-µm-thick slides and stained with hematoxylin and eosin. Via a routine histopathological examination, we assessed the histological type of tumor, histopathological grade (G), stage (pT) and the presence of lymph node metastases (pN), blood and lymphatic vessel infiltration, perineural cancer cells infiltration, peritumoral inflammation and the degree of desmoplasia. Moreover, H. pylori infection was assessed in Giemsa-stained preparations. The following information was selected from the histopathological diagnosis sheets: age and gender of the patients, tumor diameter and location, presence of distant metastases and type of cancer according to the Lauren classification.

2.3. Immunohistochemistry

Immunohistochemical (IHC) staining was performed on 90 gastric cancer tissues using the polymer method. Paraffin blocks were cut with a microtome into approximately 4-µm-thick sections on silanized slides. The microscope slides were incubated overnight at 60 °C and then deparaffinized in xylene solutions and rehydrated in a series of alcohols of decreasing concentration (2 × 99.9%, 96%, 70%). Antigen retrieval was performed in citric buffer of pH = 6.0 in a water bath at 97.5 °C for 20 min, and then in room temperature for 20 min. Endogenous peroxidase was blocked by using 3% hydrogen peroxide for 10 min. Subsequently, non-specific antibody binding was blocked by using horse serum (anti-mouse/rabbit serum produced in Horse, Vector Laboratories, Eching, Germany) for 20 min. The sections were then incubated with polyclonal anti-TNS1 (clone HPA036089, Sigma-Aldrich, Stockholm, Sweden), anti-TNS2 (clone HPA034659, Sigma Aldrich, Stockholm, Sweden) and anti-TNS3 (clone HPA055338, Sigma-Aldrich, Stockholm, Sweden) polyclonal antibodies at dilutions of: 1:200, 1:100 and 1:250, respectively, for 30 min at room temperature. Antibody binding sites were visualized using the ImmPress Universal Antibody Polymer Reagent kit (Vector Laboratories, Eching, Germany) as well as ImmPACT DAB chromogen (Vector Laboratories, Eching, Germany). Cell nuclei were stained with hematoxylin. Next, the preparations were dehydrated in a series of alcohols of increasing concentration, and washed in xylene solutions.

2.4. Validation of TNS1, TNS2, TNS3 Expression Detection

We particularly focused on obtaining reliable results of immunohistochemical staining. In order to optimize the TNS1, TNS2 and TNS3 protein staining procedure, positive and negative controls were performed and selected dilutions of primary antibodies (1:50, 1:75, 1:100, 1:200, 1:250, 1:400), as well as selected incubation times (30 min, 60 min, 120 min) were tested. Antigen retrieval in pH = 6.0 and pH = 9.0 buffers was also performed during the controls.

2.5. Microscopic Evaluation

The slides were viewed and evaluated by two independent pathomorphologists on an Olympus BX41 light microscope. The expression of TNS1, TNS2 and TNS3 proteins was assessed at 100× magnification in 10 representative fields of view. In each of them, we evaluated ≥100 tumor cells. Protein expression was observed in both the cell membrane and cytoplasm. Cut-off points were statistically evaluated. The presence of TNS1 protein in >5% of tumor cells was considered positive expression; for TNS2, it was the presence in >10% of tumor cells, and for TNS3, in ≥20% of tumor cells.

2.6. Statistical Analysis

The comparison of protein expression between the study groups was performed using Student’s t-test. TNS1 expression was compared with the expression of TNS2 protein; TNS1 was compared with TNS3, and TNS2 with TNS3. The comparison of TNS1, TNS2 and TNS3 expressions with the selected clinicopathological parameters was performed by means of the Mann-Whitney U test for two groups, and the Kruskal-Wallis test for three and more groups. Additionally, for the Kruskal-Wallis test, the Dunn’s Multiple Comparison post hoc test was conducted. A value of p < 0.05 was considered statistically significant. The overall survival analysis was performed using the Kaplan–Meier estimate. For the analysis, the Statistica 13 program (Statsoft, Krakow, Poland) was used. Missing data were eliminated in pairs.

3. Results

3.1. Expression of TNS1, TNS2 and TNS3 in Gastric Cancer Samples

The expression of TNS1, TNS2 and TNS3 was examined immunohistochemically in 90 gastric cancer samples and 20 normal gastric tissues. In all cases of control group in normal gastric mucosa, expression of these proteins was absent. The expression of the tensins was higher in cancer cells compared to normal gastric mucosa cells. A microscopic analysis demonstrated that positive expression of TNS1, TNS2 and TNS3 proteins in tumor cells was present in 7 (7.78%), 4 (4.44%) and 32 (35.56%) out of 90 patients, respectively. In tumor cells of all cases, the expression of these proteins was observed in the cell membrane and cytoplasm (Figure 1A–D).

3.2. Comparison of TNS1, TNS2 and TNS3 Expression with Clinicopathological Parameters of GC

The statistical analysis demonstrated a significant correlation between TNS1 expression and the malignancy grade (Figure 2A). The study group consisted of 28.89% moderately differentiated, 38.89% poorly differentiated and 32.22% non-differentiated cancers. TNS1 protein was more frequently present in non-differentiated tumors (17.24% of patients) than in poorly differentiated (5.71% of patients) and moderately differentiated (0.00% of patients) cancers (p = 0.016). The statistical analysis also showed that TNS1 protein expression correlates with the presence of distant organ metastases (Figure 2B). In the study group, 68.89% of tumors did not metastasize to distant tissues and 31.11% of tumors showed such metastases. TNS1 expression was more frequently observed in metastatic tumors (21.43% of patients) compared to cancers without distant metastases (1.61% of patients) (p = 0.001). The results are presented in Table 2.
A correlation was demonstrated between TNS2 expression and the malignancy grade (Figure 2C). TNS2 was more frequently observed in moderately differentiated tumors (11.54% of patients) as compared to poorly differentiated (2.86% of patients) and non-differentiated tumors (0.00% of patients) (p = 0.041). There was also a statistically significant relationship between TNS2 expression and peritumoral inflammation (Figure 2D). In the study group, there were 50% of tumors without inflammation and the same number with peritumoral inflammation. TNS2 expression occurred more frequently in cancers with peritumoral inflammation (9.52% of patients) compared to tumors without inflammation in the surrounding tissues (0.00% of patients) (p = 0.041). TNS2 protein expression was also shown to correlate with H. pylori infection (Figure 2E). The study group comprised 73.81% of cancers without H. pylori infection and 26.19% of tumors accompanied by this infection. TNS2 expression was more frequently observed in tumors with H. pylori infection (13.64% of patients) than in cases without the infection (1.61% of patients) (p = 0.023). The results are presented in Table 3.
The statistical analysis revealed a correlation between TNS3 expression and the malignancy grade (Figure 2F). TNS3 protein was more frequently present in moderately differentiated tumors (53.85% of patients) than in poorly differentiated (31.43% of patients) and non-differentiated tumors (24.14% of patients) (p = 0.023). The results are presented in Table 4.

3.3. Analysis of the Correlation of TNS1, TNS2 and TNS3 Expression Levels with the Overall Survival of Patients

The statistical analysis showed no significant correlation between TNS1, TNS2 and TNS3 expressions and the overall survival of patients (p = 0.873, p = 0.599, p = 0.634, respectively) (Figure 3A–C).

4. Discussion

Gastric cancer belongs to common malignant tumors and is a cause of death of a high number of patients. Therefore, it poses a challenge to scientists studying its pathomechanisms and searching for specific and sensitive markers of tumor cell formation that would enable early detection of cancerous lesions. Another important goal of scientists and clinicians is the search for new therapeutic targets that would increase the effectiveness of anti-cancer therapies and improve the quality of life of GC patients. From the perspective of cancer progression, the key ability of cancer cells is to migrate and invade, thus leading to the formation of distant metastases. Proteins involved in actin cytoskeleton remodeling are factors that enhance the migratory potential of cancer cells. Of the numerous types of adhesion proteins, tensins are known to enable the interaction between the intracellular environment and extracellular matrix.
The aim of our study was to evaluate the immunohistochemical expression of proteins from the tensin family: TNS1-3 in patients diagnosed with stages I-IV of gastric cancer, and then to analyze the correlation between protein expression levels and selected clinicopathological parameters. In tumor cells of all cases, we observed both membranous and cytoplasmic expressions of TNS1, TNS2, and TNS3 proteins. No correlations between immunostaining intensity and the cellular location of these proteins were found in the literature. We noted that the positive expression of TNS1 and TNS2 was not common in tumor cells, whereas the positive expression of TNS3 occurred in tumor cells of about one-third of GC patients. The observed dependencies may result from differences in the expression pattern of TNS1-3. TNS1, similarly to TNS2, is expressed in the heart, kidneys, skeletal muscles and liver [15].
Our study demonstrated that the positive expression of TNS1-3 proteins correlated with the malignancy grade of GC. TNS1 expression was over three times more frequent in non-differentiated gastric tumors than in the poorly differentiated type, whereas no TNS1 was detected in moderately differentiated tumors. Although there are no reports in the literature indicating a role of TNS1 in the development of non-differentiated cancer, we hypothesized that this protein may be overexpressed in intensely growing tissues. TNS1 is known to be essential for the formation of fibrils in extracellular vesicles [27] and myofibroblast differentiation [28] during embryogenesis, while the absence of TNS1 leads, inter alia, to kidney degeneration [29]. In contrast, positive expression of TNS2 and TNS3 was observed more frequently in moderately differentiated gastric cancer than in poorly or undifferentiated tumors. It is known that the degree of differentiation of glandular ducts in the stomach enables to determine the disease progression. Higher GC cell differentiation is related with better patient’s prognosis [30]. According to the results of our research, the studied proteins may be involved in the process of differentiation of gastric cancer cells, although their role varies depending on the tensin type. The available literature provides no reports confirming a significant correlation between the expression level of these proteins and the degree of tumor malignancy, which underlines the uniqueness of our study. TNS1 expression is more common in non-differentiated GC associated with a worse prognosis.
According to our further observations, positive TNS1 expression is correlated with the presence of distant organ metastases, which supports the hypothesis that TNS1 may play an important role as a prognostic factor for GC. In primary prostate cancer, Zhu et al. [31] observed a similar relationship between TNS1 mRNA expression levels and tumor metastases to bones in which the expression of this gene in metastatic cells was significantly higher. Moreover, Martuszewska et al. [32] suggested that the presence of TNS3 at the cell periphery stabilizes the structure of the cells by limiting their motility, and thus reduces metastatic potential. An RNA-seq analysis revealed a significant involvement of TNS2 in the formation of focal adhesions which are associated with the development of metastases in numerous cancers [33]. Our findings did not show any relationships between TNS2 and TNS3 expression and metastases.
In our study, we observed statistically significant differences in TNS2 expression between cancers with and without a peritumoral inflammation. Moreover, we noted a significant correlation between this protein expression and the concomitant H. pylori infection. The tumor microenvironment is diverse in terms of cell types. Tumor cells are often accompanied by inflammatory cells whose role in tumorigenesis remains unclear: they may have either pro- or anti-cancer effects [34]. An immune response should lead to the recognition and destruction of cancer cell clones; however, in many tumors, we observed disturbances in the functioning of the immune system. This is probably due to the defense mechanisms of the developing tumor, i.e., the presence of poorly immunogenic antigens, as well as disturbances in antigen presentation by inflammatory cells, or production of immunosuppressive factors by cancer cells [35]. It is probable that the observed positive TNS2 expression with simultaneous peritumoral inflammation is related to H. pylori infection. Numerous malignancies are known to develop at sites of infection and ongoing inflammation [34]. Since we observed a correlation between higher TNS2 expression in moderately differentiated GC with peritumoral inflammation and H. pylori infection, further studies are required to confirm the existence of such a relationship.
What is more, we did not demonstrate a statistically significant correlation between TNS1-3 expression levels and the survival of patients. However, this absence of statistical significance may result from the low number of subjects in our study group as well as the lack of available survival data in the case of some patients. These limitations of the study prevented us from drawing conclusions about the relationship of TNS1-3 with the survival rate of gastric cancer patients. Zhan et al. [36] observed that TNS1 overexpression correlates with prolonged metastasis-free survival, whereas in metastatic breast cancer, the expression of this protein is significantly reduced. In contrast, Zhang et al. [37] demonstrated that TNS1 expression levels and the overall survival of bladder cancer patients were negatively correlated. According to other studies, reduced TNS2 expression is positively correlated with short relapse-free survival of breast and lung cancer patients [38]. It was also shown that patients with kidney cancer and TNS3 detected in their cell membrane prognosed better survival than those lacking TNS3 or with TNS3 present only in the cytoplasm [32].

5. Conclusions

In conclusion, our study suggests that TNS1 expression is associated with a GC type of a poorer prognosis and with the occurrence of distant metastases. In contrast, higher TNS2 expression is accompanied by peritumoral inflammation as well as H. pylori infection, which favor GC with a better prognosis, as does higher expression of TNS3 protein.

Author Contributions

Conceptualization, M.N. and A.P.; methodology, M.N., J.Z., K.Z., K.G.-U. and A.P.; formal analysis, M.N. and A.P.; investigation, M.N., J.Z., K.Z., K.G.-U. and A.P.; resources, M.N.; data curation, M.N. and A.P.; writing—original draft preparation, M.N.; writing—review and editing, A.P.; visualization, M.N.; supervision, A.P.; project administration, M.N.; funding acquisition, M.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Medical University of Bialystok; grant number SUB/1/DN/20/002/3314.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved Bioethics Committee of the Medical University of Bialystok (R-I-002/29/2019, date of approval: 31 January 2019).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Data supporting reported results can be obtained from the corresponding author upon request.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
  2. Rona, K.A.; Schwameis, K.; Zehetner, J.; Samakar, K.; Green, K.; Samaan, J.; Sandhu, K.; Bildzukewicz, N.; Katkhouda, N.; Lipham, J.C. Gastric cancer in the young: An advanced disease with poor prognostic features. J. Surg. Oncol. 2017, 115, 371–375. [Google Scholar] [CrossRef]
  3. Song, Z.; Wu, Y.; Yang, J.; Yang, D.; Fang, X. Progress in the treatment of advanced gastric cancer. Tumour Biol. 2017, 39, 1010428317714626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Hall, E.H.; Balsbaugh, J.L.; Rose, K.L.; Shabanowitz, J.; Hunt, D.F.; Brautigan, D.L. Comprehensive analysis of phosphorylation sites in Tensin1 reveals regulation by p38MAPK. Mol. Cell Proteom. 2010, 9, 2853–2863. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Cui, Y.; Liao, Y.C.; Lo, S.H. Epidermal growth factor modulates tyrosine phosphorylation of a novel tensin family member, tensin3. Mol. Cancer Res. 2004, 2, 225–232. [Google Scholar]
  6. Zhao, Z.; Tan, S.H.; Machiyama, H.; Kawauchi, K.; Araki, K.; Hirata, H.; Sawada, Y. Association between tensin 1 and p130Cas at focal adhesions links actin inward flux to cell migration. Biol. Open 2016, 5, 499–506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Lo, S.H. Tensin. Int. J. Biochem. Cell Biol. 2004, 36, 31–34. [Google Scholar] [CrossRef]
  8. Lo, S.H. Tensins. Curr. Biol. 2017, 27, R331–R332. [Google Scholar] [CrossRef] [PubMed]
  9. Georgiadou, M.; Ivaska, J. Tensins: Bridging AMP-Activated Protein Kinase with Integrin Activation. Trends Cell Biol. 2017, 27, 703–711. [Google Scholar] [CrossRef]
  10. Schaller, M.D. Paxillin: A focal adhesion-associated adaptor protein. Oncogene 2001, 20, 6459–6472. [Google Scholar] [CrossRef] [Green Version]
  11. Hong, S.Y.; Shih, Y.P.; Li, T.; Carraway, K.L.; Lo, S.H. CTEN prolongs signaling by EGFR through reducing its ligand-induced degradation. Cancer Res. 2013, 73, 5266–5276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Hiura, K.; Lim, S.S.; Little, S.P.; Lin, S.; Sato, M. Differentiation dependent expression of tensin and cortactin in chicken osteoclasts. Cell Motil. Cytoskelet. 1995, 30, 272–284. [Google Scholar] [CrossRef]
  13. Mueller, S.C.; Yeh, Y.; Chen, W.T. Tyrosine phosphorylation of membrane proteins mediates cellular invasion by transformed cells. J. Cell Biol. 1992, 119, 1309–1325. [Google Scholar] [CrossRef] [PubMed]
  14. Nizioł, M.; Pryczynicz, A. The role of tensins in malignant neoplasms. Arch. Med Sci. 2021. [Google Scholar] [CrossRef]
  15. Chen, H.; Ishii, A.; Wong, W.K.; Chen, L.B.; Lo, S.H. Molecular characterization of human tensin. Biochem. J. 2000, 351 Pt 2, 403–411. [Google Scholar] [CrossRef]
  16. Lo, S.H.; Janmey, P.A.; Hartwig, J.H.; Chen, L.B. Interactions of tensin with actin and identification of its three distinct actin-binding domains. J. Cell Biol. 1994, 125, 1067–1075. [Google Scholar] [CrossRef]
  17. Haynie, D.T. Molecular physiology of the tensin brotherhood of integrin adaptor proteins. Proteins 2014, 82, 1113–1127. [Google Scholar] [CrossRef] [PubMed]
  18. Salgia, R.; Brunkhorst, B.; Pisick, E.; Li, J.L.; Lo, S.H.; Chen, L.B.; Griffin, J.D. Increased tyrosine phosphorylation of focal adhesion proteins in myeloid cell lines expressing p210BCR/ABL. Oncogene 1995, 11, 1149–1155. [Google Scholar] [PubMed]
  19. Jiang, B.; Yamamura, S.; Nelson, P.R.; Mureebe, L.; Kent, K.C. Differential effects of platelet-derived growth factor isotypes on human smooth muscle cell proliferation and migration are mediated by distinct signaling pathways. Surgery 1996, 120, 427–431, discussion 432. [Google Scholar] [CrossRef]
  20. Ishida, T.; Ishida, M.; Suero, J.; Takahashi, M.; Berk, B.C. Agonist-stimulated cytoskeletal reorganization and signal transduction at focal adhesions in vascular smooth muscle cells require c-Src. J. Clin. Investig. 1999, 103, 789–797. [Google Scholar] [CrossRef] [Green Version]
  21. Cheng, L.C.; Chen, Y.L.; Cheng, A.N.; Lee, A.Y.; Cho, C.Y.; Huang, J.S.; Chuang, S.E. AXL phosphorylates and up-regulates TNS2 and its implications in IRS-1-associated metabolism in cancer cells. J. Biomed. Sci. 2018, 25, 80. [Google Scholar] [CrossRef]
  22. Legate, K.R.; Montañez, E.; Kudlacek, O.; Fässler, R. ILK, PINCH and parvin: The tIPP of integrin signalling. Nat. Rev. Mol. Cell Biol. 2006, 7, 20–31. [Google Scholar] [CrossRef]
  23. Touaitahuata, H.; Morel, A.; Urbach, S.; Mateos-Langerak, J.; de Rossi, S.; Blangy, A. Tensin 3 is a new partner of Dock5 that controls osteoclast podosome organization and activity. J. Cell Sci. 2016, 129, 3449–3461. [Google Scholar] [CrossRef] [Green Version]
  24. Shinchi, Y.; Hieda, M.; Nishioka, Y.; Matsumoto, A.; Yokoyama, Y.; Kimura, H.; Matsuura, S.; Matsuura, N. SUV420H2 suppresses breast cancer cell invasion through down regulation of the SH2 domain-containing focal adhesion protein tensin-3. Exp. Cell Res. 2015, 334, 90–99. [Google Scholar] [CrossRef]
  25. Qian, X.; Li, G.; Vass, W.C.; Papageorge, A.; Walker, R.C.; Asnaghi, L.; Steinbach, P.J.; Tosato, G.; Hunter, K.; Lowy, D.R. The Tensin-3 protein, including its SH2 domain, is phosphorylated by Src and contributes to tumorigenesis and metastasis. Cancer Cell 2009, 16, 246–258. [Google Scholar] [CrossRef] [Green Version]
  26. Carter, J.A.; Górecki, D.C.; Mein, C.A.; Ljungberg, B.; Hafizi, S. CpG dinucleotide-specific hypermethylation of the TNS3 gene promoter in human renal cell carcinoma. Epigenetics 2013, 8, 739–747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Shinde, A.; Paez, J.S.; Libring, S.; Hopkins, K.; Solorio, L.; Wendt, M.K. Transglutaminase-2 facilitates extracellular vesicle-mediated establishment of the metastatic niche. Oncogenesis 2020, 9, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Bernau, K.; Torr, E.E.; Evans, M.D.; Aoki, J.K.; Ngam, C.R.; Sandbo, N. Tensin 1 Is Essential for Myofibroblast Differentiation and Extracellular Matrix Formation. Am. J. Respir. Cell Mol. Biol. 2017, 56, 465–476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Lo, S.H.; Yu, Q.C.; Degenstein, L.; Chen, L.B.; Fuchs, E. Progressive kidney degeneration in mice lacking tensin. J. Cell Biol. 1997, 136, 1349–1361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Adachi, Y.; Yasuda, K.; Inomata, M.; Sato, K.; Shiraishi, N.; Kitano, S. Pathology and prognosis of gastric carcinoma: Well versus poorly differentiated type. Cancer 2000, 89, 1418–1424. [Google Scholar] [CrossRef]
  31. Zhu, Z.; Wen, Y.; Xuan, C.; Chen, Q.; Xiang, Q.; Wang, J.; Liu, Y.; Luo, L.; Zhao, S.; Deng, Y.; et al. Identifying the key genes and microRNAs in prostate cancer bone metastasis by bioinformatics analysis. FEBS Open Bio 2020, 10, 674–688. [Google Scholar] [CrossRef]
  32. Martuszewska, D.; Ljungberg, B.; Johansson, M.; Landberg, G.; Oslakovic, C.; Dahlbäck, B.; Hafizi, S. Tensin3 is a negative regulator of cell migration and all four Tensin family members are downregulated in human kidney cancer. PLoS ONE 2009, 4, e4350. [Google Scholar] [CrossRef]
  33. Yang, X.; Wang, G.; Huang, X.; Cheng, M.; Han, Y. RNA-seq reveals the diverse effects of substrate stiffness on epidermal ovarian cancer cells. Aging 2020, 12, 20493–20511. [Google Scholar] [CrossRef]
  34. Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef]
  35. Whiteside, T.L. Immune suppression in cancer: Effects on immune cells, mechanisms and future therapeutic intervention. Semin. Cancer Biol. 2006, 16, 3–15. [Google Scholar] [CrossRef] [PubMed]
  36. Zhan, Y.; Liang, X.; Li, L.; Wang, B.; Ding, F.; Li, Y.; Wang, X.; Zhan, Q.; Liu, Z. MicroRNA-548j functions as a metastasis promoter in human breast cancer by targeting Tensin1. Mol. Oncol. 2016, 10, 838–849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Zhang, P.B.; Huang, Z.L.; Xu, Y.H.; Huang, J.; Huang, X.Y. Systematic analysis of gene expression profiles reveals prognostic stratification and underlying mechanisms for muscle-invasive bladder cancer. Cancer Cell Int. 2019, 19, 337. [Google Scholar] [CrossRef] [PubMed]
  38. Hong, S.Y.; Shih, Y.P.; Sun, P.; Hsieh, W.J.; Lin, W.C.; Lo, S.H. Down-regulation of tensin2 enhances tumorigenicity and is associated with a variety of cancers. Oncotarget 2016, 7, 38143–38153. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Tensin (TNS) protein expression levels in gastric cancer (GC): (A) TNS1 in non-differentiated GC, magnification, ×200; (B) TNS2 in moderately differentiated GC, magnification, ×200; (C) TNS3 in moderately differentiated GC, magnification, ×200; (D) comparison of TNS1-3 in GC. Statistical analysis performed using Student’s t-test.
Figure 1. Tensin (TNS) protein expression levels in gastric cancer (GC): (A) TNS1 in non-differentiated GC, magnification, ×200; (B) TNS2 in moderately differentiated GC, magnification, ×200; (C) TNS3 in moderately differentiated GC, magnification, ×200; (D) comparison of TNS1-3 in GC. Statistical analysis performed using Student’s t-test.
Biomolecules 11 00640 g001
Figure 2. Comparison of TNS1-3 expression levels with the selected clinicopathological parameters: (A) TNS1 and malignancy grade, G2 vs. G4 statistically significant at p = 0.02; (B) TNS1 and distant metastasis; (C) TNS2 and malignancy grade, G2 vs. G4 statistically significant at p = 0.045; (D) TNS2 and peritumoral inflammation; (E) TNS2 and H. pylori infection; (F) TNS3 and malignancy grade, G2 vs. G4 statistically significant at p = 0.025. The comparison of TNS1-3 expressions with the selected clinicopathological parameters was performed using the Mann-Whitney U test for two groups, and the Kruskal-Wallis test for 3 and more groups. Dunn’s Multiple Comparison post hoc test was applied for Kruskal-Wallis test. A value of p < 0.05 was considered statistically significant. a, b, c p < 0.05 G2 vs. G4 TNS1-3 expression levels in gastric cancer. G2—moderately differentiated GC, G3—poorly differentiated GC, G4—non-differentiated GC.
Figure 2. Comparison of TNS1-3 expression levels with the selected clinicopathological parameters: (A) TNS1 and malignancy grade, G2 vs. G4 statistically significant at p = 0.02; (B) TNS1 and distant metastasis; (C) TNS2 and malignancy grade, G2 vs. G4 statistically significant at p = 0.045; (D) TNS2 and peritumoral inflammation; (E) TNS2 and H. pylori infection; (F) TNS3 and malignancy grade, G2 vs. G4 statistically significant at p = 0.025. The comparison of TNS1-3 expressions with the selected clinicopathological parameters was performed using the Mann-Whitney U test for two groups, and the Kruskal-Wallis test for 3 and more groups. Dunn’s Multiple Comparison post hoc test was applied for Kruskal-Wallis test. A value of p < 0.05 was considered statistically significant. a, b, c p < 0.05 G2 vs. G4 TNS1-3 expression levels in gastric cancer. G2—moderately differentiated GC, G3—poorly differentiated GC, G4—non-differentiated GC.
Biomolecules 11 00640 g002
Figure 3. Total survival analysis performed using a Kaplan-Meier test of tensins in gastric cancer (A) TNS1; (B) TNS2; (C) TNS3.
Figure 3. Total survival analysis performed using a Kaplan-Meier test of tensins in gastric cancer (A) TNS1; (B) TNS2; (C) TNS3.
Biomolecules 11 00640 g003
Table 1. The characteristics of study group.
Table 1. The characteristics of study group.
ParameterNumber of Cases (%)
Age
 <6029 (32.22%)
 ≥6061 (67.78%)
Gender
 Female30 (33.33%)
 Male60 (66.67%)
Tumor diameter
 <5 cm19 (21.11%)
 ≥5 cm71 (78.89%)
Tumor localization
 Upper 1/317 (18.89%)
 Middle1/334 (37.78%)
 Lower 1/316 (17.78%)
 Whole stomach23 (25.55%)
Histological type
 Adenocarcinoma55 (61.11%)
 Adenocarcinoma mucinosum35 (38.89%)
Histological differentiation
 Moderately differentiated26 (28.89%)
 Poorly differentiated35 (38.89%)
 Non-differentiated29 (32.22%)
Depth of invasion
 T17 (7.78%)
 T27 (7.78%)
 T366 (73.33%)
 T410 (11.11%)
Lymph node metastasis
 Absent18 (20.00%)
 Present72 (80.00%)
Distant metastasis
 Absent62 (68.89%)
 Present28 (31.11%)
Blood vessel infiltration
 Absent49 (84.48%)
 Present9 (15.52%)
Lymphatic vessel infiltration
 Absent22 (32.84%)
 Present45 (61.16%)
Perineural cancer cells infiltration
 Absent29 (34.94%)
 Present54 (65.06%)
Peritumoral inflammation
 Absent42 (50.00%)
 Present42 (50.00%)
Desmoplasia
 Small56 (66.67%)
 Diffuse28 (33.33%)
H. pylori infection
 Absent62 (73.81%)
 Present22 (26.19%)
Lauren’s classification
 Intestinal47 (56.63%)
 Diffuse36 (43.37%)
Table 2. Correlation between TNS1 protein expression level in gastric cancer and clinicopathological parameters.
Table 2. Correlation between TNS1 protein expression level in gastric cancer and clinicopathological parameters.
ParameterExpression of TNS1 Proteinp-Value a
NegativePositive
Age 0.296
 <6028 (96.55%)1 (3.45%)
 ≥6055 (90.16%)6 (9.84%)
Gender 0.271
 Female29 (96.97%)1 (3.33%)
 Male54 (90.00%)6 (10.00%)
Tumor diameter 0.158
 <5cm19 (100%)0 (0.00%)
 ≥5cm64 (90.14%)7 (9.86%)
Tumor localization 0.201
 Upper 1/316 (94.12%)1 (5.88%)
 Middle1/332 (94.12%)2 (5.88%)
 Lower 1/316 (100%)0 (0.00%)
 Whole stomach19 (82.61%)4 (17.39%)
Histological type 0.067
 Adenocarcinoma53 (96.36%)2 (3.64%)
 Adenocarcinoma mucinosum30 (85.71%)5 (14.29%)
Histological differentiation 0.02
 Moderately differentiated26 (100.00%)0 (0.00%)(0.016)
 Poorly differentiated33 (94.29%)2 (5.71%)
 Non-differentiated24 (82.76%)5 (17.24%)
Depth of invasion 0.300
 T17 (100%)0 (0.00%)
 T27 (100%)0 (0.00%)
 T360 (90.91%)6 (9.09%)
 T49 (90.00%)1 (10.00%)
Lymph node metastasis 0.172
 Absent18 (100%)0 (0.00%)
 Present65 (90.28%)7 (9.72%)
Distant metastasis 0.001
 Absent61 (98.39%)1 (1.61%)
 Present22 (78.57%)6 (21.43%)
Blood vessel infiltration 0.546
 Absent47 (95.92%)2 (4.08%)
 Present9 (100%)0 (0.00%)
Lymphatic vessel infiltration 0.107
 Absent22 (100%)0 (0.00%)
 Present40 (88.89%)5 (11.11%)
Perineural cancer cells infiltration 0.064
 Absent29 (100.00%)0 (0.00%)
 Present48 (88.89%)6 (11.11%)
Peritumoral inflammation 0.403
 Absent38 (90.48%)4 (9.52%)
 Present40 (95.24%)2 (4.76%)
Desmoplasia 1.000
 Small52 (92.86%)4 (7.14%)
 Large26 (92.86%)2 (7.14%)
H. pylori infection 0.133
 Absent56 (90.32%)6 (9.68%)
 Present22 (100.00%)0 (0.00%)
Lauren’s classification 0.260
 Intestinal43 (95.56%)4 (4.44%)
 Diffuse32 (88.89%)4 (11.11%)
a in brackets are p-values before Dunn’s Multiple Comparison post hoc test.
Table 3. Correlation between TNS2 protein expression level in gastric cancer and clinicopathological parameters.
Table 3. Correlation between TNS2 protein expression level in gastric cancer and clinicopathological parameters.
ParameterExpression of TNS2 Proteinp-Value a
NegativePositive
Age 0.755
 <6028 (96.55%)1 (3.45%)
 ≥6058 (95.08%)3 (4.92%)
Gender 0.475
 Female28 (93.33%)2 (6.67%)
 Male58 (96.67%)2 (3.33%)
Tumor diameter 0.848
 <5 cm18 (94.74%)1 (5.26%)
 ≥5 cm68 (95.77%)3 (4.23%)
Tumor localization 0.636
 Upper 1/317 (100.00%)0 (0.00%)
 Middle1/332 (94.12%)2 (5.88%)
 Lower 1/315 (93.75%)1 (6.25%)
 Whole stomach22 (95.65%)1 (4.35%)
Histological type 0.105
 Adenocarcinoma51 (92.73%)4 (7.27%)
 Adenocarcinoma mucinosum35 (100.00%)0 (0.00%)
Histological differentiation 0.045(0.041)
 Moderately differentiated23 (88.46%)3 (11.54%)
 Poorly differentiated34 (97.14%)1 (2.86%)
 Non-differentiated29 (100.00%)0 (0.00%)
Depth of invasion 0.272
 T16 (85.71%)1 (14.29%)
 T27 (100%)0 (0.00%)
 T363 (95.45%)3 (4.55%)
 T410 (100.00%) 0 (0.00%)
Lymph node metastasis 0.801
 Absent17 (94.44%)1 (5.56%)
 Present69 (95.83%)3 (4.17%)
Distant metastasis 0.173
 Absent58 (93.55%)4 (6.45%)
 Present28 (100%)0 (0.00%)
Blood vessel infiltration 0.546
 Absent47 (95.92%)2 (4.08%)
 Present9 (100%)0 (0.00%)
Lymphatic vessel infiltration 0.208
 Absent20 (90.91%)2 (9.09%)
 Present44 (97.78%)1 (2.22%)
Perineural cancer cells infiltration 0.087
 Absent26 (89.66%)3 (10.34%)
 Present53 (98.15%)1 (1.851%)
Peritumoral inflammation 0.041
 Absent42 (100.00%)0 (0.00%)
 Present38 (90.48%)4 (9.52%)
Desmoplasia 0.721
 Small53 (94.64%)3 (5.36%)
 Large27 (96.43%)1 (3.57%)
H. pylori infection 0.023
 Absent61 (98.39%)1 (1.61%)
 Present19 (86.36%)3 (13.64%)
Lauren’s classification 0.697
 Intestinal43 (95.56%)2 (4.44%)
 Diffuse35 (97.22%)1 (2.77%)
a in brackets are p-values before Dunn’s Multiple Comparison post hoc test.
Table 4. Correlation between TNS3 protein expression level in gastric cancer and clinicopathological parameters.
Table 4. Correlation between TNS3 protein expression level in gastric cancer and clinicopathological parameters.
ParameterExpression of TNS3 Proteinp-Value a
NegativePositive
Age 0.885
 <6019 (65.52%)10 (34.48%)
 ≥6039 (63.93%)22 (36.07%)
Gender 0.878
 Female19 (63.33%)11 (36.67%)
 Male39 (65.00%)21 (35.00%)
Tumor diameter 0.897
 <5 cm12 (63.16%)7 (36.84%)
 ≥5 cm46 (64.79%)25 (35.21%)
Tumor localization 0.308
 Upper 1/313 (76.47%)4 (23.53%)
 Middle 1/322 (64.71%)12 (35.29%)
 Lower 1/39 (56.25%)7 (43.75%)
 Whole stomach14 (60.87%)9 (39.13%)
Histological type 0.520
 Adenocarcinoma34 (61.82%)21 (38.18%)
 Adenocarcinoma mucinosum24 (68.57%)11 (31.43%)
Histological differentiation 0.025
 Moderately differentiated12 (46.15%)14 (53.85%)(0.023)
 Poorly differentiated24 (68.57%)11 (31.47%)
 Non-differentiated22 (75.86%)7 (24.14%)
Depth of invasion 0.514
 T15 (71.43%)2 (28.57%)
 T23 (42.86%)4 (57.14%)
 T342 (63.64%)24 (36.36%)
 T48 (80.00%) 2 (20.00%)
Lymph node metastasis 0.447
 Present45 (62.50%)5 (27.78%)
 Absent13 (72.22%)27 (37.50%)
Distant metastasis 0.654
 Absent39 (62.90%)23 (37.10%)
 Present19 (67.86%)9 (32.14%)
Blood vessel infiltration 0.584
 Absent32 (65.31%)17 (34.69%)
 Present5 (55.56%)4 (44.44%)
Lymphatic vessel infiltration 0.912
 Absent14 (63.64%)8 (36.36%)
 Present28 (62.22%)17 (37.78%)
Perineural cancer cells infiltration 0.473
 Absent17 (58.62%)12 (41.38%)
 Present36 (66.67%)18 (33.33%)
Peritumoral inflammation 0.824
 Absent27 (64.29%)15 (35.71%)
 Present26 (61.90%)16 (38.10%)
Desmoplasia 0.268
 Small33 (58.93%)23 (41.07%)
 Large20 (71.43%)8 (28.57%)
H. pylori infection 0.952
 Absent39 (62.90%)23 (37.10%)
 Present14 (63.64%)8 (36.36%)
Lauren’s classification 0.420
 Intestinal26 (57.78%)19 (42.22%)
 Diffuse24 (66.67%)12 (33.33%)
a in brackets are p-values before Dunn’s Multiple Comparison post hoc test.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Nizioł, M.; Zińczuk, J.; Zaręba, K.; Guzińska-Ustymowicz, K.; Pryczynicz, A. Immunohistochemical Analysis of the Expression of Adhesion Proteins: TNS1, TNS2 and TNS3 in Correlation with Clinicopathological Parameters in Gastric Cancer. Biomolecules 2021, 11, 640. https://doi.org/10.3390/biom11050640

AMA Style

Nizioł M, Zińczuk J, Zaręba K, Guzińska-Ustymowicz K, Pryczynicz A. Immunohistochemical Analysis of the Expression of Adhesion Proteins: TNS1, TNS2 and TNS3 in Correlation with Clinicopathological Parameters in Gastric Cancer. Biomolecules. 2021; 11(5):640. https://doi.org/10.3390/biom11050640

Chicago/Turabian Style

Nizioł, Marcin, Justyna Zińczuk, Konrad Zaręba, Katarzyna Guzińska-Ustymowicz, and Anna Pryczynicz. 2021. "Immunohistochemical Analysis of the Expression of Adhesion Proteins: TNS1, TNS2 and TNS3 in Correlation with Clinicopathological Parameters in Gastric Cancer" Biomolecules 11, no. 5: 640. https://doi.org/10.3390/biom11050640

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop