Transglutaminase 3 protein modulates human esophageal cancer cell growth by targeting the NF-κB signaling pathway

  • Authors:
    • Wei Li
    • Zhongmian Zhang
    • Wenchao Zhao
    • Na Han
  • View Affiliations

  • Published online on: July 8, 2016     https://doi.org/10.3892/or.2016.4921
  • Pages: 1723-1730
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

It has recently been found that the expression of the transglutaminase 3 (TGM3) is significantly reduced in esophageal cancer (EC). However, the potential of TGM3 as a useful biomarker or as a molecular target for EC diagnosis and treatment is unclear. The aim of the present study was to explore the role of the TGM3 in EC. The expression level of TGM3 were measured by real-time polymerase chain reaction and western blot analysis in EC cell lines, including SKGT-4, KYSE-510, OE33, OE21 and the normal esophageal epithelial cell line HEEC; fifty-eight pairs of tissues samples were also measured. through exogenous expression of TGM3 in EC cells it was found that the expression of TGM3 is closely associated with tumor cell growth and apoptosis. TGM3 was found downregulated in EC and closely associated with tumor proliferation and migration. In addition, overexpression of TGM3 apparently induces EC cell proliferation, migration, invasion and promotes cell apoptosis in vitro. Subsequent experiments identified the NF-κB signaling pathway as direct target of TGM3. Our data collectively demonstrate that TGM3 can be a candidate tumor suppressor that is able to induce EC cell proliferation and migration by downregulating the NF-κB signaling pathway, indicating that TGM3 may serve as a useful biomarker and therapeutic target for EC treatment.

Introduction

Esophageal cancer (EC) is the seventh most common malignant tumor in the world, the mortality of which is ranked sixth among the malignant tumors worldwide (1); its incidence has increased significantly in recent years (2), especially in China. Despite the clinical advances in diagnosis and treatment improvement for malignant tumors, EC remains one of the leading causes of cancer-associated mortality. The overall 5-year survival rate for all patients with EC is <20% (3). Owing to its aggressive nature and poor response to chemotherapy, EC cure is still a problem (4). The incidence and treatment rates of EC will continue to increase substantially with the development of an aging population and the social economic development in China. Therefore, research to identify and develop more effective molecular biomarkers to prevent or treat EC is urgently needed. Further studies have reported that transglutaminase 3 (TGM3) is significantly downregulated in ECs (58).

The transglutaminase 3 (TGM 3) enzyme is an enzyme with the ability to catalyze the irreversible cross-linking of peptide-bound glutamine residues either with peptide-bound lysines or with primary amines (5). It is encoded by the TGM3 gene and widely expressed in the small intestine, brain, skin and mucosa (6). In the skin and mucosa, TGM3 is predominantly expressed in the suprabasal layers of the stratified squamous epithelium (7,8). It has been demonstrated that TGM3 is required for the cross-linking of the structural protein trichohyalin and the keratin intermediate filaments to form a rigid structure within the inner root sheath cells (9,10). Recent studies have revealed that downregulation of the TGM3 gene is closely linked with a variety of human cancer types, including laryngeal carcinoma, esophageal and oral squamous cell carcinoma (OSCC) (1113). Moreover, Uemura et al (12) identified TGM3 as a novel prognostic indicator in ESCC; the prognostic performance of TGM3 was confirmed by immunohistochemistry in 76 ESCC cases. In addition, Mendez et al (14) reported that the TGM3 gene is differentially expressed in node-positive and node-negative primary tumors in patients with OSCC, implying that decreased TGM3 expression might contribute to the metastatic potential of OSCC. However, the biological function and molecular mechanism of the TGM3 gene in cancer initiation and progression have not been reported. In addition, whether the TGM3 gene might be a valuable diagnostic or therapeutic biomarker for cancer, especially for EC, needs to be further investigated.

The nuclear factor kappa B (NF-κB) transcription factor family is composed of p50, p52, RelA/p65, c-rel and Rel B. The homodimers and heterodimers of these molecules are sequestered in the cytoplasm in an inactive form by the inhibitor of kappa B (IκB). Upon stimulation, the IκB kinase complex (IKK) phosphorylates the κB inhibitor, which then releases NF-κB and allows its phosphorylation, nuclear translocation, binding and subsequent activation of target genes involved in the regulation of cell proliferation, survival, angiogenesis and metastasis (15). Constitutively active NF-κB is common in human cancer cell lines as well as tumor tissues derived from patients, but is rare in normal cells (16). In some types of human cancer, there is strong evidence of NF-κB being involved in cancer progression (17), thus, making NF-κB and its downstream signals promising targets for therapeutic intervention. However, the role of the NF-κB signaling pathway in the tumorigenesis of human EC is not fully understood.

In the present study, we confirmed that the expression levels of TGM3 are downregulated in EC cell lines compared with normal primary esophageal epithelial cells, and tissue specimens compared with paired adjacent normal tissues, by means of RT-PCR and western blotting. We further evaluated the effect of ectopic TGM3 expression in the SKGT-4, KYSE-510, OE33 and OE21 cell lines. We provide the first evidence that the ectopic expression of TGM3 in EC cell lines inhibits cell proliferation and migration and induces apoptosis in cancer cells in vitro. In addition, we demonstrate that the NF-κB signaling pathway is a direct target of TGM3 and show that TGM3 functions as an oncogene by activating the NF-κB signaling pathway. These results suggest that TGM3 might be a candidate tumor suppressor contributing to EC and could act as a valuable prognostic predictor for patients with EC.

Materials and methods

Tissue samples

Fifty-eight pairs of primary EC tissue samples and adjacent non-cancerous tissues (located >3 cm away from the tumor) were obtained from the Department of Oncology, The Second Affiliated Hospital, Zhengzhou University (Zhengzhou, China) between June 2014 and December 2015. All subjects were diagnosed and confirmed by a pathological evaluation. None of the subjects received any biotherapy or chemotherapy treatment before recruitment to this study. The study was approved by the Institutional Ethics Committee of the Second Affiliated Hospital, Zhengzhou University and all of the patients provided written informed consent in accordance with the institutional guidelines.

Cell culture and transfection

The EC cell lines SKGT-4 and KYSE-510 were purchased from the American Type Culture Collection (ATCC; Manassas, VA, USA). OE33 and OE21 were obtained from the European Collection of Animal Cell Cultures (ECACC; Porton Down, Salisbury, UK). All EC cell lines were grown in RPMI-1640 medium supplemented with 10% filtered fetal calf serum, 100 units/ml penicillin, 100 μg/ml streptomycin and 2 mM L-glutamine. In addition, the normal human esophageal epithelial cell line HEEC was grown in Dulbecco's modified Eagle's medium (DMEM) with 10% FBS. All cells were cultured in a humidified atmosphere of 5% CO2 at 37°C.

Bay11-7082, a specific inhibitor of NF-κB signaling, was purchased from CsA (Biomol, Plymouth Meeting, PA) and diluted in culture medium to obtain the desired concentration. Recombinant human TGM3 was obtained from Abcam Inc. (Cambridge, MA, USA), and the rabbit anti-TGM3 antibody was from Adipo Bioscience Inc. (Santa Clara, CA, USA). EC cells were transfected with recombinant human TGM3 (10 μg) or TGM3 antibody (1:400; Neomarkers, Fremond, CA, USA) with Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA) according to the manufacturer's instructions; EC cells without any treatment were used as controls.

RNA isolation and quantitative real-time PCR (qRT-PCR)

Total RNA was isolated using the TRIzol reagent (Invitrogen Life Technologies). The concentration of RNA were determined using a NanoDrop ND-1000 instrument (Thermo Fisher Scientific, Waltham, MA, USA), and aliquots of the samples were stored at −80°C. For reverse transcription, cDNA was synthesized by using a reverse transcription kit (Promega, Madison, WI, USA). Quantitative real-time polymerase chain reaction (qRT-PCR) analysis was performed (StepOne; Applied Biosystems, Foster City, CA, USA) using SYBR-Green (Takara, Shiga, Japan). All PCR experiments were performed in triplicate. The TGM3 primer sequences are as follows: sense: 5′-TCAACTGGCAGACGGCCTTCA-3′ and antisense 5′-GTACCGTCCTATGGGTGCGCT-3′.

Western blot analysis

Total protein was extracted using RIPA lysis buffer and the concentrations in different samples were determined using the BCA kit (Beyotime Institute of Biotechnology, Haimen, China). An equivalent amount of protein (30 μg) from different samples was separated by SDS-PAGE, and then transferred onto a nitrocellulose membrane (Millipore, Bedford, MA, USA). The membrane was blocked with 3.0% non-fat milk at room temperature for 1 h and then blotted with rabbit anti-TGM3 antibody or mouse anti-β-actin antibody (1:10,000) purchased from Sigma (San Francisco, CA, USA) overnight at 4°C. After incubation with horseradish peroxidase-conjugated secondary antibodies (1:2,000; Bioss, Beijing, China) for 1 h at room temperature, the protein complexes was detected using enhanced chemiluminescence reagents (Pierce, Rockford, IL, USA). β-actin was used as the internal control. The signal intensity on the scanned images was analyzed using Image-Pro Plus 6.0 software.

Cell proliferation assay

Cells were seeded in 96-well plates at 6×103 cells/well and the surviving fractions were determined at 0, 24, 48, 72 and 96 h using the MTT assay, as previously described (18). The absorbance of each well was measured with a spectrophotometer (Bio-Rad Laboratories, Hercules, CA, USA) at 570 nm. Each experiment was performed in triplicate (19).

Cell migration and invasion assays

EC cells were grown to confluence in 12-well plastic dishes and treated with recombinant human TGM3 protein or a scramble peptide. Then, 24 h after transfection, linear scratch wounds (in triplicate) were created on the confluent cell monolayers using a 200 μl pipette tip. To remove cells from the cell cycle prior to wounding, cells were maintained in serum-free media. A total of 10 areas were selected randomly from each well, and the cells in three wells from each group were quantified.

For the invasion assays, 24 h after transfection, 1×105 cells in serum-free media were seeded in Transwell migration chambers. The upper chamber of the Transwell inserts was coated with Matrigel (Sigma-Aldrich, St. Louis, MO, USA). Medium containing 20% FBS was added to the lower chamber. After 24 h, the non-invading cells were removed with cotton wool. Invasive cells located on the lower surface of the chamber were stained with May-Grunwald-Giemsa stain (Sigma-Aldrich) and counted using a microscope (Olympus Corp., Tokyo, Japan). From these images, the number of invasive cells was counted. Experiments were independently repeated three times.

Apoptosis assay

Apoptosis was assayed by an Annexin V apoptosis detection kit (BD Biosciences, San Diego, CA, USA). Following transfection for 72 h, the cells were collected and detected using an Annexin V fluorescein isothiocyanate kit (FITC) according to the manufacturer's instructions. In brief, cells transfected with the recombinant human vasostatin-2 or vasostatin-2 antibody were resuspended in 100 ml binding buffer, at a density of 1×106 cells/ml, then incubated with Annexin V-FITC and PI for 15 min. The cells were analyzed with Beckman CXP software on a FC-500 flow cytometer (Beckman Coulter, Pasadena, CA, USA) within 1 h of cell collection.

ELISA analysis

The levels of p65 concentrations in culture medium were determined by ELISA (Tanjin Biotechnology Co., Shanghai, China) following the manufacturer's instructions. The absorbance was assessed at 450 nm using a 680XR microplate reader (Bio-Rad Laboratories).

Statistical analysis

The differences were analyzed using Student's t-test for two groups and examined by the χ2 test. A P-value <0.05 was considered to indicate statistically significant differences. Data were processed as mean ± standard deviation (SD). All statistical analyses were performed using SPSS 16.0 software (SPSS, Inc., Chicago, IL, USA).

Results

TGM3 is downregulated in EC tissues and cell lines

In order to confirm the potential roles of TGM3 in EC, RT-PCR was performed to investigate the mRNA levels of TGM3 in 58 paired EC specimens compared to the corresponding adjacent non-cancerous tissues (NCTs) (Fig. 1A). The TGM3 mRNA levels in the EC specimens were significantly lower than those of non-cancerous tissues. Western blot assay was performed to investigate the protein levels of TGM3 in EC specimens, same as above, the protein levels of TGM3 were significantly downregulated in the EC specimens compared to the corresponding adjacent non-cancerous tissues (Fig. 1B). In addition, RT-PCR and western blot assay were performed to investigate the expression level of four EC cell lines and a normal human esophageal epithelial cell line. The TGM3 mRNA levels in the EC cell lines were significantly lower than those of normal esophageal epithelial cells (Fig. 1C). Correspondingly, the protein levels of TGM3 were markedly downregulated in the four EC cell lines compared with the levels in normal epithelial cells using western blot analysis (Fig. 1D). These data show that the expression of TGM3 is universally downregulated in EC cells and patients, indicating that a change of TGM3 expression may affect the development of EC.

Ectopic expression of TGM3 directly affects cell proliferation in vitro

It is well-known that cell proliferation is a key event in the formation and development of cellular oncogenesis. In order to investigate the role of TGM3 in EC cell proliferation, TGM3 was overexpressed or repressed in SKGT-4 cells and KYSE-510 cells by transfection with recombinant human TGM3 or TGM3 antibody, qRT-PCR (Fig. 2A and B) and western blot analysis (Fig. 2C and D) evaluation demonstrated TGM3 expression in SKGT-4 and KYSE-510 cell lines. The MTT assay showed that overexpression of TGM3 inhibited cell proliferation. Conversely, downregulating the expression of TGM3 accelerated cell proliferation in comparison with the control group (Fig. 2E and F). Similar data were also obtained in the other two cell lines (data not shown).

Ectopic expression of TGM3 protein affects cell apoptosis in EC cells

One of the hallmarks of cancer is its ability to evade apoptosis (20). To investigate the effect of TGM3 on EC cell apoptosis, we performed a series of experiments in EC cell lines, which were stimulated by transfection with recombinant human TGM3 or TGM3 antibody to obtain TGM3 protein ectopic expression. We then investigated the effect of TGM3 on EC cell apoptosis using Annexin V-FITC staining. As shown in Fig. 3A, overexpression of TGM3 significantly increased apoptosis in SKGT-4 cells. The percentage of apoptotic cells increased to 9.8% after transfection with recombinant human TGM3. Suppressed expression of TGM3 increased apoptosis in SKGT-4 cells (Fig. 3B). Overall, these data indicate that TGM3 may play the role of promoting apoptosis in EC cells.

Ectopic expression of TGM3 directly affects cell migration and invasion in vitro

To analyze the role of TGM3 in cell migration and invasion, which are the key determinants of malignant progression and metastasis, scratch assays and Transwell assays were performed in the SKGT-4 and KYSE-510 cell lines. Cells treated with recombinant human TGM3 were distinctively less migratory and invasive; however, cells treated with the TGM3 antibody were distinctively more migratory and invasive than untreated cells at 48 h after scratching (Fig. 4). These results indicate that TGM3 plays an important role in regulating the proliferation of EC cells and strongly suggest that introduction of TGM3 could inhibit cellular oncogenesis by suppressing the migration and invasion of EC cells.

Overexpression of TGM3 promotes activation of the NF-κB signaling pathway

The NF-κB signaling pathway has been demonstrated to play an important role in cellular energy homeostasis and reported to be extensively involved in cell proliferation, migration and differentiation (2123). To further explore the mechanisms involved in TGM3 induced tumor cell growth and apoptosis, we analyzed the NF-κB pathway at the protein level via ELISA and western blotting. The results show that elevated p65 concentrations in culture medium of SKGT-4 cells were found with TGM3 overexpression (Fig. 5A). Furthermore, the protein expression of p65 was also found to be markedly increased after TGM3 pre-treatment (Fig. 5B); the same effect was observed in the other three cell lines. These results suggest that TGM3 induces activation of the NF-κB pathway.

TGM3 modulates EC cell growth and apoptosis by targeting the NF-κB signaling pathway

To further delineate the effect of NF-κB on TGM3 modulated EC cell growth and apoptosis, the NF-κB signaling inhibitor Bay11-7082 (20 mol/l) was used. After a series of functional restoration assays, we found that the decrease in SKGT-4 cell proliferation (Fig. 6A) and migration (Fig. 6B) induced by TGM3 upregulation was markedly strengthened following Bay11-7082 stimulation. Moreover, disruption of NF-κB signaling by Bay11-7082 treatment also significantly decreased the apoptosis rate (Fig. 6C). These results indicate that TGM3 may repress EC cell proliferation and migration and promote apoptosis by modulating the NF-κB signaling pathway.

Discussion

A variety of treatments is currently available for EC. The choice of treatment is crucial, recurrent or distant metastases as the limitations of conventional treatments, therefore, it is necessary to search for novel approaches. Recently, several research groups have reported certain genes and signaling molecules that are potentially involved in EC initiation and progression (24,25). These molecular targets may provide significant clues for early diagnosis, prognosis and new targeted therapies (2628). Previous studies have found that TGM3 expression is downregulation in head and neck cancer, laryngeal carcinoma and oral squamous cell carcinoma (2932), other reports found that TGM3 expression is decreased in EC tissues compared with normal tissues (29). However, the exact function and mechanisms of TGM3 in EC are largely unknown. In this study, we verified this finding by assessing the mRNA and protein levels of TGM3 in four EC cell lines and 58 EC specimens. We found that the levels of this TGM3 were much lower in EC tissues and cell lines compared with adjacent controls. Our findings are consistent with previous reports, these results allowed us to speculate that the overexpression of TGM3 may confer a survival advantage to EC patients. Since metastasis is the major cause of morbidity and mortality in EC patients, we investigated the roles of TGM3 in cell migration and invasion of EC cell lines. Transglutaminase (TGM) enzymes are widespread in both plants and animals, they are a family of calcium-dependent enzymes that catalyze the formation of isopeptide bonds (34,35). TGM3 is expressed predominantly during the late stages of the terminal differentiation of the epidermis and in certain cell types of hair follicles (36). Although TGM3 downregulation has been found in many cases, however, little information is available concerning its function and mechanisms involvement in EC. The results of this study indicate that TGM3 plays an important role in regulating the proliferation of EC cells and strongly suggest that the introduction of TGM3 could inhibit cellular oncogenesis by suppressing the migration and invasion of EC cells. We consider TGM3 to be a strong biomarker candidate of EC, because its expression clearly correlated with cancer progression in our study and it has also been shown to be potentially relevant to ESCC (esophageal squamous cell carcinoma) (37).

Further studies identified that TGM3 promotes activation of the NF-κB signaling pathway. Recently, abundant studies have confirmed the important roles of NF-κB signaling in cancer development and progression, including proliferation, survival, angiogenesis and metastasis (38). The significance of NF-κB and its therapeutic value as a target for cancer therapy have been investigated in several types of human cancer (39,40). Only a handful of studies to date have explored the involvement of NF-κB activation in EC (4143) in which the NF-κB status correlated positively with metastasis, resistance to chemotherapy and patient survival (4446). The present study broadens our understanding of the roles that TGM3 plays in EC by targeting NF-κB signaling and thus indicates that it can be a powerful target for cancer therapy.

In conclusion, this study showed that TGM3 expression is low in EC tissues and cell lines, this result is consistent with the previous studies. We also found that in EC cell lines overexpression of TGM3 inhibits cell proliferation and induces apoptosis by activation of NF-κB signaling pathway. As the TGM3 expression level is associated with EC cell growth process, promotion of TGM3 expression in EC tissues may inhibit tumor cell amplification. Therefore, the present study may provide a therapeutic strategy for patients with EC.

Acknowledgments

The present study is supported by grants from the Key Scientific Research Project of Henan Province (14A310017) and the Key Project of Henan Provincial Department of Science and Technology (142102310311).

References

1 

Kamangar F1, Qiao YL, Schiller JT, Dawsey SM, Fears T, Sun XD, Abnet CC, Zhao P, Taylor PR and Mark SD: Human papillomavirus serology and the risk of esophageal and gastric cancers: results from a cohort in a high-risk region in China. Int J Cancer. 119:579–584. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Siegel R, Naishadham D and Jemal A: Cancer statistics for Hispanics/Latinos, 2012. CA Cancer J Clin. 62:283–298. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Gaur P, Kim MP and Dunkin BJ: Esophageal cancer: Recent advances in screening, targeted therapy, and management. J Carcinog. 13:112014. View Article : Google Scholar : PubMed/NCBI

5 

Ahvazi B, Kim HC, Kee SH, Nemes Z and Steinert PM: Three-dimensional structure of the human transglutaminase 3 enzyme: Binding of calcium ions changes structure for activation. EMBO J. 21:2055–2067. 2002. View Article : Google Scholar : PubMed/NCBI

6 

Hitomi K, Horio Y, Ikura K, Yamanishi K and Maki M: Analysis of epidermal-type transglutaminase (TGase 3) expression in mouse tissues and cell lines. Int J Biochem Cell Biol. 33:491–498. 2001. View Article : Google Scholar : PubMed/NCBI

7 

Hitomi K: Transglutaminases in skin epidermis. Eur J Dermatol. 15:313–319. 2005.PubMed/NCBI

8 

Hitomi K, Presland RB, Nakayama T, Fleckman P, Dale BA and Maki M: Analysis of epidermal-type transglutaminase (transglutaminase 3) in human stratified epithelia and cultured keratinocytes using monoclonal antibodies. J Dermatol Sci. 32:95–103. 2003. View Article : Google Scholar : PubMed/NCBI

9 

Kalinin AE, Kajava AV and Steinert PM: Epithelial barrier function: Assembly and structural features of the cornified cell envelope. BioEssays. 24:789–800. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Eckert RL, Sturniolo MT, Ann-Marie B, Monica R and Rorke EA: Transglutaminase function in epidermis. JJ Invest Dermatol. 124:481–492. 2005. View Article : Google Scholar

11 

Guang HE, Zhao Z, Weineng FU, Sun X, Zhenming XU and Sun K: Study on the loss of heterozygosity and expression of transglutaminase 3 gene in laryngeal carcinoma. Zhonghua Yi Xue Yi Chuan Xue Za Zhi. 19:120–123. 2002.In Chinese.

12 

Uemura N, Nakanishi YH, Saito S, Nagino M, Hirohashi S and Kondo T: Transglutaminase 3 as a prognostic biomarker in esophageal cancer revealed by proteomics. Int J Cancer. 124:2106–1255. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Negishi A, Masuda M, Ono M, Honda K, Shitashige M, Satow R, Sakuma T, Kuwabara H, Nakanishi Y, Kanai Y, et al: Quantitative proteomics using formalin-fixed paraffin-embedded tissues of oral squamous cell carcinoma. Cancer Sci. 100:1605–1611. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Méndez E, Fan W, Choi P, Agoff SN, Whipple M, Farwell DG, Futran ND, Weymuller EA Jr, Zhao LP and Chen Cl: Tumor-specific genetic expression profile of metastatic oral squamous cell carcinoma. Head Neck. 29:803–814. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Viatour P, Merville MP, Bours V and Chariot A: Phosphorylation of NF-kappaB and IkappaB proteins: Implications in cancer and inflammation. Trends Biochem Sci. 30:43–52. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Li B, Li YY, Tsao SW and Cheung AL: Targeting NF-kappaB signaling pathway suppresses tumor growth, angiogenesis, and metastasis of human esophageal cancer. Mol Cancer Ther. 8:2635–2644. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Bassères DS and Baldwin AS: Nuclear factor-kappaB and inhibitor of kappaB kinase pathways in oncogenic initiation and progression. Oncogene. 25:6817–6830. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Zhang L, Liu X, Jin H, Guo X, Xia L, Chen Z, Bai M, Liu J, Shang X, Wu K, et al: miR-206 inhibits gastric cancer proliferation in part by repressing cyclinD2. Cancer Lett. 332:94–101. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Cotter TG: Apoptosis and cancer: The genesis of a research field. Nat Rev Cancer. 9:501–507. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Darius W, Ilja M, Margitta E, Christian K and Barbara K: Tumor necrosis factor alpha triggers proliferation of adult neural stem cells via IKK/NF-kappaB signaling. BMC Neurosci. 7:1–18. 2006. View Article : Google Scholar

22 

Palumbo R, Galvez BG, Pusterla T, De Marchis F, Cossu G, Marcu KB and Bianchi ME: Cells migrating to sites of tissue damage in response to the danger signal HMGB1 require NF-kappaB activation. J Cell Biol. 179:33–40. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Wu Y and Zhou BP: TNF-alpha/NF-kappaB/Snail pathway in cancer cell migration and invasion. Br J Cancer. 102:639–644. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Wong GS, Lee JS, Park YY, Klein-Szanto AJ, Waldron TJ, Cukierman E, Herlyn M, Gimotty P, Nakagawa H and Rustgi AK: Periostin cooperates with mutant p53 to mediate invasion through the induction of STAT1 signaling in the esophageal tumor microenvironment. Oncogenesis. 2:e592013. View Article : Google Scholar : PubMed/NCBI

25 

Kaz AM, Luo Y, Dzieciatkowski S, Chak A, Willis JE, Upton MP, Leidner RS and Grady WM: Aberrantly methylated PKP1 in the progression of Barrett's esophagus to esophageal adenocarcinoma. Genes Chromosomes Cancer. 51:384–393. 2012. View Article : Google Scholar

26 

Shigaki H, Baba Y, Watanabe M, Murata A, Ishimoto T, Iwatsuki M, Iwagami S, Nosho K and Baba H: PIK3CA mutation is associated with a favorable prognosis among patients with curatively resected esophageal squamous cell carcinoma. Clin Cancer Res. 19:2451–2459. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Shah AK, Cao KA, Choi E, Chen D, Gautier B, Nancarrow D, Whiteman DC, Saunders NA, Barbour AP, Joshi V, et al: Serum glycoprotein biomarker discovery and qualification pipeline reveals novel diagnostic biomarker candidates for esophageal adenocarcinoma. Mol Cell Proteomics. 14:3023–3039. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Isozaki Y, Hoshino I, Nohata N, Kinoshita T, Akutsu Y, Hanari N, Mori M, Yoneyama Y, Akanuma N, Takeshita N, et al: Identification of novel molecular targets regulated by tumor suppressive miR-375 induced by histone acetylation in esophageal squamous cell carcinoma. Int J Oncol. 41:985–994. 2012.PubMed/NCBI

29 

Uemura N, Nakanishi Y, Kato H, Saito S, Nagino M, Hirohashi S and Kondo T: Transglutaminase 3 as a prognostic biomarker in esophageal cancer revealed by proteomics. Int J Cancer. 124:2106–2115. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Wu X, Cao W, Wang X, Zhang J, Lv Z, Qin X, Wu Y and Chen W: TGM3, a candidate tumor suppressor gene, contributes to human head and neck cancer. Mol Cancer. 12:1512013. View Article : Google Scholar : PubMed/NCBI

31 

Liu J, Zhou Y, Wan J and Liu Z: Expression of TGM3 protein and its significance in laryngeal carcinoma. Lin Chung Er Bi Yan Hou Tou Jing Wai Ke Za Zhi. 26:101–103. 2012.In Chinese. PubMed/NCBI

32 

Ye H, Yu T, Temam S, Ziober BL, Wang J, Schwartz JL, Mao L, Wong DT and Zhou X: Transcriptomic dissection of tongue squamous cell carcinoma. BMC Genomics. 9:692008. View Article : Google Scholar : PubMed/NCBI

33 

Kondoh N, Ishikawa T, Ohkura S, Arai M, Hada A, Yamazaki Y, et al: Gene expression signatures that classify the mode of invasion of primary oral squamous cell carcinomas. Mol Carcinog. 47:744–756. 2008. View Article : Google Scholar : PubMed/NCBI

34 

Lorand L and Graham RM: Transglutaminases: Crosslinking enzymes with pleiotropic functions. Nat Rev Mol Cell Biol. 4:140–156. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Griffin M, Casadio R and Bergamini CM: Transglutaminases: Nature's biological glues. Biochem J. 368:377–396. 2002. View Article : Google Scholar : PubMed/NCBI

36 

Candi E, Schmidt R and Melino G: The cornified envelope: A model of cell death in the skin. Nat Rev Mol Cell Biol. 6:328–340. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Liu W, Yu ZC, Cao WF, Ding F and Liu ZH: Functional studies of a novel oncogene TGM3 in human esophageal squamous cell carcinoma. World J Gastroenterol. 12:3929–3932. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Karin M and Greten FR: NF-kappaB: Linking inflammation and immunity to cancer development and progression. Nat Rev Immunol. 5:749–759. 2005. View Article : Google Scholar : PubMed/NCBI

39 

Sourbier C, Danilin S, Lindner V, Steger J, Rothhut S, Meyer N, Jacqmin D, Helwig JJ, Lang H and Massfelder T: Targeting the nuclear factor-kappaB rescue pathway has promising future in human renal cell carcinoma therapy. Cancer Res. 67:11668–11676. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Robe PA, Bentires-Alj M, Bonif M, Rogister B, Deprez M, Haddada H, Khac MT, Jolois O, Erkmen K, Merville MP, et al: In vitro and in vivo activity of the nuclear factor-kappaB inhibitor sulfasalazine in human glioblastomas. Clin Cancer Res. 10:5595–5603. 2004. View Article : Google Scholar : PubMed/NCBI

41 

Abdel-Latif MMM, O'Riordan J, Windle HJ, Carton E, Ravi N, Kelleher D and Reynolds JV: NF-kappaB activation in esophageal adenocarcinoma: Relationship to Barrett's metaplasia, survival, and response to neoadjuvant chemoradiotherapy. Ann Surg. 239:491–500. 2004. View Article : Google Scholar : PubMed/NCBI

42 

Abdel Latif MM, O'Riordan JN, Kelleher D and Reynolds JV: Activated nuclear factor-kappaB and cytokine profiles in the esophagus parallel tumor regression following neoadjuvant chemoradiotherapy. Dis Esophagus. 18:246–252. 2005. View Article : Google Scholar

43 

Konturek PC, Nikiforuk A, Kania J, Raithel M, Hahn EG and Mühldorfer S: Activation of NFkappaB represents the central event in the neoplastic progression associated with Barrett's esophagus: A possible link to the inflammation and overexpression of COX-2, PPARgamma and growth factors. Dig Dis Sci. 49:1075–1083. 2004. View Article : Google Scholar : PubMed/NCBI

44 

Izzo JG, Correa AM, Wu TT, Malhotra U, Chao CK, Luthra R, Ensor J, Dekovich A, Liao Z, Hittelman WN, et al: Pretherapy nuclear factor-kappaB status, chemoradiation resistance, and metastatic progression in esophageal carcinoma. Mol Cancer Ther. 5:2844–2850. 2006. View Article : Google Scholar : PubMed/NCBI

45 

Izzo JG, Malhotra U, Wu TT, Ensor J, Luthra R, Lee JH, Swisher SG, Liao Z, Chao KS, Hittelman WN, et al: Association of activated transcription factor nuclear factor kappab with chemoradiation resistance and poor outcome in esophageal carcinoma. J Clin Oncol. 24:748–754. 2006. View Article : Google Scholar : PubMed/NCBI

46 

Izzo JG, Malhotra U, Wu TT, Luthra R, Correa AM, Swisher SG, Hofstetter W, Chao KS, Hung MC and Ajani JA: Clinical biology of esophageal adenocarcinoma after surgery is influenced by nuclear factor-kappaB expression. Cancer Epidemiol Biomarkers Prev. 16:1200–1205. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2016
Volume 36 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li W, Zhang Z, Zhao W and Han N: Transglutaminase 3 protein modulates human esophageal cancer cell growth by targeting the NF-κB signaling pathway. Oncol Rep 36: 1723-1730, 2016
APA
Li, W., Zhang, Z., Zhao, W., & Han, N. (2016). Transglutaminase 3 protein modulates human esophageal cancer cell growth by targeting the NF-κB signaling pathway. Oncology Reports, 36, 1723-1730. https://doi.org/10.3892/or.2016.4921
MLA
Li, W., Zhang, Z., Zhao, W., Han, N."Transglutaminase 3 protein modulates human esophageal cancer cell growth by targeting the NF-κB signaling pathway". Oncology Reports 36.3 (2016): 1723-1730.
Chicago
Li, W., Zhang, Z., Zhao, W., Han, N."Transglutaminase 3 protein modulates human esophageal cancer cell growth by targeting the NF-κB signaling pathway". Oncology Reports 36, no. 3 (2016): 1723-1730. https://doi.org/10.3892/or.2016.4921