Type III TGF-β receptor inhibits cell proliferation and migration in salivary glands adenoid cystic carcinoma by suppressing NF-κB signaling

  • Authors:
    • Dongyang Xu
    • Duo Li
    • Zhiyong Lu
    • Xingli Dong
    • Xiaofeng Wang
  • View Affiliations

  • Published online on: November 3, 2015     https://doi.org/10.3892/or.2015.4390
  • Pages: 267-274
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

It is known that the TGF-β superfamily receptors act as master regulators of cancer progression. However, alteration and role of type III TGF-β receptor (TβRIII, or betaglycan) as the most abundant of the TGF-β receptor has not been explored in salivary gland adenoid cystic carcinoma (ACC). Here, we reported that tumor biopsies and matched normal human salivary glands from patients with ACC were examined for the expression of TβRIII. The expression of TβRIII protein is significantly decreased in ACC patients based on immunohistochemistry and western blot analysis. In vitro, a transient overexpression of TβRIII markedly induced apoptosis and cell cycle arrest in the G2/M phase, thereby inhibited cell viability and migration of ACC-M cells. Co-immunoprecipitation revealed that TβRIII, scaffolding protein-arrestin2 (β-arrestin2) and IκBα formed a complex. Transient overexpression of TβRIII decreased p-p65 expression and increased IκBα expression, which was abolished by knockdown of β-arrestin2. The present study defines TβRIII as a biomarker exerting antitumor action on ACC progression.Gene therapy of TβRIII may be a powerful new approach for ACC disease.

Introduction

Salivary gland adenoid cystic carcinoma (ACC) is the most common epithelial malignant neoplasm of salivary glands (1,2), which is characterized by slow but aggressive growth, intensive local invasion, distant metastasis to the lungs at early or late stages, multiple recurrence, and poor long-term survival rates (3,4). The primary treatment of ACC is radical surgery, which can be followed by post-operative radiotherapy. Unfortunately, surgery, chemotherapy, and radiation therapy provide little improvement in survival (5). Therefore, there is a need for better understanding of the biology of ACC and the development of therapeutic approaches based on relevant targets.

Recently, many efforts have been made to examine the antitumor effect of TGF-β superfamily co-receptor, the type III TGF-β receptor (TβRIII, also known as betaglycan) in both in vitro and in vivo cancer models, including breast (6,7), lung (8,9), prostate (10,11), pancreas (12), ovary (13) and oral squamous cell carcinomas (14). Conversely, a report by Gatza et al (15) showed that TβRIII enhances colon cancer cell migration and growth. A recent study by Jovanović et al (16) also demonstrated that TβRIII is a tumor promoter in mesenchymal-stem like triple-negative breast cancer. These findings suggest that TβRIII exerts dual action on cancer progression depending upon the cell type. To date, no studies have investigated the expression and role of TβRIII in progression of ACC disease.

Here, we demonstrated that the expression of TβRIII is reduced in ACC. Transient transfection of TβRIII significantly decreased viability, migration and induced apoptosis in human high metastasis cell lines of ACCs (ACC-M), with significant inhibition of nuclear factor κB (NF-κB) signaling through its interaction with β-arrestin2.

Materials and methods

Tissue samples

Human adenoid cystic carcinoma and their adjacent non-cancer tissues were collected immediately after surgery from the Department of Oral and Maxillofacial Surgery at the Second Affiliated Hospital of Harbin Medical University in China, and informed consent was obtained from all participating patients or their guardians. The study was approved by the Institutional Review Board of Harbin Medical University.

Cell culture

Human high metastasis cell lines of ACCs (ACC-M) were cultured as previously described (17). ACC-M cells were cultured in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% (v/v) fetal bovine serum and 100 U/ml penicillin and streptomycin.

Plasmids and transfections

Cells were transfected with pc-DNA3.1-mTβRΙΙΙ plasmid (GeneChem Co., Ltd., Shanghai, China) with DNA concentrations of 0.5 and 1 µg/ml. The pc-DNA3.1-plasmid was used as an empty vector. Transient transfections were carried out by Fugene 6 (Roche Molecular Biochemicals, Mannheim, Germany) according to the manufacturer's instructions.

Cell viability assay

Cells were seeded in a 96-well microplate at a density of 1×104 cells/well and treated as designated. Cells were incubated with 20 µl of MTT (5 mg/ml; Sigma-Aldrich, St. Louis, MO, USA) for 4 h at 37°C in the dark. After medium was removed, 150 µl of DMSO was added to the wells. The absorbance was measured using a microplate spectrophotometer (Tecan, Austria) at 490 nm. The cell viability was calculated as a ratio of the mean OD value in treated versus untreated cells.

Assessment of electron microscopy

ACC-M cells were cultured in 60-mm plates, collected in phosphate-buffered saline (PBS) solution and fixed with 2% (v/v) paraformaldehyde (PFA) containing 2.5% (w/v) glutaraldehyde (Paesel-Lorei) buffered in HMSS (Hank's modified salt solution) at 4°C for 4 h. The cells were further fixed in 1% (w/v) OsO4 solution buffered by 0.1 M cacodylate (pH 7.2) at 4°C for 2 h. Then, the cells were scraped off from the plastic and dehydrate dinethanol. Dehydration was completed in propylene oxide. The specimens were embedded in Epon medium and dissected into 60–70-nm sections. Specimens were analyzed and documented with a JEOL 1200 electron microscope (JEOL Ltd., Tokyo, Japan).

Quantification of apoptosis and cell cycle by flow cytometry

Apoptosis in ACC-M cells, transfected with pcDNA3.1 expressing empty vector or TβRIII for 24 h, was quantified using the Annexin V-fluorescein isothiocyanate (FITC) apoptosis detection kit (Invitrogen, Carlsbad, CA, USA). Briefly, the cells were harvested, washed in cold PBS, and resuspended in Annexin V-labeling solution. After a 15-min incubation at room temperature, cells were then stained with 5 µg/ml propidium iodide (PI) and immediately subjected to Coulter Epics XL flow cytometer (Beckman Coulter, Miami, FL, USA). The effect of TβRIII overexpression on cell cycle distribution was determined by flow cytometry of DNA content from the nuclei of the cells. After treatment, ACC-M cells were harvested, washed with PBS and fixed in 70% ethanol and treated with 80 mg/ml RNase A at 37°C for 30 min. DNA was stained with 50 mg/ml PI and analyzed using a Coulter Epics XL flow cytometer (Beckman Coulter).

RNA interference

RNA interference was performed with siRNA duplexes (GenePharma, Shanghai) with sequences specifically targeting β-arrestin2 (5′-AAGGACCGCAAAGU GUUUGUG-3′), or control non-specific siRNA using Lipofactamine 2000 (Invitrogen) according to the manufacturer's protocol. Knockdown of expression of the target was determined by western blot analysis.

Co-immunoprecipition (Co-IP) and western blot analysis

Total protein samples were extracted from the cultured cells. Approximately 5 µg of antibody specific to TβRIII or β-arrestin2 protein was added to cell lysates and then incubated for 12 h at 4°C. The antibody-protein immune complexes were precipitated together with protein A/G PLUS-Agarose (rabbit polyclonal; Santa Cruz Biotechnology), which binds most antibodies, then incubated at 4°C overnight. When TβRIII or β-arrestin2 protein binds to one of them, TβRIII or β-arrestin2 protein can be identified by western blot analysis. Briefly, proteins were resolved by SDS-polyacrylamide gel electrophoresis, and transferred to PVDF membranes (Amersham Biosciences). All the blots were blocked with 5% non-fat dry milk powder in Tris-buffered saline with Tween-20 for 2 h and then incubated with either primary rabbit anti-TβRIII, anti-p-p65, anti-β-arrestin2, or anti-IκBα (1:1,000; Cell Signaling Technology, Beverly, MA, USA). Western blot bands were quantified using Odyssey v1.2 software by measuring the band intensity (area × OD) for each group and normalizing it to GAPDH or β-actin (Zhongshan, Beijing, China) as an internal control.

Scratch wound-healing assay

For the scratch assay, ACC-M cells were seeded at 1×105 cells/6-well plate and allowed to grow overnight. Cells were transiently transfected with pcDNA3.1 expressing empty vector or TβRIII. After a 24-h transfection, a scratch wound was applied using a pipette tip, and a baseline image was obtained. Scratch wound closure was monitored over a 24 h period. The healing of the wounds through cell migration was quantified by measuring the wound distance.

Transwell migration assay

Transwell migration and invasion assay were performed using the 24-well cell culture inserts without Matrigel and Matrigel invasion chambers (8-µm pore; BD Biosciences), respectively. Briefly, 5×104 cells were resuspended in 250 µl serum-free RPMI-1640 and added into the inserts. A total of 500 µl DMEM with 10% FBS was added to the lower chamber. After allowing cells to migrate for 4 h or invasion for 22 h, cells on the upper surface of the membrane were removed using a cotton swab, and the membranes were fixed with methanol and stained with crystal violet. The number of migrating or invading cells was determined by averaging cell counts from nine randomly selected ×100 fields.

Immunohistochemistry (IHC)

Serial sections (5–6-µm thick) were made from paraffin-embedded tissue blocks and mounted on silane-coated glass slides (Matsunami Glass, Osaka, Japan). One section from each tissue block was stained with hematoxylin and eosin (H&E), and the others were used for IHC. IHC staining was performed using the standard streptavidin-biotin-peroxidase complex method. Briefly, paraffin sections of ACC tissues were deparaffinized, blocked with 10% normal goat serum for 10 min, and incubated with anti-TβRIII overnight at 4°C. The tissue section was then incubated with biotinylated goat anti-rabbit immunoglobulin at a concentration of 1:75 at 37°C for 30 min. The status of TβRIII expression was assessed by two independent investigators without prior knowledge of the clinicopathological data.

Statistical analysis

The results are expressed as the mean ± standard deviation (SD). Statistical significance was evaluated using the unpaired Student's t-test. A p-value <0.05 was considered statistically significant.

Results

Tumor characteristics

Table I summarizes the clinical attributes of the patients, who provided the 12 ACC tumor samples. All the tumors had arisen sporadically. Of these, 8 occurred in men, and the median age at the time of presentation was 61 years (range, 48–77 years). Tumors arose at the following sites: submandibular gland (7 tumors), parotid gland (3 tumors), and tongue (2 tumors). The tumors were classified as follows based on morphologic subtype: combined tubular and cribriform (8 tumors), cribriform (2 tumors), combined tubular and solid (2 tumors).

Table I

Clinicopathological features of ACC samples.

Table I

Clinicopathological features of ACC samples.

CaseGenderAge (years)Tumor locationHistological type
1M74Submandibular glandTubular + cribriform
2M69Submandibular glandTubular + cribriform
3F57Parotid glandTubular + cribriform
4F77Submandibular glandTubular + cribriform
5M63TongueCribriform
6M51Parotid glandTubular + solid
7M48Parotid glandTubular + solid
8F66TongueCribriform
9M54Submandibular glandTubular + cribriform
10F66Submandibular glandTubular + cribriform
11M59Submandibular glandTubular + cribriform
12M49Submandibular glandTubular + cribriform

[i] M, male; F, female.

Decreased TβRIII expression in human adenoid cystic carcinoma

Loss or reduced expression of TβRIII has been demonstrated in multiple human cancers. To confirm the expression of TβRIII in human adenoid cystic carcinoma, we performed IHC analysis for TβRIII expression in matched normal human salivary glands and adenoid cystic carcinoma specimens. Representative samples are shown in Fig. 1A. H&E staining showed that adenoid cystic carcinoma exhibited cribriform architecture with pseudocystic spaces filled with eosiniphilic coagulum. IHC showed moderate to strong TβRIII expression in normal salivary glands and negative TβRIII expression in ACC specimens. We also examined the expression of TβRIII in matched normal human salivary glands and adenoid cystic carcinoma specimens. Lower levels of TβRIII protein were observed in human adenoid cystic carcinoma specimens (Fig. 1B). These results suggest that TβRIII expression is significantly decreased in adenoid cystic carcinoma, with loss of TβRIII expression correlating with adenoid cystic carcinoma progression.

TβRIII overexpression suppresses the viability of ACC-M cells

To validate the positive functional involvement of TβRIII in adenoid cystic carcinoma, we transfected ACC-M cells with a plasmid encoding the constitutively active TβRIII. Successful transfection of TβRIII was verified by our data shown in Fig. 2A. Western blot analysis showed that the TβRIII protein level was increased in a concentration-dependent manner in cells treated with 0.5 and 1 µg/ml of TβRIII plasmid DNA (Fig. 2A). Cell viability was determined by MTT assay. Fig. 2B shows that the viability of ACC-M cells transfected with 0.5 or 1 µg/ml TβRIII plasmid was reduced dose-dependently.

TβRIII overexpression induces apoptosis in ACC-M cells

TGF-β signaling pathway induces programmed cell death in a variety of cell types. To determine the effects of TβRIII overexpression on ACC-M cell apoptosis, we used electron microscopy and flow cytometry to confirm the apoptotic changes. Under an electron microscope the cells with TβRIII overexpression exhibited robust changes in microstructure, including cell surface microvilli reduction, nuclear chromatin condensation, margination, and membrane blistering (Fig. 3A). We transfected ACC-M cells with a plasmid encoding TβRIII or NC plasmid, and apoptosis was assessed by flow cytometry in live cells stained with Annexin V-FITC/PI. As shown in Fig. 3B, TβRIII overexpression induced apoptosis in ACC-M cells in a dose-dependent manner.

TβRIII affects cell cycle progression in ACC-M cells

To define the mechanisms by which TβRIII regulated viability of ACC-M cells further, we examined the effect of increasing TβRIII expression on the cell cycle progression in ACC-M cells in vitro. Cell cycle analysis revealed that TβRIII over-expression resulted in a remarkable G2/M arrest in ACC-M cells (Fig. 4).

TβRIII delays and decreases ACC-M cells migration in vitro

We further examined whether TβRIII overexpression affected cell migration ability in ACC-M cells. Wound healing and Transwell assays were evaluated. As shown in Fig. 5, both wound healing and Transwell assays showed that TβRIII over-expression significantly inhibited the cellular transmigration ability compared with controls. These results strongly indicated that TβRIII may also regulate the cell migration ability of adenoid cystic carcinoma.

TβRIII negatively regulates NF-κB signaling through interacting with β-arrestin2 in ACC-M cells

NF-κB is a dimeric transcription factor that regulates genes involved in cell survival and proliferation (18). Increased NF-κB activity has been demonstrated in diverse human malignancies, including ACC-M, which is believed to enhance tumor cell survival by accelerating mitosis and inhibiting apoptosis (19). TGF-β has been reported to activate (20) or inhibit (21) NF-κB signaling through mechanisms yet to be fully defined. You et al (22) reported that TβRIII was involved in NF-κB regulation via its interaction with β-arrestin2 in MCF10A breast epithelial and MDA-MB-231 breast cancer cells. We next investigated the effect of TβRIII overexpression on NF-κB activity in ACC-M. Our results indicated that transiently increasing TβRIII expression decreased p-p65 expression (Fig. 6A) and increased IκBα expression (Fig. 6B). These results suggest that inhibition of NF-κB signaling represents a potential mechanism for TβRIII-mediated inhibition of cell apoptosis, migration, and mitotic arrest in ACC-M. Our results also confirmed that TβRIII interacted with β-arrestin2, and β-arrestin2 interacted with IκBα (Fig. 6C). TβRIII overexpression decreased p-p65 expression, and co-transfection of TβRIII and siRNA of β-arrestin2 resulted in an increase in phosphorylation of p65 (Fig. 6E). These results suggest that TβRIII through its interaction with β-arrestin2, negatively regulates NF-κB signaling in ACC-M.

Discussion

In the present study, we report the expression status of TβRIII in salivary gland adenoid cystic carcinoma for the first time. We define an important role for TβRIII expression in salivary gland adenoid cystic carcinoma as an inhibitor of cell growth and migration through its inhibitory effects on NF-κB signaling via its interaction with β-arrestin2.

Here, we demonstrated that the TβRIII expression was markedly repressed in adenoid cystic carcinoma specimens compared with matched normal human salivary glands, suggesting that a low TβRIII expression level is associated with salivary gland adenoid cystic carcinoma progression. Loss of TβRIII expression has also been described in breast, lung, prostate, pancreas, ovary, and oral squamous cell carcinomas as mentioned above. Turley et al (10) reported that the restoration of TβRIII expression in prostate cancer cells inhibits migration and invasion. Zheng et al (23) reported that transient overexpression of TβRIII induces apoptosis in human nasopharyngeal carcinoma CNE-2Z cells. Forced overexpression of TβRIII upregulated pro-apoptotic Bad and Bax protein, and downregulated anti-apoptotic p-Bad, Bcl-2, and XIAP protein. These results support that the TβRIII regulated multiple targets involved in cell proliferation by mediating the TGF-β superfamily ligand independent signaling. Furthermore, some studies reported that TβRIII had direct effects on regulating tumor migration, invasion, and proliferation without any cytokines and ligands (23,24). These data indicated that gene therapy of TβRIII may be a powerful new approach for cancer.

How does decreased TβRIII expression promote salivary gland adenoid cystic carcinoma progression? The great potential for hematogenous metastasis at an early stage is one of the unique characteristics of ACCs. Zhang and Peng (19) reported that the high expression levels of NF-κB were significantly correlated with ACC metastasis. Our results showed that the reason of high levels of NF-κB in ACC may be correlated with reduced expression of TβRIII. Regulation of TβRIII expression occurs at multiple levels. At the transcriptional level, TβRIII expression is negatively regulated by TGF-β1 through inhibition of the promoter in multiple cell types. In some tumors, especially late-stage tumors, a large amount of TGF-β1 was secreted, and inhibited the expression of TβRIII (24).

Zanotto-Filho et al (25) demonstrated that the pharmacological NF-κB inhibitors BAY117082 and MG132 induce cell arrest and apoptosis in leukemia cells through ROS-mitochondria pathway activation. Other groups also confirmed the result that inhibition of NF-κB pathways induces G2/M arrest and apoptosis in different cancer cells, including prostate cancer (26), glioblastoma (27) and melanoma (28). We studied the effects of TβRIII overexpression on cell viability, apoptosis, cell cycle, and in vitro cell migration in highly metastatic cell lines of human ACC-M. We demonstrated that TβRIII inhibited ACC-M cell growth and migration through its inhibitory effects on NF-κB signaling. As NF-κB activation should be accompanied by proteasome-mediated degradation of IκBα, we also investigated IκBα protein expression in response to TβRIII stimulation. Our results indicated that transiently increasing TβRIII expression decreased p-p65 expression and increased IκBα expression in ACC-M.

We also showed a novel interaction of TβRIII with the scaffolding protein, β-arrestin2, which results in TβRIII internalization and downregulation of TGF-β signaling. β-arrestin2 also scaffolds interacting receptors with the IκBα (Fig. 6C). The results are consistent with other groups. TβRIII has been shown to interact with β-arrestin2 through its cytoplasmic domain (29). Gao et al (30) have shown that β-arrestin2 directly interacts with IκBα and prevents phosphorylation and degradation of IκBα. Similar results have also been obtained from Witherow et al (31). You et al (22) demonstrated that TβRIII, through its interaction with β-arrestin2, negatively regulates NF-κB signaling in breast cancer.

In summary, we demonstrated that the TβRIII expression was markedly repressed in adenoid cystic carcinoma. Transient TβRIII overexpression induced apoptosis and G2/M arrest, inhibited cell growth and migration in ACC-M cells. We established the TβRIII/β-arrestin2 as an important negative regulator of NF-κB signaling in ACC-M and a potential mechanism for TβRIII-mediated inhibition of ACC-M migration and ACC progression. The present study defines TβRIII as a biomarker exerting antitumor action on ACC progression, thus, gene therapy of TβRIII may be a new approach for ACC disease.

Acknowledgments

This study was supported by the Natural Science Foundation for Youth of HeilongJiang Province (QC2011C006).

Abbreviations:

ACC

adenoid cystic carcinoma

TGF-β

transforming growth factor β

NF-κB

nuclear factor κB

BMPs

bone morphogenetic proteins

GDFs

growth and differentiation factors

TβRIII

the type III TGF-β receptor

ACC-M

high metastasis cell lines of ACCs

MTT

3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide

FITC

fluorescein isothiocyanate

PI

propidium iodide

H&E

hematoxylin and eosin

References

1 

Tian Z, Li L, Wang L, Hu Y and Li J: Salivary gland neoplasms in oral and maxillofacial regions: A 23-year retrospective study of 6982 cases in an eastern Chinese population. Int J Oral Maxillofac Surg. 39:235–242. 2010. View Article : Google Scholar

2 

Li J, Wang BY, Nelson M, Li L, Hu Y, Urken ML and Brandwein-Gensler M: Salivary adenocarcinoma, not otherwise specified: A collection of orphans. Arch Pathol Lab Med. 128:1385–1394. 2004.PubMed/NCBI

3 

Spiro RH, Huvos AG and Strong EW: Adenoid cystic carcinoma: Factors influencing survival. Am J Surg. 138:579–583. 1979. View Article : Google Scholar : PubMed/NCBI

4 

Zhang CY, Zhao YX, Xia RH, Han J, Wang BS, Tian Z, Wang LZ, Hu YH and Li J: RASSF1A promoter hypermethylation is a strong biomarker of poor survival in patients with salivary adenoid cystic carcinoma in a Chinese population. PLoS One. 9:e1101592014. View Article : Google Scholar : PubMed/NCBI

5 

Laurie SA and Licitra L: Systemic therapy in the palliative management of advanced salivary gland cancers. J Clin Oncol. 24:2673–2678. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Dong M, How T, Kirkbride KC, Gordon KJ, Lee JD, Hempel N, Kelly P, Moeller BJ, Marks JR and Blobe GC: The type III TGF-beta receptor suppresses breast cancer progression. J Clin Invest. 117:206–217. 2007. View Article : Google Scholar

7 

Bandyopadhyay A, López-Casillas F, Malik SN, Montiel JL, Mendoza V, Yang J and Sun LZ: Antitumor activity of a recombinant soluble betaglycan in human breast cancer xenograft. Cancer Res. 62:4690–4695. 2002.PubMed/NCBI

8 

Finger EC, Turley RS, Dong M, How T, Fields TA and Blobe GC: TbetaRIII suppresses non-small cell lung cancer invasiveness and tumorigenicity. Carcinogenesis. 29:528–535. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Hempel N, How T, Cooper SJ, Green TR, Dong M, Copland JA, Wood CG and Blobe GC: Expression of the type III TGF-beta receptor is negatively regulated by TGF-beta. Carcinogenesis. 29:905–912. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Turley RS, Finger EC, Hempel N, How T, Fields TA and Blobe GC: The type III transforming growth factor-beta receptor as a novel tumor suppressor gene in prostate cancer. Cancer Res. 67:1090–1098. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Sharifi N, Hurt EM, Kawasaki BT and Farrar WL: TGFBR3 loss and consequences in prostate cancer. Prostate. 67:301–311. 2007. View Article : Google Scholar

12 

Gordon KJ, Kirkbride KC, How T and Blobe GC: Bone morphogenetic proteins induce pancreatic cancer cell invasiveness through a Smad1-dependent mechanism that involves matrix metalloproteinase-2. Carcinogenesis. 30:238–248. 2009. View Article : Google Scholar :

13 

Mythreye K and Blobe GC: The type III TGF-beta receptor regulates epithelial and cancer cell migration through beta-arrestin2-mediated activation of Cdc42. Proc Natl Acad Sci USA. 106:8221–8226. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Meng W, Xia Q, Wu L, Chen S, He X, Zhang L, Gao Q and Zhou H: Downregulation of TGF-beta receptor types II and III in oral squamous cell carcinoma and oral carcinoma-associated fibroblasts. BMC Cancer. 11:882011. View Article : Google Scholar : PubMed/NCBI

15 

Gatza CE, Holtzhausen A, Kirkbride KC, Morton A, Gatza ML, Datto MB and Blobe GC: Type III TGF-β receptor enhances colon cancer cell migration and anchorage-independent growth. Neoplasia. 13:758–770. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Jovanović B, Beeler JS, Pickup MW, Chytil A, Gorska AE, Ashby WJ, Lehmann BD, Zijlstra A, Pietenpol JA and Moses HL: Transforming growth factor beta receptor type III is a tumor promoter in mesenchymal-stem like triple negative breast cancer. Breast Cancer Res. 16:R692014. View Article : Google Scholar

17 

Chu WF, Wu DM, Liu W, Wu LJ, Li DZ, Xu DY and Wang XF: Sulforaphane induces G2-M arrest and apoptosis in high metastasis cell line of salivary gland adenoid cystic carcinoma. Oral Oncol. 45:998–1004. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Vlantis K, Wullaert A, Sasaki Y, Schmidt-Supprian M, Rajewsky K, Roskams T and Pasparakis M: Constitutive IKK2 activation in intestinal epithelial cells induces intestinal tumors in mice. J Clin Invest. 121:2781–2793. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Zhang J and Peng B: In vitro angiogenesis and expression of nuclear factor kappaB and VEGF in high and low metastasis cell lines of salivary gland adenoid cystic carcinoma. BMC Cancer. 7:952007. View Article : Google Scholar : PubMed/NCBI

20 

Lu T, Tian L, Han Y, Vogelbaum M and Stark GR: Dose-dependent cross-talk between the transforming growth factor-beta and interleukin-1 signaling pathways. Proc Natl Acad Sci USA. 104:4365–4370. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Monteleone G, Mann J, Monteleone I, Vavassori P, Bremner R, Fantini M, Del Vecchio Blanco G, Tersigni R, Alessandroni L, Mann D, et al: A failure of transforming growth factor-beta1 negative regulation maintains sustained NF-kappaB activation in gut inflammation. J Biol Chem. 279:3925–3932. 2004. View Article : Google Scholar

22 

You HJ, How T and Blobe GC: The type III transforming growth factor-beta receptor negatively regulates nuclear factor kappa B signaling through its interaction with beta-arrestin2. Carcinogenesis. 30:1281–1287. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Zheng F, He K, Li X, Zhao D, Sun F, Zhang Y, Nie D, Li X, Chu W, Sun Y, et al: Transient overexpression of TGFBR3 induces apoptosis in human nasopharyngeal carcinoma CNE-2Z cells. Biosci Rep. 33:e000292013. View Article : Google Scholar : PubMed/NCBI

24 

Gatza CE, Oh SY and Blobe GC: Roles for the type III TGF-beta receptor in human cancer. Cell Signal. 22:1163–1174. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Zanotto-Filho A, Delgado-Cañedo A, Schröder R, Becker M, Klamt F and Moreira JC: The pharmacological NFkappaB inhibitors BAY117082 and MG132 induce cell arrest and apoptosis in leukemia cells through ROS-mitochondria pathway activation. Cancer Lett. 288:192–203. 2010. View Article : Google Scholar

26 

Yan X, Shen H, Jiang H, Zhang C, Hu D, Wang J and Wu X: External Qi of Yan Xin Qigong induces G2/M arrest and apop tosis of androgen-independent prostate cancer cells by inhibiting Akt and NF-kappa B pathways. Mol Cell Biochem. 310:227–234. 2008. View Article : Google Scholar

27 

Zanotto-Filho A, Braganhol E, Battastini AM and Moreira JC: Proteasome inhibitor MG132 induces selective apoptosis in glioblastoma cells through inhibition of PI3K/Akt and NFkappaB pathways, mitochondrial dysfunction, and activation of p38-JNK1/2 signaling. Invest New Drugs. 30:2252–2262. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Zheng M, Ekmekcioglu S, Walch ET, Tang CH and Grimm EA: Inhibition of nuclear factor-kappaB and nitric oxide by curcumin induces G2/M cell cycle arrest and apoptosis in human melanoma cells. Melanoma Res. 14:165–171. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Chen W, Kirkbride KC, How T, Nelson CD, Mo J, Frederick JP, Wang XF, Lefkowitz RJ and Blobe GC: Beta-arrestin 2 mediates endocytosis of type III TGF-beta receptor and down-regulation of its signaling. Science. 301:1394–1397. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Gao H, Sun Y, Wu Y, Luan B, Wang Y, Qu B and Pei G: Identification of beta-arrestin2 as a G protein-coupled receptor-stimulated regulator of NF-kappaB pathways. Mol Cell. 14:303–317. 2004. View Article : Google Scholar : PubMed/NCBI

31 

Witherow DS, Garrison TR, Miller WE and Lefkowitz RJ: Beta-arrestin inhibits NF-kappaB activity by means of its interaction with the NF-kappaB inhibitor IkappaBalpha. Proc Natl Acad Sci USA. 101:8603–8607. 2004. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2016
Volume 35 Issue 1

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
Xu D, Li D, Lu Z, Dong X and Wang X: Type III TGF-β receptor inhibits cell proliferation and migration in salivary glands adenoid cystic carcinoma by suppressing NF-κB signaling. Oncol Rep 35: 267-274, 2016
APA
Xu, D., Li, D., Lu, Z., Dong, X., & Wang, X. (2016). Type III TGF-β receptor inhibits cell proliferation and migration in salivary glands adenoid cystic carcinoma by suppressing NF-κB signaling. Oncology Reports, 35, 267-274. https://doi.org/10.3892/or.2015.4390
MLA
Xu, D., Li, D., Lu, Z., Dong, X., Wang, X."Type III TGF-β receptor inhibits cell proliferation and migration in salivary glands adenoid cystic carcinoma by suppressing NF-κB signaling". Oncology Reports 35.1 (2016): 267-274.
Chicago
Xu, D., Li, D., Lu, Z., Dong, X., Wang, X."Type III TGF-β receptor inhibits cell proliferation and migration in salivary glands adenoid cystic carcinoma by suppressing NF-κB signaling". Oncology Reports 35, no. 1 (2016): 267-274. https://doi.org/10.3892/or.2015.4390