Elsevier

Cellular Signalling

Volume 26, Issue 3, March 2014, Pages 564-569
Cellular Signalling

FOXP3 inhibits NF-κB activity and hence COX2 expression in gastric cancer cells

https://doi.org/10.1016/j.cellsig.2013.11.030Get rights and content

Highlights

  • Expression of COX2 was closely correlated with NF-κB activity.

  • The ratio of FOXP3 and nuclear p65 correlates better with COX2 expression.

  • Interaction between FOXP3 and p65 inhibits NF-κB activation and COX2 expression.

  • FOXP3 inhibits cell migration by repressing NF-κB activity.

Abstract

Gastric cancer remains the main cause of cancer related deaths all over the world, and upregulated COX2 is a key player in its development. The mechanism as to how COX2 is regulated during the gastric cancer development is largely unknown. In this study, we found that the expression of COX2 was closely correlated with NF-κB activity. Strikingly, NF-κB activity was not absolutely consistent with its nuclear localization. Especially, in some cancer cell lines, such as MKN28, there were abundant nuclear localized NF-κB, while NF-κB luciferase activity in this cell line was relatively low. Furthermore, FOXP3 was found to be abundantly expressed in these cells. When the nuclear localized NF-κB expression was adjusted with the expression of FOXP3, it then correlated well with NF-κB activity. Molecularly, increased FOXP3 expression can interact with NF-κB and thus repress its activity. Knockdown of FOXP3 could increase NF-κB activity, COX2 expression, and cell migration. Taken together, our study revealed that function of FOXP3 as a negative regulator of NF-κB activity and thus plays a tumor suppressor role by reducing cell metastasis.

Introduction

Gastric cancer is one of the most common malignant tumors in the world [1]. Previous studies have shown that expression of Cyclooxygenase-2 (COX2) is significantly higher than in normal gastric tissue and plays as a key molecule in the development of gastric cancer [2]. Expression of COX2 has been found to be significantly correlated with lymph node metastasis, poor differentiation and advanced tumor/node/metastasis (TNM) staging. The detection of COX2 is helpful in judging the prognosis of gastric cancer [3]. Among the regulators of COX2 expression, NF-κB, nuclear factor κB, is recognized as the key regulator [4]. NF-κB itself plays an important role in tumor growth and metastasis, and COX2 in turn is the most important downstream mediator [5]. Constitutive activation of NF-κB is a common feature of several tumor cells. Expression of COX2 is up-regulated by activated NF-κB in various kinds of stimuli or other carcinogenic factors, thus inducing apoptosis, proliferation imbalance and other changes, eventually inducing carcinogenesis. Up to now, how COX2 is regulated by NF-κB in gastric cancer and whether there are endogenous co-activators and co-repressors is still largely unknown.

Forkhead box protein 3 (FOXP3) belongs to the forkhead/winged-helix family of transcriptional regulators. While most studies mainly focused on regulatory T cells (Tregs) and their role in immunosuppression [6], [7], there are still some studies indicating that FOXP3 was also expressed in some cancer cells [8]. FOXP3 expression in different tumor cells may have different functions. Most of the studies suggest a tumor suppressor role for FOXP3. Researchers reported that the expression of FOXP3 inhibited the expression of HER-2 [9], SKP2 [10]. It is reported that FOXP3 played a cancer suppressor role in MCF-7 cells by transcriptionally activating p21 expression, suggesting a tumor suppressor role in breast cancer [11]. The interaction between FOXP3 and NF-κB in tumor cells and its functional role in inflammatory microenvironment have also been reported. Foxp3 interacts with NF-κB to repress cytokine gene expression and effector functions of T helper cells [12]. We also report that p65 could interact with FOXP3 and in turn inhibit FOXP3 mediated p21 transcription activation [13], and to some extent dilute the tumor suppressor role of FOXP3. However, how FOXP3 regulates the activity of NF-κB still awaits elucidation. It is especially interesting to see whether how NF-κB and FOXP3 interaction affects NF-κB target gene, such as COX2. Here, we examined the expression of COX2, nuclear localization of NF-κB, NF-κB activity and the expression of FOXP3 in different gastric cancer cell lines. Our study here revealed that expression of COX2, is correlated with NF-κB activity rather than nuclear localization only. Further study revealed that FOXP3 interacts with p65 and in turn inhibits NF-κB activation. FOXP3 and NF-κB expression levels together determine the NF-κB activity and thus COX2 expression. In summary, our study here revealed that FOXP3 functions as a negative regulator of NF-κB activity and thus plays a tumor suppressor role by reducing cell metastasis.

Section snippets

Cell culture

Normal gastric epithelial cell lines (GES-1), and gastric cancer cell lines AGS, MKN45, MKN28 and SGC7901, were cultured in 1640 medium (Invitrogen, Carlsbad, CA), supplemented with 2 mmol/L glutamine, 0.06 g/L penicillin, 0.1 g/L streptomycin, and 10% fetal bovine serum (FBS) at 37 °C in a humidified atmosphere of 5% CO2.

RNA interference and gene transfection

FOXP3 siRNAs (GenePharma, Shanghai, China) were synthesized to knockdown FOXP3 expression in cells. A scrambled siRNA (GenePharma) that targets no known genes was used as a

NF-κB activity correlates with COX2 expression better than solely p65 nuclear localization

First, different gastric cancer cell lines and the normal gastric epithelium were used to study the relationship between COX2 and NF-κB. COX2 expression in both was found upregulated in cancerous cell lines, both at mRNA and protein levels (Fig. 1A and B). COX2 was highly expressed in some low differentiated cell lines. As COX2 is a well-known NF-κB target gene, we next examined nuclear p65 expression in these cell lines. Strikingly, COX2 expression is not closely correlated with p65 nuclear

Discussion

Gastric cancer remains the main cause of cancer deaths worldwide, and the key reason for death was gastric cancer metastasis. Our study here has revealed that COX2 expression in gastric cancer cells are coordinately regulated by FOXP3 and NF-κB. By inhibiting NF-κB activity, which is the key factor for COX2 induction, FOXP3 inhibits the expression of COX2 and hence cell metastasis, thus playing a tumor suppressor role.

COX2 expression was induced to attenuate the degree of atrophic gastritis,

Conclusions

Taken together, our data demonstrates that FOXP3 interacts with p65 and in turn inhibits NF-κB activation. FOXP3 and NF-κB expression levels together determine the NF-κB activity and thus COX2 expression. Our study here reveals that FOXP3 functions as a negative regulator of NF-κB activity and thus plays a tumor suppressor role by reducing cell metastasis. Our study has revealed an instinctive negative regulator of NF-κB, and thus rescue of FOXP3 expression and function might pave the way to

Acknowledgments

This work was supported by grants from the Natural Science Foundation of China (NSFC, Nos. 81001182, 31000406, 30972672, 81171978). We appreciate the valuable comments from the other members of our laboratories.

References (24)

  • J. Chen et al.

    J. Biol. Chem.

    (2005)
  • Q. Hao et al.

    Biochem. Biophys. Res. Commun.

    (2013)
  • C. Rodriguez et al.

    Cell. Signal.

    (2008)
  • J.H. Kim et al.

    Hum. Pathol.

    (2009)
  • G. Helbig et al.

    J. Biol. Chem.

    (2003)
  • R. Siegel et al.

    CA Cancer J. Clin.

    (2012)
  • S.Q. Ji et al.

    Ai Zheng

    (2002)
  • D. Ratnasinghe et al.

    Oncol. Rep.

    (1999)
  • P.A. Konstantinopoulos et al.

    Cell. Oncol.: Off. J. Int. Soc. Cell. Oncol.

    (2007)
  • J.D. Fontenot et al.

    Nat. Immunol.

    (2005)
  • S. Sakaguchi

    Nat. Immunol.

    (2005)
  • V. Karanikas et al.

    J. Transl. Med.

    (2008)
  • Cited by (35)

    • Lipopolysaccharide-induced splenic ferroptosis in goslings was alleviated by polysaccharide of atractylodes macrocephala koidz associated with proinflammatory factors

      2022, Poultry Science
      Citation Excerpt :

      NADPH oxidase (NOX), cyclooxygenase (COX) and LOX constantly generate free radicals such as ROS and NO, which, in addition to being used to kill pathogenic microorganisms, can activate NF-κB to induce the expression of proinflammatory cytokine genes in neighboring cells (Lu et al., 2017). In turn, NF-κB regulates the expression of COX-2 and 5-LOX (Hao et al., 2014), the proinflammatory cytokines TNF-α and IL-1β are stimulators of NOX, and ROS production triggers a positive feedback loop that contributes to the onset and persistence of inflammation (Liang et al., 2019). It can be seen that the elevated iron content and the accumulation of ROS in spleen are likely due to changes in the expression of cytokines as well as ferroptosis-related genes, which in turn affect the expression of inflammatory cytokines and ferroptosis-related genes.

    • Epstein–Barr virus is a promoter of lymphoma cell metastasis

      2020, Pathology
      Citation Excerpt :

      In addition, NF-κB is transferred to the nucleus to play the role of a transcription factor.27 In addition, CTAR2 indirectly binds with TRAF2 through the action of tumour necrosis factor associated dead domain (TRADD) and activates NF-κB, while NF-κB further regulates the expression of many genes that contain κB and adhesion molecules, in order to upregulate anti-apoptotic gene products, such as Bcl-2 and A20, and upregulates cytokines such as IL-6 and IL-8, cell surface antigens such as CD54 (ICAM-1) and CD40, and angiogenesis factors such as COX2 and vascular endothelial growth factor (VEGF).28 Furthermore, this also regulates downstream genes, such as epidermal growth factor receptor (EGFR) and p53.29

    • Members of FOX family could be drug targets of cancers

      2018, Pharmacology and Therapeutics
      Citation Excerpt :

      Demethylation of FOXP3 promoter reduced immune responses and promotes tumor progression in NSCLC (Ke et al., 2016). FOXP3 reduced cell metastasis by inhibiting NF-κB activity (Hao et al., 2014). FOXP4 regulates growth of tumor and independently associates with miR-138 in NSCLC cells (Yang et al., 2015).

    • PIM kinases: From survival factors to regulators of cell motility

      2017, International Journal of Biochemistry and Cell Biology
    • Targeted superparamagnetic nanoparticles coated with 2-deoxy-D-gloucose and doxorubicin more sensitize breast cancer cells to ionizing radiation

      2017, Breast
      Citation Excerpt :

      To decrease side effects of the related single modality cancer treatments, increased cytotoxicity and more localization on tumor cells by effective drug delivery we used combined chemo-radiotherapy with the synthesized SPmHA nanocomposites because of the excellent properties of SPmHA such as biocompatibility, bioactivity, non-mutagenicity, non-toxicity, diverse morphologies and surface properties. Nanometer pores in the structure of this platform and SPIONs, as a core of mesoporous HA, can make it much more applicable for targeted therapy and may have the potential for targeted drug delivery and hyperthermia [42–47]. To put it another way, there are few published reports on the synthesis of mesoporous HA as a shell for SPIONs cores for targeted drug delivery.

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text