Skip to main content
Top
Published in: Tumor Biology 6/2016

01-06-2016 | Original Article

Overexpression of the transcription factor FOXP3 in lung adenocarcinoma sustains malignant character by promoting G1/S transition gene CCND1

Authors: Yinan Li, Dong Li, Wei Yang, Haiying Fu, Yaqing Liu, Yi Li

Published in: Tumor Biology | Issue 6/2016

Login to get access

Abstract

The Forkhead box P3 (FOXP3) transcription factor is the key driver of the differentiation and immunosuppressive function of regulatory T cells (Tregs). Additionally, FOXP3 has been reported to be expressed in many solid tumor cell lines and tissues. However, its role in tumorigenesis and tumor progression is conflicting, both tumor suppressive and promoting functions have been described. In this study, we demonstrated that FOXP3 was expressed in both lung adenocarcinoma tissues and the lung adenocarcinoma cell line A549. FOXP3 inhibition decreased cell proliferation, migration, and invasion as well as the secretion of inhibitory cytokines (e.g., transforming growth factor beta 1 (TGF-β1), interleukin 35 (IL-35), and heme oxygenase-1 (HMOX1)), suggesting a positive role for FOXP3 in tumor development. Importantly, we found that FOXP3 could enhance lung adenocarcinoma cell proliferation via upregulating the levels of the cell cycle G1/S checkpoint gene CCND1. These data demonstrated that FOXP3 could be regarded as a novel therapeutic target for inhibiting lung adenocarcinoma progression.
Literature
1.
go back to reference Brunkow ME, Jeffery EW, Hjerrild KA, et al. Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat Genet. 2001;27:68–73.CrossRefPubMed Brunkow ME, Jeffery EW, Hjerrild KA, et al. Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat Genet. 2001;27:68–73.CrossRefPubMed
2.
go back to reference Bennett CL, Christie J, Ramsdell F, et al. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet. 2001;27:20–1.CrossRefPubMed Bennett CL, Christie J, Ramsdell F, et al. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet. 2001;27:20–1.CrossRefPubMed
3.
go back to reference Schneider T, Kimpfler S, Warth A, et al. FOXP3+ regulatory T cells and naturalkiller cells distinctly infiltrate primary tumors and draininglymph nodes in pulmonary adenocarcinoma. J Thorac Oncol. 2011;6:432–8.CrossRefPubMed Schneider T, Kimpfler S, Warth A, et al. FOXP3+ regulatory T cells and naturalkiller cells distinctly infiltrate primary tumors and draininglymph nodes in pulmonary adenocarcinoma. J Thorac Oncol. 2011;6:432–8.CrossRefPubMed
4.
go back to reference Shimizu K, Nakata M, Hirami Y, et al. Tumor-infiltrating FOXP3+ regulatory T cells are correlated with cyclooxygenase-2 expression and are associated with recurrence in resected non-small cell lung cancer. J Thora Oncol. 2010;5:585–90.CrossRef Shimizu K, Nakata M, Hirami Y, et al. Tumor-infiltrating FOXP3+ regulatory T cells are correlated with cyclooxygenase-2 expression and are associated with recurrence in resected non-small cell lung cancer. J Thora Oncol. 2010;5:585–90.CrossRef
6.
go back to reference Hinz S, Pagerols-Raluy L, Oberg HH, et al. Foxp3 expression in pancreatic carcinoma cells as a noveral mechanism of immune evasion in cancer. Cancer Res. 2007;67:8344–50.CrossRefPubMed Hinz S, Pagerols-Raluy L, Oberg HH, et al. Foxp3 expression in pancreatic carcinoma cells as a noveral mechanism of immune evasion in cancer. Cancer Res. 2007;67:8344–50.CrossRefPubMed
7.
go back to reference Triulzi T. FOXP3 expression in tumor cells and implications for cancer progression.Taqliabue E, Balsari A, et al. J Cell Physiol. 2013;1:30–5.CrossRef Triulzi T. FOXP3 expression in tumor cells and implications for cancer progression.Taqliabue E, Balsari A, et al. J Cell Physiol. 2013;1:30–5.CrossRef
8.
go back to reference Redpath M, Xu B, van Kempen LC, et al. The dual role of the X-liked FoxP3 gene in human cancers. Mol Oncol. 2011;2:156–63.CrossRef Redpath M, Xu B, van Kempen LC, et al. The dual role of the X-liked FoxP3 gene in human cancers. Mol Oncol. 2011;2:156–63.CrossRef
9.
go back to reference Jia T, Fu HY, Sun JT, et al. Foxp3 expression in A549 cells is regulated by TLR4 through NF-κB. Mol Med Rep. 2012;6:167–72.PubMed Jia T, Fu HY, Sun JT, et al. Foxp3 expression in A549 cells is regulated by TLR4 through NF-κB. Mol Med Rep. 2012;6:167–72.PubMed
10.
go back to reference Milkova L, Voelcker V, Forstreuter I, et al. The NF-kappaB signaling pathway is involved un the LPS/IL-2-induced upregulation of Foxp3 expression in human CD4+CD25high regulatory T cells. Exp Med. 2003;4:403–11. Milkova L, Voelcker V, Forstreuter I, et al. The NF-kappaB signaling pathway is involved un the LPS/IL-2-induced upregulation of Foxp3 expression in human CD4+CD25high regulatory T cells. Exp Med. 2003;4:403–11.
11.
go back to reference Li B, Samanta A, Song X, et al. FOXP3 ensembles in T-cell regulation. Immunol Rev. 2006;212:99–113.CrossRefPubMed Li B, Samanta A, Song X, et al. FOXP3 ensembles in T-cell regulation. Immunol Rev. 2006;212:99–113.CrossRefPubMed
12.
14.
go back to reference Niu J, Jiang C, Li C, et al. Foxp3 expression in melanoma cells as a possible mechanism of resistance to immune destruction. Cancer Immunol Immunother. 2011;8:1109–18.CrossRef Niu J, Jiang C, Li C, et al. Foxp3 expression in melanoma cells as a possible mechanism of resistance to immune destruction. Cancer Immunol Immunother. 2011;8:1109–18.CrossRef
15.
go back to reference Ma C, Peng C, Lu X, et al. Downregulation of FOXP3 inhibits invasion and immune escape in cholangiocarcinoma. Biochem Biophys Res Commun. 2015;2:234–9.CrossRef Ma C, Peng C, Lu X, et al. Downregulation of FOXP3 inhibits invasion and immune escape in cholangiocarcinoma. Biochem Biophys Res Commun. 2015;2:234–9.CrossRef
16.
go back to reference Chen YH, Zhang HH, He GX, et al. Expression of FOXP3 in hepatoma cells can directly promote tumor metastasis in vitro and in vivo. Hepatology. 2012;794A. Chen YH, Zhang HH, He GX, et al. Expression of FOXP3 in hepatoma cells can directly promote tumor metastasis in vitro and in vivo. Hepatology. 2012;794A.
17.
go back to reference Wang G, Liu G, Li X, et al. FOXP3 expression in esophageal cancer cells is caaosiated with poor prognosis in esophageal cancer. Hepatogastroenterology. 2012;119:2186–91. Wang G, Liu G, Li X, et al. FOXP3 expression in esophageal cancer cells is caaosiated with poor prognosis in esophageal cancer. Hepatogastroenterology. 2012;119:2186–91.
18.
go back to reference Zuo T, Liu R, Zhang H, et al. FOXP3 is a novel transcriptional repressor for the breast cancer oncogene SKP2. J Clinl Invest. 2007;117:3765–73. Zuo T, Liu R, Zhang H, et al. FOXP3 is a novel transcriptional repressor for the breast cancer oncogene SKP2. J Clinl Invest. 2007;117:3765–73.
19.
go back to reference Zuo T, Wang L, Morrison C, et al. FOXP3 is an X-linked breast cancer suppressor gene and an important repressor of the HER-2/ErbB2 oncogene. Cell. 2007;129:1275–86.CrossRefPubMedPubMedCentral Zuo T, Wang L, Morrison C, et al. FOXP3 is an X-linked breast cancer suppressor gene and an important repressor of the HER-2/ErbB2 oncogene. Cell. 2007;129:1275–86.CrossRefPubMedPubMedCentral
20.
go back to reference Ladoire S, Arnould L, Mignot G, et al. Presence of Foxp3 expression in tumor cells predicts better survival in HER2-overexpressing breast cancer patients treated with neoadjuvant chemotherapy. Breast Cancer Res Treat. 2011;125:65–72.CrossRefPubMed Ladoire S, Arnould L, Mignot G, et al. Presence of Foxp3 expression in tumor cells predicts better survival in HER2-overexpressing breast cancer patients treated with neoadjuvant chemotherapy. Breast Cancer Res Treat. 2011;125:65–72.CrossRefPubMed
21.
go back to reference Douqlass S, Meeson AP, Overbeck-Zubrzycka D, et al. Breast cancer metastasis: demonsrtation that FOXP3 regulates CXCR4 expression and the response to CXCL12. J Pathol. 2014;1:74–85.CrossRef Douqlass S, Meeson AP, Overbeck-Zubrzycka D, et al. Breast cancer metastasis: demonsrtation that FOXP3 regulates CXCR4 expression and the response to CXCL12. J Pathol. 2014;1:74–85.CrossRef
22.
23.
go back to reference Li W, Wang L, Katoh H, et al. Identification of a tumor suppressor relay between the FOXP3 and the Hippo pathways in breast and prostate cancers. Cancer Res. 2011;71:2162–71.CrossRefPubMedPubMedCentral Li W, Wang L, Katoh H, et al. Identification of a tumor suppressor relay between the FOXP3 and the Hippo pathways in breast and prostate cancers. Cancer Res. 2011;71:2162–71.CrossRefPubMedPubMedCentral
24.
go back to reference Zhang HY, Sun H. Up-regulation of Foxp3 inhibits cell proliferation, migration and invasion in epithelial ovarian cancer. Cancer Lett. 2010;287:91–7.CrossRefPubMed Zhang HY, Sun H. Up-regulation of Foxp3 inhibits cell proliferation, migration and invasion in epithelial ovarian cancer. Cancer Lett. 2010;287:91–7.CrossRefPubMed
25.
go back to reference Lopes JE, Torqerson TR, Schubert LA, et al. Analysis of FOXP3 reveals multiple domains required for its functions as a transcriptional repressor. J Immunol. 2006;5:3133–42.CrossRef Lopes JE, Torqerson TR, Schubert LA, et al. Analysis of FOXP3 reveals multiple domains required for its functions as a transcriptional repressor. J Immunol. 2006;5:3133–42.CrossRef
26.
go back to reference Chen C, Rowell EA, Thomas RM, et al. Transcriptional regulation by Foxp3 is associated with direct promoter occupancy and modulation of histone acetylation. J Biol Chem. 2006;281:36828–34.CrossRefPubMed Chen C, Rowell EA, Thomas RM, et al. Transcriptional regulation by Foxp3 is associated with direct promoter occupancy and modulation of histone acetylation. J Biol Chem. 2006;281:36828–34.CrossRefPubMed
27.
go back to reference Du JG, Huang CJ, Zhou BH, et al. Isoform specific inhibition of ror mediated transcriptional activation human foxp3. J Immunol. 2008;180:4785–92.CrossRefPubMed Du JG, Huang CJ, Zhou BH, et al. Isoform specific inhibition of ror mediated transcriptional activation human foxp3. J Immunol. 2008;180:4785–92.CrossRefPubMed
28.
go back to reference Harada K, Shimoda S, Kimura Y, et al. Significance of immunoglobulin G4 (IgG4)-positive cells in extrahepatic cholangiocarcinoma: molecular mechanism of IgG4 reaction in cancer tissue. Hepatology. 2012;1:157–64.CrossRef Harada K, Shimoda S, Kimura Y, et al. Significance of immunoglobulin G4 (IgG4)-positive cells in extrahepatic cholangiocarcinoma: molecular mechanism of IgG4 reaction in cancer tissue. Hepatology. 2012;1:157–64.CrossRef
29.
go back to reference Ebert LM, Tan BS, Browning J, et al. The regulatory T cell-associated transcription factor FoxP3 is expressed by tumor cells. Cancer Res. 2008;8:3001–9.CrossRef Ebert LM, Tan BS, Browning J, et al. The regulatory T cell-associated transcription factor FoxP3 is expressed by tumor cells. Cancer Res. 2008;8:3001–9.CrossRef
30.
go back to reference Sathe A, Koshy N, Schmid SC, et al. CDK4/6-inhibition controls proliferation of bladder cancer and transcription of RB1. J Urol. 2015;15:04685–6. Sathe A, Koshy N, Schmid SC, et al. CDK4/6-inhibition controls proliferation of bladder cancer and transcription of RB1. J Urol. 2015;15:04685–6.
31.
Metadata
Title
Overexpression of the transcription factor FOXP3 in lung adenocarcinoma sustains malignant character by promoting G1/S transition gene CCND1
Authors
Yinan Li
Dong Li
Wei Yang
Haiying Fu
Yaqing Liu
Yi Li
Publication date
01-06-2016
Publisher
Springer Netherlands
Published in
Tumor Biology / Issue 6/2016
Print ISSN: 1010-4283
Electronic ISSN: 1423-0380
DOI
https://doi.org/10.1007/s13277-015-4616-3

Other articles of this Issue 6/2016

Tumor Biology 6/2016 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine