Suppression of NF-κB/p65 Inhibits the Proliferation in Oral Squamous Cancer Cells

Abstract

Object: Hypoxia occurs when oxygen tension drops below normal limits, and malignant tumors often experience hypoxia, which activates the expression of genes through oxygen-sensitive transcription factors, including the hypoxiainducible factor (HIF) and the nuclear factor-κB (NF-κB). NF-κB pathway represents an attractive therapeutic target in both cancer cells and ischemic cells, which involves immune response. To investigate the expression of NF-κB target genes in oral squamous cancer carcinoma (OSCC) under hypoxia, we performed cDNA plate array analyses of 23 NF-κB-regulated genes in different cell lines. Our aim was to clarify the functions of NF-κB in OSCC under hypoxia. Results: We conducted an NF-κB reporter assay to examine NF-κB activation under hypoxia. This luciferase-based reporter assay showed that hypoxia induced NF-κB activation after 24 h of hypoxia. We also found that vascular endothelial growth factor-C (VEGF-C) of NF-κB-regulated genes was upregulated in both cell lines under hypoxia. We then tested the influence of NF-κB/p65 knockdown on these cells and NF-κB/p65 knockdown inhibited the proliferation of OSCC cells by colony assay. Conclusion: Based on these findings, we postulate that one mechanism by which hypoxic OSCC cells may involve NF-κB-mediated upregulation. Our results also indicate that the knockdown of NF-κB/p65 subunit lead to growth inhibition during hypoxia in OSCC cells. These results suggest that knockdown of NF-κB/p65 subunit could be a potential therapeutic option for patients with OSCC.

Share and Cite:

N. Anbo, K. Ogi, Y. Sogabe, M. Shimanishi, T. Kaneko, H. Dehari, A. Miyazaki and H. Hiratsuka, "Suppression of NF-κB/p65 Inhibits the Proliferation in Oral Squamous Cancer Cells," Journal of Cancer Therapy, Vol. 4 No. 4, 2013, pp. 891-897. doi: 10.4236/jct.2013.44100.

Conflicts of Interest

The authors declare no conflicts of interest.

References

[1] J. Sudbo and A. Reith, “The Evolution of Predictive Oncology and Molecular-Based Therapy for Oral Cancer Prevention,” International Journal of Cancer, Vol. 115, No. 3, 2005, pp. 339-345. doi:10.1002/ijc.20896
[2] R. T. Greenlee, M. B. Hill-Harmon, T. Murray, et al., “Cancer Statistics,” A Cancer Journal for Clinicians, Vol. 5, No. 1, 2001, pp. 15-36.
[3] M. Shimanishi, K. Ogi, T. Kaneko, et al., “Silencing of GLUT-1 Inhibits Sensitization of Oral Cancer Cells to Cisplatin during Hypoxia,” Journal of Oral Pathology and Medicine, Vol. 42, No. 5, 2013, pp. 382-388. doi:10.1111/jop.12028
[4] A. L. Harris, “Hypoxia—A Key Regulatory Factor in Tumor Growth,” Nature Reviews Cancer, Vol. 2, No. 1, 2002, pp. 38-47. doi:10.1038/nrc704
[5] G. L. Semenza, “Targeting Hif-1 Alpha for Cancer Therapy,” Nature Reviews Cancer, Vol. 10, No. 3, 2003, pp. 721-732. doi:10.1038/nrc1187
[6] J. Yang, L. Zhang, P. J. Erbel, et al., “Functions of the Per/ARNT/Sim Domains of the Hypoxia-Inducible Factor,” The Journal of Biological Chemistry, Vol. 280, No. 43, 2005, pp. 36047-36054. doi:10.1074/jbc.M501755200
[7] G. L. Semenza, “HIF-1: Mediator of Physiological and Pathophysiological Responses to Hypoxia,” Journal of Applied Physiology, Vol. 88, No. 4, 2000, pp. 1474-1480.
[8] E. P. Cummins and C. T. Taylor, “Hypoxia-Responsive Transcription Factors,” Pflügers Archiv—European Journal of Physiology, Vol. 450, No. 6, 2005, pp. 363-371.
[9] K. R. Laderoute, “The Interaction between HIF-1 and AP-1 Transcription Factors in Response to Low Oxygen,” Seminars in Cell & Developmental Biology, Vol. 16, No. 4-5, 2005, pp. 502-513. doi:10.1016/j.semcdb.2005.03.005
[10] T. Tanaka, H. Nakayama, Y. Yoshitake, et al, “Selective Inhibition of Nuclear Factor-κB by Nuclear Factor-κB Essential Modulator-Binding Domain Peptide Suppresses the Metastasis of Highly Metastatic Oral Squamous Cell Carcinoma,” Japanese Journal of Cancer Research, Vol. 103, No. 3, 2012, pp. 455-463. doi:10.1111/j.1349-7006.2011.02174.x
[11] A. W. Ken, S. M. Karen, H. Jason, et al., “Hypoxia-Inducible Factor and Nuclear Factor Kappa-B Activation in Blood-Brain Barrier Endothelium under Hypoxic/Reoxygenation Stress,” Journal of Neurochemistry, Vol. 92, No. 1, 2005, pp. 203-214. doi:10.1111/j.1471-4159.2004.02871.x
[12] S. Y. Nam, Y S. Ko, J. Jung, et al., “A Hypoxia-Dependent Upregulation of Hypoxia-Inducible Factor-1 by Nuclear Factor-κB Promotes Gastric Tumour Growth and Angiogenesis,” British Journal of Cancer, Vol. 104, No. 1, 2011, pp. 166-174. doi:10.1038/sj.bjc.6606020
[13] A. C. Koong, E. Y. Chen and A. J. Giaccia, “Hypoxia Causes the Activation of Nuclear Factor Kappa B through the Phosphorylation of I Kappa B Alpha on Tyrosine Residues,” Cancer Research, Vol. 54, No. 6, 1994, pp. 1425-1414.
[14] J. F. Schmedtje Jr, Y. S. Ji, R. N. DuBois, et al., “Hypoxia Induces Cyclooxygenase-2 via the NF-KappaB p65 Transcription Factor in Human Vascular Endothelial Cells,” The Journal of Biological Chemistry, Vol. 272, No. 1, 1997, pp. 601-608. doi:10.1074/jbc.272.1.601
[15] C. T. Taylor, A. L. Dzus and S. P. Colgan, “Autocrine Regulation of Epithelial Permeability by Hypoxia: Role for Polarized Release of Tumor Necrosis Factor Alpha,” Gastroenterology, Vol. 114, No. 4, 1998, pp. 657-668. doi:10.1016/S0016-5085(98)70579-7
[16] H. Matsui, Y. Ihara, Y Fujio, et al., “Induction of Interleukin (IL)-6 by Hypoxia Is Mediated by Nuclear Factor (NF)-Kappa B and NF-IL6 in Cardiac Myocytes,” Cardiovascular Research, Vol. 42, No. 1, 1999, pp. 104-112. doi:10.1016/S0008-6363(98)00285-5
[17] A. Zampetaki, S. A. Mitsialis, J. Pfeilschifter, et al., “Hypoxia Induces Macrophage Inflammatory Protein-2 (MIP-2) Gene Expression in Murine Macrophages via NF kappaB: The Prominent Role of p42/p44 and PI3 Kinase Pathways,” Official Publication of the Federation of American Societies for Experimental Biology, Vol. 18, No. 10, 2004, pp. 1090-1092.
[18] E. P. Cummins, E. Berra, K. M. Comerford, et al., “Prolyl Hydroxylase-1 Negatively Regulates IkB Kinase-Beta Giving Insight into Hypoxia-Induced NFkB Activity,” Proceedings of the National Academy of Sciences of the United States of America, Vol. 103, No. 48, 2006, pp. 18154-18159. doi:10.1073/pnas.0602235103
[19] J. Rius, M. Guma, C. Schachtrup, et al., “NF-KappaB Links Innate Immunity to the Hypoxic Response through Transcriptional Regulation of HIF-1alpha,” Nature, Vol. 453, No. 7196, 2008, pp. 807-811. doi:10.1038/nature06905
[20] R. S. Belaiba, S. Bonello, C. Zahringer, et al., “Hypoxia Up-Regulates Hypoxia-Inducible Factor-1alpha Transcription by Involving Phosphatidylinositol 3-Kinase and Nuclear Factor KappaB in Pulmonary Artery Smooth Muscle Cells,” Molecular Biology of the Cell, Vol. 18, No. 12, 2007, pp. 4691-4697. doi:10.1091/mbc.E07-04-0391
[21] P. van Uden, N. S. Kenneth, S. Rocha, et al., “Regulation of Hypoxia-Inducible Factor-1alpha by NF-KappaB,” The Biochemical Journal, Vol. 412, No. 3, 2008, pp. 477-484. doi:10.1042/BJ20080476
[22] S. R. Walmsley, C. Print, N. Farahi, et al., “HypoxiaInduced Neutrophil Survival Is Mediated by HIF-1alphaDependent NF-KappaB Activity,” The Journal of Experimental Medicine, Vol. 201, No. 1, 2005, pp. 105-115. doi:10.1084/jem.20040624
[23] C. T. Taylor, “Interdependent Roles for Hypoxia Inducible Factor and Nuclear Factor-KappaB in Hypoxic Inflammation,” The Journal of Physiology, Vol. 586, No. 187, 2008, pp. 4055-4059. doi:10.1113/jphysiol.2008.157669
[24] D. B. Mendonca, G. Mendonca, F. J. Aragao, et al., “NF-κB Suppresses HIF-1α Response by Competing for P300 Binding,” Biochemical and Biophysical Research Communications, Vol. 404, No. 4, 2011, pp. 997-1003. doi:10.1016/j.bbrc.2010.12.098
[25] C. Huang, Z. Sun, Y. Sun, et al., “Association of Increased Ligand Cyclophilin A and Receptor CD147 with Hypoxia, Angiogenesis, Metastasis and Prognosis of Tongue Squamous Cell Carcinoma,” Histopathology, Vol. 60, No. 5, 2012, pp. 793-803. doi:10.1111/j.1365-2559.2011.04130.x
[26] H. Kubo, R. Cao, E. Brakenhielm, et al., “Blockade of Vascular Endothelial Growth Factor Receptor-3 Signaling Inhibits Fibroblast Growth Factor-2-Induced Lymphangiogenesis in Mouse Cornea,” Proceedings of the National Academy of Sciences of the United States of America, Vol. 99, No. 13, 2002, pp. 8868-8873. doi:10.1073/pnas.062040199
[27] S. E. S. Faustino, D. T. Oliveira, S. Nonogaki, et al., “Expression of Vascular Endothelial Growth Factor-C Does Not Predict Occult Lymph-Node Metastasis in Early Oral Squamous Cell Carcinoma,” International Journal of Oral and Maxillofacial Surgery, Vol. 37, No. 4, 2008, pp. 372-378. doi:10.1016/j.ijom.2007.11.021
[28] T. Naruse, G. Kawasaki, S. Yanamoto, et al., “Immunohistochemical Study of VEGF Expression in Oral Squamous Cell Carcinomas: Correlation with the mTOR-HIF-1alpha Pathway,” Anticancer Research, Vol. 31, No. 12, 2011, pp. 4429-4437.
[29] S. Shintani, C. Li, T. Ishikawa, et al., “Expression of Vascular Endothelial Growth Factor A, B, C, and D in Oral Squamous Cell Carcinoma,” Oral Oncology, Vol. 40, No. 1, 2004, pp. 13-20. doi:10.1016/S1368-8375(03)00127-1
[30] B. S. Siriwardena, Y. Kudo, I. Ogawa, et al., “VEGF-C Is Associated with Lymphatic Status and Invasion in Oral Cancer,” Journal of Clinical Pathology, Vol. 61, No. 1, 2008, pp. 103-108. doi:10.1136/jcp.2007.047662
[31] E. Sasabe, X. Zhou, D. Li, et al., “ The Involvement of Hypoxia-Inducible Factor-1alpha in the Susceptibility to Gamma-Rays and Chemotherapeutic Drugs of Oral Squamous Cell Carcinoma Cells,” International Journal of Cancer, Vol. 120, No. 2, 2007, pp. 268-277. doi:10.1002/ijc.22294

Copyright © 2024 by authors and Scientific Research Publishing Inc.

Creative Commons License

This work and the related PDF file are licensed under a Creative Commons Attribution 4.0 International License.