Published in

American Association for Cancer Research, Cancer Research, 13_Supplement(77), p. 2698-2698, 2017

DOI: 10.1158/1538-7445.am2017-2698

Links

Tools

Export citation

Search in Google Scholar

Abstract 2698: IL-17A alone or in combination with IL-4/IFN-γ induces Duox2/DuoxA2 expression and reactive oxygen production in human colon and pancreatic cancer cells

This paper was not found in any repository, but could be made available legally by the author.
This paper was not found in any repository, but could be made available legally by the author.

Full text: Unavailable

Green circle
Preprint: archiving allowed
Orange circle
Postprint: archiving restricted
Red circle
Published version: archiving forbidden
Data provided by SHERPA/RoMEO

Abstract

Abstract NADPH oxidase (NOX)-derived reactive oxygen species (ROS) contribute significantly to inflammation-associated carcinogenesis. Expression of dual oxidase 2 (Duox2), one of seven members of the NOX gene family, is up-regulated in inflammatory bowel disease, chronic pancreatitis, and in many human malignancies including carcinomas of the colon, pancreas, prostate, lung, and breast. Previously, we demonstrated that Stat1 and/or NF-kB play a critical role in modulating the enhanced expression of Duox2, and its cognate maturation factor DuoxA2, by the pro-inflammatory mediators IFN-γ and lipopolysaccharide in human pancreatic cancer cells. This cytokine-mediated increase in Duox2 expression was responsible for a significant increase in tumor cell H2O2 production, DNA damage, and HIF-1α and VEGFA upregulation in pancreatic cancer cell lines. Using the T84 human colon cancer line, and BxPC-3, AsPC-1, and HTB134 pancreatic cancer cell lines, we now report that the pro-inflammatory cytokine IL-17A, pivotal in the adaptive immune response against various pathogens, and in the promotion of colon and pancreatic cancer progression, strongly induces Duox2/DuoxA2 mRNA and protein expression (alone or in combination with IL-4 or IFN-γ). In BxPC-3 cells, IL-17A enhances the expression of Duox2/DuoxA2 in a time- and concentration-dependent manner, which leads to a significant increase in the generation of extracellular H2O2. When IL-17A is combined with IL-4 in either T-84 or AsPc-1 lines, Duox2-mediated ROS formation is sufficient to produce a DNA damage response, indicated by the strong induction of γH2AX expression; moreover, in AsPc-1 cells, Duox2-specific siRNA significantly attenuated cytokine-induced ROS production and γH2AX phosphorylation. IL-17A in combination with IL-4 also significantly increases Stat3 phosphorylation and promotes proliferation in AsPC-1 cells. Ongoing cellular experiments are focused on whether the cytokine-mediated upregulation of Duox2/DuoxA2 is responsible for Stat3 activation and cell growth promotion in the K-ras mutated AsPC-1 cells. Bio-informatic analysis with the TCGA data set, as well as RT-PCR using surgically-resected human colon cancer samples compared to adjacent, uninvolved colonic epithelium revealed that Duox2 was significantly upregulated in human colon cancers. These studies, in concert with our previous data demonstrating high-level Duox protein expression in pancreatitis, pancreatic intra-epithelial neoplasia (PanIN), and frank pancreatic cancer, strongly suggest that ROS, produced as a consequence of cytokine-related Duox2 up-regulation, may be associated with the development of inflammation-related gastrointestinal malignancies. Citation Format: James H. Doroshow, Yongzhong Wu, Jiamo Lu, Smitha Antony, Jennifer Meitzler, Agnes Juhasz, Guojian Jiang, Iris Dahan, Mariam Konate, Krishnendu Roy. IL-17A alone or in combination with IL-4/IFN-γ induces Duox2/DuoxA2 expression and reactive oxygen production in human colon and pancreatic cancer cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2698. doi:10.1158/1538-7445.AM2017-2698