American Association for Cancer Research
Browse
15357163mct150309-sup-148100_2_supp_3209077_nx2zg9.png (66.69 kB)

Supplementary Table from GSK-3β Governs Inflammation-Induced NFATc2 Signaling Hubs to Promote Pancreatic Cancer Progression

Download (66.69 kB)
figure
posted on 2023-04-03, 14:46 authored by Sandra Baumgart, Nai-Ming Chen, Jin-San Zhang, Daniel D. Billadeau, Irina N. Gaisina, Alan P. Kozikowski, Shiv K. Singh, Daniel Fink, Philipp Ströbel, Caroline Klindt, Lizhi Zhang, William R. Bamlet, Alexander Koenig, Elisabeth Hessmann, Thomas M. Gress, Volker Ellenrieder, Albrecht Neesse

Tissue microarray analysis of NFATc2 and Stat3

Funding

NCI

Deutsche Krebshilfe

National Natural Science Foundation of China

History

ARTICLE ABSTRACT

We aimed to investigate the mechanistic, functional, and therapeutic role of glycogen synthase kinase 3β (GSK-3β) in the regulation and activation of the proinflammatory oncogenic transcription factor nuclear factor of activated T cells (NFATc2) in pancreatic cancer. IHC, qPCR, immunoblotting, immunofluorescence microscopy, and proliferation assays were used to analyze mouse and human tissues and cell lines. Protein–protein interactions and promoter regulation were analyzed by coimmunoprecipitation, DNA pulldown, reporter, and ChIP assays. Preclinical assays were performed using a variety of pancreatic cancer cells lines, xenografts, and a genetically engineered mouse model (GEMM). GSK-3β–dependent SP2 phosphorylation mediates NFATc2 protein stability in the nucleus of pancreatic cancer cells stimulating pancreatic cancer growth. In addition to protein stabilization, GSK-3β also maintains NFATc2 activation through a distinct mechanism involving stabilization of NFATc2–STAT3 complexes independent of SP2 phosphorylation. For NFATc2–STAT3 complex formation, GSK-3β–mediated phosphorylation of STAT3 at Y705 is required to stimulate euchromatin formation of NFAT target promoters, such as cyclin-dependent kinase-6, which promotes tumor growth. Finally, preclinical experiments suggest that targeting the NFATc2–STAT3–GSK-3β module inhibits proliferation and tumor growth and interferes with inflammation-induced pancreatic cancer progression in KrasG12D mice. In conclusion, we describe a novel mechanism by which GSK-3β fine-tunes NFATc2 and STAT3 transcriptional networks to integrate upstream signaling events that govern pancreatic cancer progression and growth. Furthermore, the therapeutic potential of GSK-3β is demonstrated for the first time in a relevant Kras and inflammation-induced GEMM for pancreatic cancer. Mol Cancer Ther; 15(3); 491–502. ©2016 AACR.