scholarly journals GATA4 Regulates Inflammation-Driven Pancreatic Ductal Adenocarcinoma Progression

Author(s):  
Weiliang Jiang ◽  
Congying Chen ◽  
Li Huang ◽  
Jie Shen ◽  
Lijuan Yang

Cancer-associated inflammation is a key molecular feature in the progression of pancreatic ductal adenocarcinoma (PDAC). GATA4 is a transcription factor that participates in the regulation and normal development of several endoderm- and mesoderm-derived tissues such as the pancreas. However, it remains unclear whether GATA4 is involved in the inflammation-driven development of pancreatic cancer. Here, we employed quantitative reverse transcription PCR, immunohistochemistry, and differential expression analysis to investigate the association between GATA4 and inflammation-driven PDAC. We found that overexpression of GATA4 in pancreatic tumor tissue was accompanied by increased levels of inflammatory macrophages. We used macrophage-conditioned medium to validate inflammation models following treatment with varying concentrations of lipopolysaccharide and determined whether GATA4-dependent inflammatory stimuli affected pancreatic cancer cell invasion and growth in vitro. Nude mouse models of dibutyltin dichloride-induced chronic pancreatitis with orthotopic tumor xenografts were used to evaluate the effect of the inflammatory microenvironment on GATA4 expression in vivo. Our findings indicate that overexpression of GATA4 dramatically aggravated inflammatory stimuli-induced pancreatic cancer cell invasion and growth via NF-κB and STAT3 signaling, whereas silencing of GATA4 attenuated invasion and growth. Overall, our findings suggest that inflammation-driven cancer progression is dependent on GATA4 expression and is mediated through the STAT3 and NF-κB signaling pathways.

2016 ◽  
Vol 5 (S6) ◽  
pp. S1093-S1097 ◽  
Author(s):  
Rémi Samain ◽  
Christine Jean ◽  
Corinne Bousquet

2001 ◽  
Vol 120 (5) ◽  
pp. A616
Author(s):  
Jiro Okami ◽  
Shoji Nakamori ◽  
Masato Sakon ◽  
Hirofumi Yamamoto ◽  
Nobuaki Hiraok ◽  
...  

2002 ◽  
Vol 122 (2) ◽  
pp. 308-317 ◽  
Author(s):  
Toshiyuki Kusama ◽  
Mutsuko Mukai ◽  
Teruo Iwasaki ◽  
Masaharu Tatsuta ◽  
Yoshirou Matsumoto ◽  
...  

Cancer ◽  
2007 ◽  
Vol 109 (9) ◽  
pp. 1811-1820 ◽  
Author(s):  
Sarah K. Johnson ◽  
Vishnu C. Ramani ◽  
Leah Hennings ◽  
Randy S. Haun

2015 ◽  
Vol 15 (1) ◽  
Author(s):  
Xu Ying ◽  
Li Jing ◽  
Shijie Ma ◽  
Qianjun Li ◽  
Xiaoling Luo ◽  
...  

2020 ◽  
Author(s):  
Brenna A. Rheinheimer ◽  
Lukas Vrba ◽  
Bernard W Futscher ◽  
Ronald L Heimark

AbstractBackgroundSLIT2 has been shown to serve as a tumor suppressor in breast, lung, colon, and liver cancers. Additionally, expression of SLIT2 has been shown to be epigenetically regulated in prostate cancer. Therefore, we sought to determine transcriptional regulation of SLIT2 in pancreatic ductal adenocarcinoma.MethodsRNA expression of SLIT2, SLIT3, and ROBO1 was examined in a panel of pancreatic ductal adenocarcinoma cell lines while protein expression of ROBO1 and SLIT2 was examined in tumor tissue. Methylation of the SLIT2 promoter was determined using Sequenom while histone modifications were queried by chromatin immunoprecipitation. Reexpression of SLIT2 was tested by treatment with 5-aza-2’deoxycytidine and Trichostatin A.ResultsPancreatic cancer cell lines fall into three distinct groups based on SLIT2 and ROBO1 expression. The SLIT2 promoter is methylated in pancreatic ductal adenocarcinoma and SLIT2 expression is dependent on the level of methylation at specific CpG sites. Treatment with 5-aza-2’deoxycytidine (but not Trichostatin A) led to SLIT2 reexpression. The SLIT2 promoter is bivalent in pancreatic ductal adenocarcinoma and histone marks around the transcriptional start site are responsible for transcription.ConclusionsLoss of SLIT2 expression modulated by epigenetic silencing may play a role in pancreatic ductal adenocarcinoma progression.


Sign in / Sign up

Export Citation Format

Share Document