4.7 Article

Paracrine activin B-NF-κB signaling shapes an inflammatory tumor microenvironment in gastric cancer via fibroblast reprogramming

出版社

BMC
DOI: 10.1186/s13046-023-02861-4

关键词

Gastric cancer; Activin B; Fibroblast reprogramming; NF-kappa B; Tumor microenvironment

类别

向作者/读者索取更多资源

Our study revealed that INHBB is significantly upregulated in gastric cancer (GC) and its increased expression is associated with poor survival. INHBB promotes the proliferation, migration, and invasion of GC cells in vitro. Additionally, activin B reprograms fibroblasts into cancer-associated fibroblasts (CAFs) and promotes GC development. The high expression of INHBB in GC cells activates the NF-kappa B pathway in normal gastric fibroblasts via the secretion of activin B, leading to fibroblast proliferation, migration, and invasion. Activin B induces NF-kappa B pathway activation in fibroblasts through controlling TRAF6 autoubiquitination and TAK1 phosphorylation. Activated fibroblasts release pro-inflammatory factors IL-1 beta, which induce p65 phosphorylation in GC cells. p65 directly binds to the INHBB promoter, increasing INHBB transcription in GC cells and establishing a positive regulatory feedback loop that promotes GC progression.
Background Important roles of INHBB in various malignancies are increasingly identified. The underlying mechanisms in gastric cancer (GC) microenvironment are still greatly unexplored.Methods The clinical significance of INHBB and the correlation between INHBB and p-p65 in GC were assessed through analyzing publicly available databases and human paraffin embedded GC tissues. The biological crosstalk of INHBB between GC cells and fibroblasts was explored both in vitro and in vivo. RNA-seq analyses were performed to determine the mechanisms which regulating fibroblasts reprogramming. Luciferase reporter assay and chromatin immunoprecipitation (CHIP) assay were used to verify the binding relationship of p65 and INHBB in GC cells.Results Our study showed that INHBB level was significantly higher in GC, and that increased INHBB was associated with poor survival. INHBB positively regulates the proliferation, migration, and invasion of GC cells in vitro. Also, activin B promotes the occurrence of GC by reprogramming fibroblasts into cancer-associated fibroblasts (CAFs). The high expression of INHBB in GC cells activates the NF-kappa B pathway of normal gastric fibroblasts by secreting activin B, and promotes fibroblasts proliferation, migration, and invasion. In addition, activin B activates NF-kappa B pathway by controlling TRAF6 autoubiquitination to induce TAK1 phosphorylation in fibroblasts. Fibroblasts activated by activin B can induce the activation of p65 phosphorylation of GC cells by releasing pro-inflammatory factors IL-1 beta. p65 can directly bind to the INHBB promoter and increase the INHBB transcription of GC cells, thus establishing a positive regulatory feedback loop to promote the progression of GC.Conclusions GC cells p65/INHBB/activin B and fibroblasts p65/IL-1 beta signal loop led to the formation of a whole tumor-promoting inflammatory microenvironment, which might be a promising therapeutic target for GC.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据