4.7 Article

A novel STAT3/ NFκB p50 axis regulates stromal-KDM2A to promote M2 macrophage-mediated chemoresistance in breast cancer

Journal

CANCER CELL INTERNATIONAL
Volume 23, Issue 1, Pages -

Publisher

BMC
DOI: 10.1186/s12935-023-03088-1

Keywords

Lysine demethylase 2A; Cancer-associated fibroblasts; Tumor-associated macrophage; Paclitaxel resistance

Categories

Ask authors/readers for more resources

This study revealed a non-canonical molecular mechanism of IL-6 upregulating KDM2A expression in CAFs via a novel STAT3/NF kappa B p50 axis. KDM2A-expressing CAFs predominantly secreted CXCR2-associated chemokines to promote M2 macrophage polarization and enhance paclitaxel resistance in breast cancer, suggesting the therapeutic potential of targeting the CXCR2 or CCR2 pathway for paclitaxel-resistant breast cancer.
BackgroundLysine Demethylase 2A (KDM2A) plays a crucial role in cancer cell growth, differentiation, metastasis, and the maintenance of cancer stemness. Our previous study found that cancer-secreted IL-6 can upregulate the expression of KDM2A to promote further the transition of cells into cancer-associated fibroblasts (CAFs). However, the molecular mechanism by which breast cancer-secreted IL-6 regulates the expression of KDM2A remains unclear. Therefore, this study aimed to elucidate the underlying molecular mechanism of IL-6 in regulating KDM2A expression in CAFs and KDM2A-mediated paclitaxel resistance in breast cancer.MethodsThe ectopic vector expression and biochemical inhibitor were used to analyze the KDM2A expression regulated by HS-578 T conditioned medium or IL-6 in mammary fibroblasts. Immunoprecipitation and chromatin immunoprecipitation assays were conducted to examine the interaction between STAT3 and NF kappa B p50. M2 macrophage polarization was assessed by analyzing M2 macrophage-specific markers using flow cytometry and RT-PCR. ESTIMATE algorithm was used to analyze the tumor microenvironment-dominant breast cancer samples from the TCGA database. The correlation between stromal KDM2A and CD163 + M2 macrophages was analyzed using the Pearson correlation coefficient. Cell viability was determined using trypan blue exclusion assay.ResultsIL-6 regulates gene expression via activation and dimerization of STAT3 or collaboration of STAT3 and NF kappa B. However, STAT3, a downstream transcription factor of the IL-6 signaling pathway, was directly complexed with NF kappa B p50, not NF kappa B p65, to upregulate the expression of KDM2A in CAFs. Enrichment analysis of immune cells/stromal cells using TCGA-breast cancer RNA-seq data unveiled a positive correlation between stromal KDM2A and the abundance of M2 macrophages. CXCR2-associated chemokines secreted by KDM2A-expressing CAFs stimulated M2 macrophage polarization, which in turn secreted CCL2 to increase paclitaxel resistance in breast cancer cells by activating CCR2 signaling.ConclusionThis study revealed the non-canonical molecular mechanism of IL-6 secreted by breast cancer upregulated KDM2A expression in CAFs via a novel STAT3/NF kappa B p50 axis, which STAT3 complexed with NF kappa B p50 in NF kappa B p50 binding motif of KDM2A promoter. KDM2A-expressing CAFs dominantly secreted the CXCR2-associated chemokines to promote M2 macrophage polarization and enhance paclitaxel resistance in breast cancer. These findings underscore the therapeutic potential of targeting the CXCR2 or CCR2 pathway as a novel strategy for paclitaxel-resistant breast cancer.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available