4.7 Article

Adipocyte coculture induces a pro-inflammatory, fibrotic, angiogenic, and proliferative microenvironment in uterine leiomyoma cells

出版社

ELSEVIER
DOI: 10.1016/j.bbadis.2022.166564

关键词

Uterine leiomyoma; Adipocyte; Inflammation; Fibrosis; Angiogenesis; Proliferation

向作者/读者索取更多资源

The aim of this study was to determine the cellular and molecular mechanisms by which adipocytes contribute to the initiation and promotion of uterine leiomyoma tumors. The results showed that adipocytes promote inflammation, fibrosis, and angiogenesis in leiomyoma cells by upregulating associated factors and transferring free fatty acids. Adipocyte-leiomyoma cell interaction increases the phosphorylation of NF-.B and enhances cell proliferation.
Objective: Obesity and its consequences are among the biggest challenges facing the healthcare system. Uterine leiomyomas are the most common gynecologic tumors. The risk of leiomyoma increases with obesity, but the underlying mechanisms of this association remain unclear. The aim of the present study to determine the cellular and molecular mechanisms by which adipocyte contributes to both leiomyoma tumor initiation and promotion. Methods: Primary myometrium and leiomyoma cells were isolated from patients who underwent a hysterectomy or myomectomy. Pro-inflammatory, fibrotic, and angiogenic factors were measured using a multiplex cytokine array in human primary and immortalized myometrial and leiomyoma cells cocultured with human adipocyte (SW872) cells, or in animal ELT3 leiomyoma cells cocultured with 3 T3-L1 adipocytes. The free fatty acids (FFAs) and fatty acid-binding protein 4 (FABP4) levels were measured using immunofluorescence assays. Other protein abundances were determined using western blots. The expression levels of TNF-a, MCP-1, phospho-NF-.B, TGF ss 3 and VEGF-A in lean and obese in different leiomyoma patients were determined by immunofluorescence staining. Results: Adipocytes promote inflammation, fibrosis, and angiogenesis in uterine leiomyoma cells by upregulating associated factors, such as IL-1 ss, TNF-a, MCP-1, GM-CSF, TGF-ss s, PLGF, VEGF, HB-EGF, G-CSF and FGF2. Coculture led to the transfer of FFAs and FABP4 from adipocytes to leiomyoma cells, suggesting that adipocytes may modulate metabolic activity in these tumor cells. Increased levels of FFA and FABP4 expressions were detected in obese leiomyoma tissue compared to lean. The adipocyte-leiomyoma cell interaction increased the phospho-NF-.B level, which plays a key role in inflammation, restructuring metabolic pathways, and angiogenesis. Obese leiomyoma patients expressed a higher amount of TNF-a, MCP-1, phospho-NF-.B, TGF ss 3 and VEGF-A than lean leiomyoma patients, consistent with in vitro findings. Furthermore, we found that adipocyte secretory factors enhance leiomyoma cell proliferation by increasing PCNA abundance. Finally, the inhibition of the inflammatory factors TNF-a, MCP-1, and NF-.B abrogated the adipocyte coculture-induced proliferation of leiomyoma cells. Conclusions: Adipocytes release inflammatory, fibrotic, and angiogenic factors, along with FFAs, which contribute to a tumor-friendly microenvironment that may promote leiomyoma growth and can represent a new target for leiomyoma prevention and treatment.

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