Suppr超能文献

癌症相关成纤维细胞(CAFs)通过外泌体转运丝氨酸蛋白酶抑制剂E2(PN1)来调节肺癌的恶性进展。

Cancer-associated Fibroblasts (CAFs) Regulate Lung Cancer Malignant Progression by Transferring SERPINE2 (PN1) Via Exosomes.

作者信息

Chen Yu, Zhu Sihong, Yang Ling, Lu YuFen, Ye Xiaoqun

机构信息

Department of Respiratory Diseases, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China.

出版信息

Curr Mol Med. 2024 Oct 25. doi: 10.2174/0115665240289093240109062852.

Abstract

BACKGROUND AND AIM

Cancer-associated fibroblasts (CAFs), one of the most abundant stromal cell types in tumor microenvironment (TME), have been a potential target for cancer treatment such as lung cancer. However, the underlying mechanism by which CAFs promote lung cancer progression remains elusive.

METHODS

We obtained primary CAFs, normal fibroblasts (NFs) and their exosomes, constructed protease nexin-1 (PN1) stably silenced or over-expressed CAFs cells using lentivirus. Bioinformatics was used to obtain the expression of PN1 in lung cancer and normal tissues, the relationship with overall survival, and the enriched pathways. The MTT assays and Transwell assays were performed to detect the proliferation, migration, and invasion abilities of lung cancer cells after treatments. Western blotting, qRT-PCR, immunohistochemistry, and xenograft models were used to illustrate how CAFs functions in lung cancer progression via exosomes.

RESULTS

CAFs-derived exosomes, in which PN1 was higher expressed compared with NFs-derived ones, promoted effectively the proliferation, migration, and invasion of lung cancer cells A549 and H1975. Meanwhile, the expression of PN1 expressed higher in lung cancer tissues compared with normal ones, and was negatively associated with the overall survival rate of lung cancer patients. More importantly, over-expressing or silencing PN1 in A549 and H1975 could also promote or inhibit cell proliferation, migration, and invasion correspondingly. Furthermore, treated with PN1 over-expressed CAFs-derived exosomes, the lung cancer cells proliferation, migration, and invasion varied positively, and accompanied by activation of Toll-like and NF-κB signaling pathways. However, this phenomenon can be reversed by AN-3485, an antagonist of Toll-like pathway. Finally, over-expressing PN1 leads to an accelerated tumor growth by increasing the expression of proliferation biomarker Ki67 and activation of NF-κB signaling pathway in vivo.

CONCLUSIONS

CAFs promoted lung cancer progression by transferring PN1 and activating Toll-like/NF-κB signaling pathway via exosomes.

摘要

背景与目的

癌症相关成纤维细胞(CAFs)是肿瘤微环境(TME)中最丰富的基质细胞类型之一,一直是肺癌等癌症治疗的潜在靶点。然而,CAFs促进肺癌进展的潜在机制仍不清楚。

方法

我们获取了原代CAFs、正常成纤维细胞(NFs)及其外泌体,使用慢病毒构建了蛋白酶nexin-1(PN1)稳定沉默或过表达的CAFs细胞。利用生物信息学获得PN1在肺癌组织和正常组织中的表达、与总生存期的关系以及富集通路。进行MTT试验和Transwell试验以检测处理后肺癌细胞的增殖、迁移和侵袭能力。采用蛋白质免疫印迹法、qRT-PCR、免疫组织化学和异种移植模型来说明CAFs如何通过外泌体在肺癌进展中发挥作用。

结果

与NFs来源的外泌体相比,CAFs来源的外泌体中PN1表达更高,能有效促进肺癌细胞A549和H1975的增殖、迁移和侵袭。同时,PN1在肺癌组织中的表达高于正常组织,且与肺癌患者的总生存率呈负相关。更重要的是,在A549和H1975中过表达或沉默PN1也能相应地促进或抑制细胞增殖、迁移和侵袭。此外,用PN1过表达的CAFs来源的外泌体处理后,肺癌细胞的增殖、迁移和侵袭呈正相关变化,并伴有Toll样和NF-κB信号通路的激活。然而,这种现象可被Toll样通路拮抗剂AN-3485逆转。最后,在体内过表达PN1通过增加增殖生物标志物Ki67的表达和激活NF-κB信号通路导致肿瘤生长加速。

结论

CAFs通过外泌体传递PN1并激活Toll样/NF-κB信号通路促进肺癌进展。

文献AI研究员

20分钟写一篇综述,助力文献阅读效率提升50倍。

立即体验

用中文搜PubMed

大模型驱动的PubMed中文搜索引擎

马上搜索

文档翻译

学术文献翻译模型,支持多种主流文档格式。

立即体验