4.7 Article

AKR1B10 promotes breast cancer cell proliferation and migration via the PI3K/AKT/NF-kappa B signaling pathway

期刊

CELL AND BIOSCIENCE
卷 11, 期 1, 页码 -

出版社

BMC
DOI: 10.1186/s13578-021-00677-3

关键词

AKR1B10; PI3K/AKT; NF-kappa B; Proliferation; Migration; Breast cancer

资金

  1. Hunan Province Science Fund for Distinguished Young Scholars [2018JJ1021]
  2. Natural Science Foundation of Hunan Province [2019JJ50022, 2020JJ5013]
  3. Health Department project of the Hunan Province [B2019001, C2019003]
  4. Education Department project of Hunan Province [18C0066]
  5. Scientific research project of Huazhong University of Science and Technology Union Hospital
  6. Science and Technique Foundation of Chenzhou [zdy2020049, zdyf2020008]

向作者/读者索取更多资源

Elevated expression of AKR1B10 in breast cancer tissues was associated with lymph node metastasis, tumor size, Ki67 expression, and p53 expression, while inversely correlated with overall and disease-free survival rates. AKR1B10 promotes proliferation, migration, and invasion of breast cancer cells via the PI3K/AKT/NF-kappa B signaling pathway, indicating its potential as a prognostic indicator and therapeutic target in breast cancer.
Background: Aberrant expression of Aldo-Keto reductase family 1 member B10 (AKR1B10) was associated with tumor size and metastasis of breast cancer in our published preliminary studies. However, little is known about the detailed function and underlying molecular mechanism of AKR1B10 in the pathological process of breast cancer. Methods: The relationship between elevated AKR1B10 expression and the overall survival and disease-free survival of breast cancer patients was analyzed by Kaplan-Meier Plotter database. Breast cancer cell lines overexpressing AKR1B10 (MCF-7/AKR1B10) and breast cancer cell lines with knockdown of AKR1B10 (BT-20/shAKR1B10) were constructed to analyze the impact of AKR1B10 expression on cell proliferation and migration of breast cancer. The expression levels of AKR1B10 were detected and compared in the breast cancer cell lines and tissues by RT-qPCR, western blot and immunohistochemistry. The proliferation of breast cancer cells was monitored by CCK8 cell proliferation assay, and the migration and invasion of breast cancer cells was observed by cell scratch test and transwell assay. The proliferation- and EMT-related proteins including cyclinD1, c-myc, Survivin, Twist, SNAI1, SLUG, ZEB1, E-cadherin, PI3K, p-PI3K, AKT, p-AKT, IKB alpha, p-IKB alpha, NF-kappa B p65, p-NF-kappa B p65 were detected by western blot in breast cancer cells. MCF-7/AKR1B10 cells were treated with LY294002, a PI3K inhibitor, to consider the impact of AKR1B10 overexpression on the PI3K/AKT/NF-kappa B signal cascade and the presence of NF-kappa B p65 in nuclear. In vivo tumor xenograft experiments were used to observe the role of AKR1B10 in breast cancer growth in mice. Results: AKR1B10 expression was significantly greater in breast cancer tissue compared to paired non-cancerous tissue. The expression of AKR1B10 positively correlated with lymph node metastasis, tumor size, Ki67 expression, and p53 expression, but inversely correlated with overall and disease-free survival rates. Gene Ontology analysis showed that AKR1B10 activity contributes to cell proliferation. Overexpression of AKR1B10 facilitated the proliferation of MCF-7 cells, and induced the migration and invasion of MCF-7 cells in vitro in association with induction of epithelial-mesenchymal transition (EMT). Conversely, knockdown of AKR1B10 inhibited these effects in BT-20 cells. Mechanistically, AKR1B10 activated PI3K, AKT, and NF-kappa B p65, and induced nuclear translocation of NF-kappa B p65, and expression of proliferation-related proteins including c-myc, cyclinD1, Survivin, and EMT-related proteins including ZEB1, SLUG, Twist, but downregulated E-cadherin expression in MCF-7 cells. AKR1B10 silencing reduced the phosphorylation of PI3K, AKT, and NF-kappa B p65, the nuclear translocation of NF-kappa B p65, and the expression of proliferation- and migration-related proteins in BT-20 cells. LY294002, a PI3K inhibitor, attenuated the phosphorylation of PI3K, AKT, and NF-kappa B p65, and the nuclear translocation of NF-kappa B p65. In vivo tumor xenograft experiments confirmed that AKR1B10 promoted breast cancer growth in mice. Conclusions: AKR1B10 promotes the proliferation, migration and invasion of breast cancer cells via the PI3K/AKT/NF-kappa B signaling pathway and represents a novel prognostic indicator as well as a potential therapeutic target in breast cancer.

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