1. Academic Validation
  2. KLF14 regulates the growth of hepatocellular carcinoma cells via its modulation of iron homeostasis through the repression of iron-responsive element-binding protein 2

KLF14 regulates the growth of hepatocellular carcinoma cells via its modulation of iron homeostasis through the repression of iron-responsive element-binding protein 2

  • J Exp Clin Cancer Res. 2023 Jan 5;42(1):5. doi: 10.1186/s13046-022-02562-4.
Hui Zhou # 1 Junru Chen # 2 Mingjie Fan # 1 3 Huajian Cai 1 Yufei Dong 1 Yue Qiu 1 Qianqian Zhuang 1 Zhaoying Lei 1 Mengyao Li 1 Xue Ding 1 Peng Yan 1 Aifu Lin 1 Shusen Zheng 4 Qingfeng Yan 5 6 7
Affiliations

Affiliations

  • 1 College of Life Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
  • 2 Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China.
  • 3 Department of Pediatrics, The First Affiliated Hospital, School of Medicine Zhejiang University, Hangzhou, 310003, Zhejiang, China.
  • 4 Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China. shusenzheng@zju.edu.cn.
  • 5 College of Life Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China. qfyan@zju.edu.cn.
  • 6 Department of Pediatrics, The First Affiliated Hospital, School of Medicine Zhejiang University, Hangzhou, 310003, Zhejiang, China. qfyan@zju.edu.cn.
  • 7 Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou, 310058, Zhejiang, China. qfyan@zju.edu.cn.
  • # Contributed equally.
Abstract

Background: Hepatocellular carcinoma (HCC) is a multifactor-driven malignant tumor with rapid progression, which causes the difficulty to substantially improve the prognosis of HCC. Limited understanding of the mechanisms in HCC impedes the development of efficacious therapies. Despite Krüpple-Like factors (KLFs) were reported to be participated in HCC pathogenesis, the function of KLF14 in HCC remains largely unexplored.

Methods: We generated KLF14 overexpressed and silenced liver Cancer cells, and nude mouse xenograft models for the in vitro and in vivo study. Luciferase reporter assay, ChIP-qPCR, Co-IP, immunofluorescence were performed for mechanism research. The expression of KLF14 in HCC samples was analyzed by quantitative RT-PCR, Western blotting, and immunohistochemistry (IHC) analysis.

Results: KLF14 was significantly downregulated in human HCC tissues, which was highly correlated with poor prognosis. Inhibition of KLF14 promoted liver Cancer cells proliferation and overexpression of KLF14 suppressed cells growth. KLF14 exerts its anti-tumor function by inhibiting Iron-responsive element-binding protein 2 (IRP2), which then causes transferrin receptor-1(TfR1) downregulation and ferritin upregulation on the basis of IRP-IREs system. This then leading to cellular iron deficiency and HCC cells growth suppression in vitro and in vivo. Interestingly, KLF14 suppressed the transcription of IRP2 via recruiting SIRT1 to reduce the histone acetylation of the IRP2 promoter, resulting in iron depletion and cell growth suppression. More important, we found fluphenazine is an activator of KLF14, inhibiting HCC cells growth through inducing iron deficiency.

Conclusion: KLF14 acts as a tumor suppressor which inhibits the proliferation of HCC cells by modulating cellular iron metabolism via the repression of IRP2. We identified Fluphenazine, as an activator of KLF14, could be a potential compound for HCC therapy. Our findings therefore provide an innovative insight into the pathogenesis of HCC and a promising therapeutic target.

Keywords

Fluphenazine; Hepatocellular carcinoma; IRP2; Iron metabolism; KLF14.

Figures
Products