Transferrin receptor regulates malignancies and the stemness of hepatocellular carcinoma-derived cancer stem-like cells by affecting iron accumulation

PLoS One. 2020 Dec 22;15(12):e0243812. doi: 10.1371/journal.pone.0243812. eCollection 2020.

Abstract

Background: Iron metabolism is essential because it plays regulatory roles in various physiological and pathological processes. Disorders of iron metabolism balance are related to various cancers, including hepatocellular carcinoma. Cancer stem-like cells (CSCs) exert critical effects on chemotherapy failure, cancer metastasis, and subsequent disease recurrence and relapse. However, little is known about how iron metabolism affects liver CSCs. Here, we investigated the expression of transferrin receptor 1 (TFR1) and ferroportin (FPN), two iron importers, and an upstream regulator, iron regulatory protein 2 (IRP2), in liver hepatocellular carcinoma (LIHC) and related CSCs.

Methods: The expression levels of TFR1, FPN and IRP2 were analysed using the GEPIA database. CSCs were derived from parental LIHC cells cultured in serum-free medium. After TFR1 knockdown, ROS accumulation and malignant behaviours were measured. The CCK-8 assay was performed to detect cell viability after TFR1 knockdown and erastin treatment.

Results: TFR1 expression was upregulated in LIHC tissue and CSCs derived from LIHC cell lines, prompting us to investigate the roles of TFR1 in regulating CSCs. Knockdown of TFR1 expression decreased iron accumulation and inhibited malignant behaviour. Knockdown of TFR1 expression decreased reactive oxygen species (ROS) accumulation induced by erastin treatment and maintained mitochondrial function, indicating that TFR1 is critical in regulating erastin-induced cell death in CSCs. Additionally, knockdown of TFR1 expression decreased sphere formation by decreasing iron accumulation in CSCs, indicating a potential role for TFR1 in maintaining stemness.

Conclusion: These findings, which revealed TFR1 as a critical regulator of LIHC CSCs in malignant behaviour and stemness that functions by regulating iron accumulation, may have implications to improve therapeutic approaches.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Carcinoma, Hepatocellular / pathology*
  • Cell Line, Tumor
  • Humans
  • Iron / metabolism*
  • Liver Neoplasms / pathology*
  • Neoplastic Stem Cells / pathology*
  • Phenotype
  • Receptors, Transferrin / metabolism*

Substances

  • Receptors, Transferrin
  • Iron

Grants and funding

This work was supported by Young Scientists Fund of the National Natural Science Foundation of China (Grant No. 81803994), General Program of National Natural Science Foundation of China (Grant No. 81774284), International Cooperation Project of Sichuan Science and Technology Department (Grant No. 2019YFH0152) and Science and Technology Developmental Foundation of Chengdu University of TCM (Grant No. QNXZ2019022) The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.