C-MYC Inhibited Ferroptosis and Promoted Immune Evasion in Ovarian Cancer Cells through NCOA4 Mediated Ferritin Autophagy

Cells. 2022 Dec 19;11(24):4127. doi: 10.3390/cells11244127.

Abstract

Objective: We aimed to construct the ferritin autophagy regulatory network and illustrate its mechanism in ferroptosis, TME immunity and malignant phenotypes of ovarian cancer.

Methods: First, we used Western blot assays and immunohistochemistry to detect the pathway expression in ovarian cancer samples (C-MYC, NCOA4). Then, we performed RIP and FISH analysis to verify the targeted binding of these factors after which we constructed ovarian cancer cell models and detected pathway regulator expression (NCOA4). Co-localization and Western blot assays were used to detect ferritin autophagy in different experimental groups. We selected corresponding kits to assess ROS contents in ovarian cancer cells. MMP was measured using flow cytometry and mitochondrial morphology was observed through TEM. Then, we chose Clone, EdU and Transwell to evaluate the proliferation and invasion abilities of ovarian cancer cells. We used Western blot assays to measure the DAMP content in ovarian cancer cell supernatants. Finally, we constructed tumor bearing models to study the effect of the C-MYC pathway on ovarian cancer tumorigenesis and TME immune infiltration in in vivo conditions.

Results: Through pathway expression detection, we confirmed that C-MYC was obviously up-regulated and NCOA4 was obviously down-regulated in ovarian cancer samples, while their expression levels were closely related to the malignancy degree of ovarian cancer. RIP, FISH and cell model detection revealed that C-MYC could down-regulate NCOA4 expression through directly targeted binding with its mRNA. Ferritin autophagy and ferroptosis detection showed that C-MYC could inhibit ferroptosis through NCOA4-mediated ferritin autophagy, thus reducing ROS and inhibiting mitophagy in ovarian cancer cells. Cell function tests showed that C-MYC could promote the proliferation and invasion of ovarian cancer cells through the NCOA4 axis. The Western blot assay revealed that C-MYC could reduce HMGB1 release in ovarian cancer cells through the NCOA4 axis. In vivo experiments showed that C-MYC could promote tumorigenesis and immune evasion in ovarian cancer cells through inhibiting HMGB1 release induced by NCOA4-mediated ferroptosis.

Conclusion: According to these results, we concluded that C-MYC could down-regulate NCOA4 expression through directly targeted binding, thus inhibiting ferroptosis and promoting malignant phenotype/immune evasion in ovarian cancer cells through inhibiting ferritin autophagy.

Keywords: ferritin autophagy; ferroptosis; immune evasion; malignant phenotype; ovarian cancer.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Autophagy
  • Carcinogenesis
  • Female
  • Ferritins / metabolism
  • Ferroptosis*
  • HMGB1 Protein / metabolism
  • Humans
  • Immune Evasion
  • Nuclear Receptor Coactivators* / genetics
  • Nuclear Receptor Coactivators* / metabolism
  • Ovarian Neoplasms* / genetics
  • Ovarian Neoplasms* / metabolism
  • Proto-Oncogene Proteins c-myc* / genetics
  • Proto-Oncogene Proteins c-myc* / metabolism
  • Reactive Oxygen Species / metabolism
  • Transcription Factors / metabolism

Substances

  • Ferritins
  • HMGB1 Protein
  • NCOA4 protein, human
  • Nuclear Receptor Coactivators
  • Reactive Oxygen Species
  • Transcription Factors
  • MYC protein, human
  • Proto-Oncogene Proteins c-myc

Grants and funding

Suzhou Youth Project of Promoting Health through Science and Education, Grant ID: KJXW2022010.