PRMT6-mediated ADMA promotes p62 phase separation to form a negative feedback loop in ferroptosis

Due to intrinsic defensive response, ferroptosis-activating targeted therapy fails to achieve satisfactory clinical benefits. Though p62-Keap1-Nrf2 axis is activated to form a negative feedback loop during ferroptosis induction, how p62 is activated remains largely unknown. MTS assay was applied to...

Full description

Saved in:
Bibliographic Details
Published inTheranostics Vol. 14; no. 10; pp. 4090 - 4106
Main Authors Feng, Lifeng, Chen, Lini, Wang, Weikai, Wei, Qi, Chen, Minqiang, Jiang, Xin, Hu, Shiman, Wu, Yuchen, Duan, Lian, Zhu, Liyuan, Wang, Xian, Jin, Hongchuan
Format Journal Article
LanguageEnglish
Published Australia Ivyspring International Publisher 01.01.2024
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Due to intrinsic defensive response, ferroptosis-activating targeted therapy fails to achieve satisfactory clinical benefits. Though p62-Keap1-Nrf2 axis is activated to form a negative feedback loop during ferroptosis induction, how p62 is activated remains largely unknown. MTS assay was applied to measure cell growth. Lipid ROS was detected with C11-BODIPY reagent by flow cytometer. Quantitative real-time PCR (qPCR) and western blotting were performed to determine mRNA and protein level. Immunofluorescence (IF) was performed to examine the distribution of proteins. Fluorescence recovery after photobleaching (FRAP) was adopted to evaluate p62 phase separation. Immunoprecipitation (IP), co-IP and Proximal ligation assay (PLA) were performed to detected protein posttranslational modifications and protein-protein interactions. Tumor xenograft model was employed to inspect growth of pancreatic cancer cells. Upon ferroptosis induction, Nuclear Factor E2 Related Factor 2 (Nrf2) protein and its downstream genes such as HMOX1 and NQO1 were upregulated. Knockdown of p62 significantly reversed Nrf2 upregulation and Keap1 decrease after ferroptosis induction. Knockdown of either p62 or Nrf2 remarkably sensitized ferroptosis induction. Due to augmented p62 phase separation, formation of p62 bodies were increased to recruit Keap1 after ferroptosis induction. Protein arginine methyltransferase 6 (PRMT6) mediated asymmetric dimethylarginine (ADMA) of p62 to increase its oligomerization, promoting p62 phase separation and p62 body formation. Knockdown of p62 or PRMT6 notably sensitized pancreatic cancer cells to ferroptosis both and through suppressing Nrf2 signaling. During ferroptosis induction, PRMT6 mediated p62 ADMA to promote its phase separation, sequestering Keap1 to activate Nrf2 signaling and inhibit ferroptosis. Therefore, targeting PRMT6-mediated p62 ADMA could be a new option to sensitize ferroptosis for cancer treatment.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
Lifeng Feng, Lini Chen and Weikai Wang contribute equally.
Competing Interests: The authors have declared that no competing interest exists.
ISSN:1838-7640
1838-7640
DOI:10.7150/thno.94789