4.7 Article

CISD3 inhibition drives cystine-deprivation induced ferroptosis

Journal

CELL DEATH & DISEASE
Volume 12, Issue 9, Pages -

Publisher

SPRINGERNATURE
DOI: 10.1038/s41419-021-04128-2

Keywords

-

Categories

Funding

  1. National Natural Science Foundation of China [81971172, 82102938]
  2. Zhejiang Public Welfare Technology Application Research Project [LGF19H080006, LGF21H010008, LGF20H080005]
  3. Medical and Health Science and Technology Project of Zhejiang Province [2019RC014, 2019RC115, 2021KY842, 2021KY483, 2021KY077]
  4. Outstanding Youth Foundation of Zhejiang Provincial People's Hospital [ZRY2020B001]
  5. Key Research and Development Program of Zhejiang Province [WKJ-ZJ-1914]
  6. Foundation of Zhejiang Provincial Administration of Traditional Chinese Medicine [2020ZB020]

Ask authors/readers for more resources

CISD3 plays a crucial regulatory role in cancer progression and ferroptosis, with its overexpression associated with poorer survival rates, while its depletion accelerates ferroptotic cell death. Depletion of CISD3 leads to metabolic reprogramming towards glutaminolysis, which is essential for mitochondrial oxidative phosphorylation.
Ferroptosis, a new form of programmed cell death, not only promotes the pathological process of various human diseases, but also regulates cancer progression. Current perspectives on the underlying mechanisms remain largely unknown. Herein, we report a member of the NEET protein family, CISD3, exerts a regulatory role in cancer progression and ferroptosis both in vivo and in vitro. Pan-cancer analysis from TCGA reveals that expression of CISD3 is generally elevated in various human cancers which are consequently associated with a higher hazard ratio and poorer overall survival. Moreover, knockdown of CISD3 significantly accelerates lipid peroxidation and accentuates free iron accumulation triggered by Xc(-) inhibition or cystine-deprivation, thus causing ferroptotic cell death. Conversely, ectopic expression of the shRNA-resistant form of CISD3 (CISD3res) efficiently ameliorates the ferroptotic cell death. Mechanistically, CISD3 depletion presents a metabolic reprogramming toward glutaminolysis, which is required for the fuel of mitochondrial oxidative phosphorylation. Both the inhibitors of glutaminolysis and the ETC process were capable of blocking the lipid peroxidation and ferroptotic cell death in the shCISD3 cells. Besides, genetic and pharmacological activation of mitophagy can rescue the CISD3 knockdown-induced ferroptosis by eliminating the damaged mitochondria. Noteworthily, GPX4 acts downstream of CISD3 mediated ferroptosis, which fails to reverse the homeostasis of mitochondria. Collectively, the present work provides novel insights into the regulatory role of CISD3 in ferroptotic cell death and presents a potential target for advanced antitumor activity through ferroptosis.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available