4.7 Article

Oxidized GAPDH transfers S-glutathionylation to a nuclear protein Sirtuin-1 leading to apoptosis

期刊

FREE RADICAL BIOLOGY AND MEDICINE
卷 174, 期 -, 页码 73-83

出版社

ELSEVIER SCIENCE INC
DOI: 10.1016/j.freeradbiomed.2021.07.037

关键词

Glutaredoxin; S-Glutathionylation; Trans-Glutathionylation; GAPDH; SirT1

资金

  1. NIH [R01 DK103750, R01 HL133013, R03 AG 051857]
  2. NIH CTSI [1UL1TR001430]
  3. American Heart Association [16GRNT27660006]
  4. Whi-taker Cardiovascular Institute
  5. Evans Junior Faculty Research Award by the Department of Medicine of Boston University
  6. Metabolic Clinical Research Collaborative

向作者/读者索取更多资源

The study showed that exposure to hydrogen peroxide induced S-glutathionylation and nuclear translocation of GAPDH in HEK 293T cells. Overexpression of Glrx or a redox dead mutant GAPDH inhibited S-glutathionylation and nuclear translocation. Nuclear GAPDH formed a complex with SirT1, transferring S-glutathionylation to SirT1 and inhibiting its deacetylase activity to initiate apoptotic signaling.
Aims: S-glutathionylation is a reversible oxidative modification of protein cysteines that plays a critical role in redox signaling. Glutaredoxin-1 (Glrx), a glutathione-specific thioltransferase, removes protein S-glutathionylation. Glrx, though a cytosolic protein, can activate a nuclear protein Sirtuin-1 (SirT1) by removing its Sglutathionylation. Glrx ablation causes metabolic abnormalities and promotes controlled cell death and fibrosis in mice. Glyceraldehyde 3-phosphate dehydrogenase (GAPDH), a key enzyme of glycolysis, is sensitive to oxidative modifications and involved in apoptotic signaling via the SirT1/p53 pathway in the nucleus. We aimed to elucidate the extent to which S-glutathionylation of GAPDH and glutaredoxin-1 contribute to GAPDH/SirT1/ p53 apoptosis pathway. Results: Exposure of HEK 293T cells to hydrogen peroxide (H2O2) caused rapid S-glutathionylation and nuclear translocation of GAPDH. Nuclear GAPDH peaked 10-15 min after the addition of H2O2. Overexpression of Glrx or redox dead mutant GAPDH inhibited S-glutathionylation and nuclear translocation. Nuclear GAPDH formed a protein complex with SirT1 and exchanged S-glutathionylation to SirT1 and inhibited its deacetylase activity. Inactivated SirT1 remained stably bound to acetylated-p53 and initiated apoptotic signaling resulting in cleavage of caspase-3. We observed similar effects in human primary aortic endothelial cells suggesting the GAPDH/ SirT1/p53 pathway as a common apoptotic mechanism. Conclusions: Abundant GAPDH with its highly reactive-cysteine thiolate may function as a cytoplasmic rheostat to sense oxidative stress. S-glutathionylation of GAPDH may relay the signal to the nucleus where GAPDH transglutathionylates nuclear proteins such as SirT1 to initiate apoptosis. Glrx reverses GAPDH S-glutathionylation and prevents its nuclear translocation and cytoplasmic-nuclear redox signaling leading to apoptosis. Our data suggest that trans-glutathionylation is a critical step in apoptotic signaling and a potential mechanism that cytosolic Glrx controls nuclear transcription factors.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据