Abstract

Glycolytic glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is a multifunctional protein that also mediates cell death under oxidative stress. We reported previously that the active-site cysteine (Cys-152) of GAPDH plays an essential role in oxidative stress-induced aggregation of GAPDH associated with cell death, and a C152A-GAPDH mutant rescues nitric oxide (NO)-induced cell death by interfering with the aggregation of wild type (WT)-GAPDH. However, the detailed mechanism underlying GAPDH aggregate-induced cell death remains elusive. Here we report that NO-induced GAPDH aggregation specifically causes mitochondrial dysfunction. First, we observed a correlation between NO-induced GAPDH aggregation and mitochondrial dysfunction, when GAPDH aggregation occurred at mitochondria in SH-SY5Y cells. In isolated mitochondria, aggregates of WT-GAPDH directly induced mitochondrial swelling and depolarization, whereas mixtures containing aggregates of C152A-GAPDH reduced mitochondrial dysfunction. Additionally, treatment with cyclosporin A improved WT-GAPDH aggregate-induced swelling and depolarization. In doxycycline-inducible SH-SY5Y cells, overexpression of WT-GAPDH augmented NO-induced mitochondrial dysfunction and increased mitochondrial GAPDH aggregation, whereas induced overexpression of C152A-GAPDH significantly suppressed mitochondrial impairment. Further, NO-induced cytochrome c release into the cytosol and nuclear translocation of apoptosis-inducing factor from mitochondria were both augmented in cells overexpressing WT-GAPDH but ameliorated in C152A-GAPDH-overexpressing cells. Interestingly, GAPDH aggregates induced necrotic cell death via a permeability transition pore (PTP) opening. The expression of either WT- or C152A-GAPDH did not affect other cell death pathways associated with protein aggregation, such as proteasome inhibition, gene expression induced by endoplasmic reticulum stress, or autophagy. Collectively, these results suggest that NO-induced GAPDH aggregation specifically induces mitochondrial dysfunction via PTP opening, leading to cell death.

Highlights

  • Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is a glycolytic enzyme that is responsible for the sixth step of glycolysis [1]

  • Our experiments using C152A-GAPDH revealed the involvement of mitochondrial dysfunction, as well as the subsequent release of cyt c and nuclear translocation of apoptosis-inducing factor (AIF) via permeability transition pore (PTP) opening, in nitric oxide (NO)-induced necrotic cell death mediated by GAPDH aggregation

  • One of the most convincing proposed mechanisms underlying mitochondrial swelling and depolarization is the PTP-induced mitochondrial swelling model [39]. Based on this model, using cyclosporin A (CsA), which binds to cyclophilin D and inhibits the opening of PTP [39], we examined whether aggregates of GAPDH induce mitochondrial dysfunction via PTP opening

Read more

Summary

GAPDH Aggregation Mediates Mitochondrial Dysfunction

Plasmic reticulum (ER) stress, and autophagy, which cause cell death (28 –32). PTP opening leads to mitochondrial depolarization and the release of cell death mediators from the intermembrane space, such as cytochrome c (cyt c) and apoptosis-inducing factor (AIF) [37]. From these observations, we focused on mitochondria to elucidate the cell death pathway that is mediated by GAPDH aggregation. Our experiments using C152A-GAPDH revealed the involvement of mitochondrial dysfunction, as well as the subsequent release of cyt c and nuclear translocation of AIF via PTP opening, in NO-induced necrotic cell death mediated by GAPDH aggregation

Results
Discussion
Experimental Procedures
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call