1. Academic Validation
  2. Selective, reversible caspase-3 inhibitor is neuroprotective and reveals distinct pathways of cell death after neonatal hypoxic-ischemic brain injury

Selective, reversible caspase-3 inhibitor is neuroprotective and reveals distinct pathways of cell death after neonatal hypoxic-ischemic brain injury

  • J Biol Chem. 2002 Aug 16;277(33):30128-36. doi: 10.1074/jbc.M202931200.
Byung Hee Han 1 Daigen Xu Junjeong Choi Yongxin Han Steven Xanthoudakis Sophie Roy John Tam John Vaillancourt John Colucci Robert Siman Andre Giroux George S Robertson Robert Zamboni Donald W Nicholson David M Holtzman
Affiliations

Affiliation

  • 1 Department of Neurology, Washington University, St. Louis, Missouri 63110, USA.
Abstract

Hypoxia-ischemia (H-I) in the developing brain results in brain injury with prominent features of both Apoptosis and necrosis. A peptide-based pan-caspase inhibitor is neuroprotective against neonatal H-I brain injury, suggesting a central role of caspases in brain injury. Because previously studied peptide-based Caspase inhibitors are not potent and are only partially selective, the exact contribution of specific caspases and other proteases to injury after H-I is not clear. In this study, we explored the neuroprotective effects of a small, reversible Caspase-3 inhibitor M826. M826 selectively and potently inhibited both Caspase-3 enzymatic activity and Apoptosis in cultured cells in vitro. In a rat model of neonatal H-I, M826 blocked Caspase-3 activation and cleavage of its substrates, which begins 6 h and peaks 24 h after H-I. Although M826 significantly reduced DNA fragmentation and brain tissue loss, it did not prevent calpain activation in the cortex. This activation, which is associated with excitotoxic/necrotic cell injury, occurred within 30 min to 2 h after H-I even in the presence of M826. Similar to calpain activation, we found evidence of caspase-2 processing within 30 min to 2 h after H-I that was not affected by M826. Caspase-2 processing appeared to be secondary to calpain-mediated cleavage and was not associated with caspase-2 activation. These data suggest that Caspase-3 specifically contributes to delayed cell death and brain injury after neonatal H-I and that calpain activation is associated with and likely a marker for the early component of excitotoxic/necrotic brain injury previously demonstrated in this model.

Figures
Products