Abstract

Hypoxia inducible factor-1 (HIF-1) is a key regulator in hypoxia and can determine the fate of brain cells during ischemia. However, the mechanism of HIF-1 regulation is still not fully understood in ischemic brains. We tested a hypothesis that both the 26S and the 20S proteasomal pathways were involved in HIF-1α degradation under ischemic conditions. Using in vitro ischemic model (oxygen and glucose deprivation) and a mouse model of middle cerebral artery occlusion, we tested effects of inhibitors of proteasomes and prolyl hydroxylase (PHD) on HIF-1α stability and brain injury in cerebral ischemia. We observed that 30 and 60 min of oxygen-glucose deprivation significantly increased the 20S proteasomal activity. We demonstrated that proteasome inhibitors increased HIF-1α stabilization and cell viability and were more effective than PHD inhibitors in primary cultured cortical neurons exposed to oxygen and glucose deprivation. Furthermore, the administration of the proteasome inhibitor, epoxomicin, to mice resulted in smaller infarct size and brain edema than a PHD inhibitor. Our results indicate that 20S proteasomes are involved in HIF-1α degradation in ischemic neurons and that proteasomal inhibition provides more HIF-1α stabilization and neuroprotection than PHD inhibition in cerebral ischemia.

Highlights

  • Hypoxia inducible factor 1 (HIF-1), a transcription factor, is considered to be the most critical factor involved in the cellular response to hypoxia

  • To determine the contributions of both 20S and 26S proteasomal pathways to the degradation of HIF-1α in ischemic neurons, we carried out experiments in the presence of proteasome inhibitors or prolyl hydroxylase (PHD) inhibitors

  • As hydroxylation is a required step for HIF-1α to be degraded through the 26S degradation pathway but not the 20S pathway, PHD and proteasomal inhibitions would differentiate the individual contribution of the two pathways to HIF-1α degradation

Read more

Summary

Introduction

Hypoxia inducible factor 1 (HIF-1), a transcription factor, is considered to be the most critical factor involved in the cellular response to hypoxia. This is mainly due to its regulation of 1–2% of human genes that play important roles in cellular adaptation to low oxygen (Mazure et al, 2004). HIF-1α is degraded through the ubiquitin-dependent proteasomal (26S) pathway. In order for a protein to be targeted to the 26S proteasome, it requires a poly-ubiquitin tail that can be detected by the 19S for subsequent processing and unfolding. It is known that von Hippel-Lindau tumor suppressor (pVHL) possesses ubiquitin ligase E3 activity that attaches ubiquitin to HIF-1α protein.

Results
Discussion
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.