Cell
Volume 158, Issue 3, 31 July 2014, Pages 534-548
Journal home page for Cell

Article
Allosteric Inhibition of the IRE1α RNase Preserves Cell Viability and Function during Endoplasmic Reticulum Stress

https://doi.org/10.1016/j.cell.2014.07.002Get rights and content
Under an Elsevier user license
open archive

Highlights

  • ER stress triggers a “terminal UPR” via hyperactivation of IRE1α, an ER kinase/RNase

  • Hyperactive IRE1α’s RNase degrades ER-localized mRNAs and miRNAs to cause apoptosis

  • Kinase-Inhibiting RNase-Attenuators (KIRAs) reduce IRE1α’s RNase activity

  • KIRA6—an advanced KIRA—preserves cell survival and function in many ER stress models

Summary

Depending on endoplasmic reticulum (ER) stress levels, the ER transmembrane multidomain protein IRE1α promotes either adaptation or apoptosis. Unfolded ER proteins cause IRE1α lumenal domain homo-oligomerization, inducing trans autophosphorylation that further drives homo-oligomerization of its cytosolic kinase/endoribonuclease (RNase) domains to activate mRNA splicing of adaptive XBP1 transcription factor. However, under high/chronic ER stress, IRE1α surpasses an oligomerization threshold that expands RNase substrate repertoire to many ER-localized mRNAs, leading to apoptosis. To modulate these effects, we developed ATP-competitive IRE1α Kinase-Inhibiting RNase Attenuators—KIRAs—that allosterically inhibit IRE1α’s RNase by breaking oligomers. One optimized KIRA, KIRA6, inhibits IRE1α in vivo and promotes cell survival under ER stress. Intravitreally, KIRA6 preserves photoreceptor functional viability in rat models of ER stress-induced retinal degeneration. Systemically, KIRA6 preserves pancreatic β cells, increases insulin, and reduces hyperglycemia in Akita diabetic mice. Thus, IRE1α powerfully controls cell fate but can itself be controlled with small molecules to reduce cell degeneration.

Cited by (0)

12

Co-first author