β-Arrestin-2 modulates radiation-induced intestinal crypt progenitor/stem cell injury

Cell Death Differ. 2016 Sep 1;23(9):1529-41. doi: 10.1038/cdd.2016.38. Epub 2016 Apr 29.

Abstract

Intestinal crypt progenitor/stem (ICPS) cell apoptosis and vascular endothelial cell apoptosis are responsible for the initiation and development of ionizing radiation (IR)-evoked gastrointestinal syndrome. The signaling mechanisms underlying IR-induced ICPS cell apoptosis remain largely unclear. Our findings provide evidence that β-arrestin-2 (βarr2)-mediated ICPS cell apoptosis is crucial for IR-stimulated intestinal injury. βArr2-deficient mice exhibited decreased ICPS cell and intestinal Lgr5(+) (leucine-rich repeat-containing G-protein-coupled receptor 5-positive) stem cell apoptosis, promoted crypt proliferation and reproduction, and protracted survival following lethal doses of radiation. Radioprotection in the ICPS cells isolated from βarr2-deficient mice depended on prolonged nuclear factor-κB (NF-κB) activation via direct interaction of βarr2 with IκBα and subsequent inhibition of p53-upregulated modulator of apoptosis (PUMA)-mediated mitochondrial dysfunction. Unexpectedly, βarr2 deficiency had little effect on IR-induced intestinal vascular endothelial cell apoptosis in mice. Consistently, βarr2 knockdown also provided significant radioresistance by manipulating NF-κB/PUMA signaling in Lgr5(+) cells in vitro. Collectively, these observations show that targeting the βarr2/NF-κB/PUMA novel pathway is a potential radiomitigator for limiting the damaging effect of radiotherapy on the gastrointestinal system. Significance statement: acute injury to the intestinal mucosa is a major dose-limiting complication of abdominal radiotherapy. The issue of whether the critical factor for the initiation of radiation-induced intestinal injury is intestinal stem cell apoptosis or endothelial cell apoptosis remains unresolved. βArrs have recently been found to be multifunctional adaptor of apoptosis. Here, we found that β-arrestin-2 (βarr2) deficiency was associated with decreased radiation-induced ICPS cell apoptosis, which prolonged survival in abdominally irradiated mice. Moreover, βarr2 deficiency-mediated intestinal progenitor/stem cell radioprotection relied on protracted NF-κB activation and subsequent suppression of PUMA induction. Our results suggest that ICPS cell apoptosis is the factor involved in the initiation and development of radiation-induced gastrointestinal syndrome. βArr2 is a potential target for lessening radiation-induced ICPS cell apoptosis.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Apoptosis / drug effects
  • Apoptosis / radiation effects*
  • Apoptosis Regulatory Proteins / metabolism
  • Cells, Cultured
  • HCT116 Cells
  • Humans
  • Intestines / cytology
  • Mice
  • Mice, Inbred C57BL
  • Mice, Knockout
  • NF-KappaB Inhibitor alpha / metabolism
  • NF-kappa B / metabolism
  • Nitriles / pharmacology
  • RNA Interference
  • Radiation, Ionizing*
  • Receptors, G-Protein-Coupled / metabolism
  • Signal Transduction
  • Stem Cells / cytology
  • Stem Cells / metabolism
  • Stem Cells / radiation effects
  • Sulfones / pharmacology
  • Tumor Suppressor Protein p53 / metabolism
  • Tumor Suppressor Proteins / metabolism
  • beta-Arrestin 2 / antagonists & inhibitors
  • beta-Arrestin 2 / genetics
  • beta-Arrestin 2 / metabolism*

Substances

  • 3-(4-methylphenylsulfonyl)-2-propenenitrile
  • Apoptosis Regulatory Proteins
  • Lgr5 protein, mouse
  • NF-kappa B
  • Nitriles
  • PUMA protein, mouse
  • Receptors, G-Protein-Coupled
  • Sulfones
  • Tumor Suppressor Protein p53
  • Tumor Suppressor Proteins
  • beta-Arrestin 2
  • NF-KappaB Inhibitor alpha