Enhanced cardiomyocyte reactive oxygen species signaling promotes ibrutinib-induced atrial fibrillation

Redox Biol. 2020 Feb:30:101432. doi: 10.1016/j.redox.2020.101432. Epub 2020 Jan 20.

Abstract

Atrial fibrillation (AF) occurs in up to 11% of cancer patients treated with ibrutinib. The pathophysiology of ibrutinib promoted AF is complicated, as there are multiple interactions involved; the detailed molecular mechanisms underlying this are still unclear. Here, we aimed to determine the electrophysiological and molecular mechanisms of burst-pacing-induced AF in ibrutinib-treated mice. The results indicated differentially expressed proteins in ibrutinib-treated mice, identified through proteomic analysis, were found to play a role in oxidative stress-related pathways. Finally, treatment with an inhibitor of NADPH oxidase (NOX) prevented and reversed AF development in ibrutinib-treated mice. It was showed that the related protein expression of reactive oxygen species (ROS) in the ibrutinib group was significantly increased, including NOX2, NOX4, p22-phox, XO and TGF-β protein expression. It was interesting that ibrutinib group also significantly increased the expression of ox-CaMKII, p-CaMKII (Thr-286) and p-RyR2 (Ser2814), causing enhanced abnormal sarcoplasmic reticulum (SR) Ca2+ release and mitochondrial structures, as well as atrial fibrosis and atrial hypertrophy in ibrutinib-treated mice, and apocynin reduced the expression of these proteins. Ibrutinib-treated mice were also more likely to develop AF, and AF occurred over longer periods. In conclusion, our study has established a pathophysiological role for ROS signaling in atrial cardiomyocytes, and it may be that ox-CaMKII and p-CaMKII (Thr-286) are activated by ROS to increase AF susceptibility following ibrutinib treatment. We have also identified the inhibition of NOX as a potential novel AF therapy approach.

Keywords: Atrial fibrillation; Electrical remodeling; Ibrutinib; Reactive oxygen species; Structural remodeling.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Acetophenones / administration & dosage*
  • Acetophenones / pharmacology
  • Adenine / adverse effects
  • Adenine / analogs & derivatives*
  • Animals
  • Atrial Fibrillation / chemically induced
  • Atrial Fibrillation / drug therapy*
  • Atrial Fibrillation / metabolism
  • Calcium / metabolism
  • Calcium-Calmodulin-Dependent Protein Kinase Type 2 / metabolism
  • Disease Models, Animal
  • Humans
  • Male
  • Mice
  • Myocytes, Cardiac / drug effects
  • Myocytes, Cardiac / metabolism*
  • Piperidines / adverse effects*
  • Protein Interaction Maps / drug effects
  • Proteomics
  • Reactive Oxygen Species / metabolism*
  • Sarcoplasmic Reticulum / metabolism
  • Signal Transduction / drug effects

Substances

  • Acetophenones
  • Piperidines
  • Reactive Oxygen Species
  • ibrutinib
  • acetovanillone
  • Calcium-Calmodulin-Dependent Protein Kinase Type 2
  • Adenine
  • Calcium