4.7 Article

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

Journal

REDOX BIOLOGY
Volume 30, Issue -, Pages -

Publisher

ELSEVIER
DOI: 10.1016/j.redox.2020.101432

Keywords

Reactive oxygen species; Ibrutinib; Atrial fibrillation; Electrical remodeling; Structural remodeling

Funding

  1. National Natural Science Foundation of China [81430098, 81870244, 81725024]
  2. National Key Research and Development Program of China [2017YFC1700400, 2018YFC1704901]
  3. National high-level talent special support plan [W02020052]
  4. Beijing Natural Science Foundation [7152049]
  5. Beijing University of Chinese Medicine [2019-JYB-XS-136]

Ask authors/readers for more resources

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-beta 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.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available