Atrial Myocyte NLRP3/CaMKII Nexus Forms a Substrate for Postoperative Atrial Fibrillation

Author:

Heijman Jordi12ORCID,Muna Azinwi Phina1,Veleva Tina1,Molina Cristina E.13,Sutanto Henry2ORCID,Tekook Marcel1,Wang Qiongling45,Abu-Taha Issam H.1,Gorka Marcel1,Künzel Stephan16,El-Armouche Ali6,Reichenspurner Hermann7,Kamler Markus8,Nikolaev Viacheslav3,Ravens Ursula910,Li Na411,Nattel Stanley1213ORCID,Wehrens Xander H.T.45ORCID,Dobrev Dobromir1ORCID

Affiliation:

1. Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (J.H., A.P.M., T.V., C.E.M., M.T., I.H.A.-T., M.G., S.N., D.D.).

2. Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, The Netherlands (J.H., H.S.,).

3. Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf and DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (C.E.M., V.N.).

4. Cardiovascular Research Institute (Q.W., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.

5. Molecular Physiology and Biophysics, Medicine, Pediatrics, Neuroscience, and Center for Space Medicine (Q.W., X.H.T.W.), Baylor College of Medicine, Houston, TX.

6. Pharmacology and Toxicology, Medical Faculty, Technische Universität Dresden, Dresden, Germany (S.K., A.E.-A.).

7. Cardiovascular Surgery, University Heart Center Hamburg and DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (H.R.).

8. Thoracic and Cardiovascular Surgery, West German Heart and Vascular Center Essen, University Hospital Essen, Essen, Germany (M.K.).

9. Institute of Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany (U.R.).

10. Institute of Physiology, Medical Faculty Carl Gustav Carus, Technical University of Dresden, Dresden, Germany (U.R.).

11. Medicine (Section of Cardiovascular Research) (N.L.), Baylor College of Medicine, Houston, TX.

12. Medicine, Montreal Heart Institute and Université de Montréal & Department of Pharmacology and Therapeutics, McGill University Montreal, Canada (S.N.).

13. IHU LIRYC and Fondation Bordeaux Université, Bordeaux, France (S.N.).

Abstract

Rationale: Postoperative atrial fibrillation (POAF) is a common and troublesome complication of cardiac surgery. POAF is generally believed to occur when postoperative triggers act on a preexisting vulnerable substrate, but the underlying cellular and molecular mechanisms are largely unknown. Objective: To identify cellular POAF mechanisms in right atrial samples from patients without a history of atrial fibrillation undergoing open-heart surgery. Methods and Results: Multicellular action potentials, membrane ion-currents (perforated patch-clamp), or simultaneous membrane-current (ruptured patch-clamp) and [Ca 2+ ] i -recordings in atrial cardiomyocytes, along with protein-expression levels in tissue homogenates or cardiomyocytes, were assessed in 265 atrial samples from patients without or with POAF. No indices of electrical, profibrotic, or connexin remodeling were noted in POAF, but Ca 2+ -transient amplitude was smaller, although spontaneous sarcoplasmic reticulum (SR) Ca 2+ -release events and L-type Ca 2+ -current alternans occurred more frequently. CaMKII (Ca 2+ /calmodulin-dependent protein kinase-II) protein-expression, CaMKII-dependent phosphorylation of the cardiac RyR2 (ryanodine-receptor channel type-2), and RyR2 single-channel open-probability were significantly increased in POAF. SR Ca 2+ -content was unchanged in POAF despite greater SR Ca 2+ -leak, with a trend towards increased SR Ca 2+ -ATPase activity. Patients with POAF also showed stronger expression of activated components of the NLRP3 (NACHT, LRR, and PYD domains-containing protein-3)-inflammasome system in atrial whole-tissue homogenates and cardiomyocytes. Acute application of interleukin-1β caused NLRP3-signaling activation and CaMKII-dependent RyR2/phospholamban hyperphosphorylation in an immortalized mouse atrial cardiomyocyte cell-line (HL-1-cardiomyocytes) and enhanced spontaneous SR Ca 2+ -release events in both POAF cardiomyocytes and HL-1-cardiomyocytes. Computational modeling showed that RyR2 dysfunction and increased SR Ca 2+ -uptake are sufficient to reproduce the Ca 2+ -handling phenotype and indicated an increased risk of proarrhythmic delayed afterdepolarizations in POAF subjects in response to interleukin-1β. Conclusions: Preexisting Ca 2+ -handling abnormalities and activation of NLRP3-inflammasome/CaMKII signaling are evident in atrial cardiomyocytes from patients who subsequently develop POAF. These molecular substrates sensitize cardiomyocytes to spontaneous Ca 2+ -releases and arrhythmogenic afterdepolarizations, particularly upon exposure to inflammatory mediators. Our data reveal a potential cellular and molecular substrate for this important clinical problem.

Funder

ZonMw

HHS | NIH | National Heart, Lung, and Blood Institute

Deutsche Forschungsgemeinschaft

Gouvernement du Canada | Canadian Institutes of Health Research

Heart and Stroke Foundation of Canada

Publisher

Ovid Technologies (Wolters Kluwer Health)

Subject

Cardiology and Cardiovascular Medicine,Physiology

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3