Your browser doesn't support javascript.
loading
Atrial Myocyte NLRP3/CaMKII Nexus Forms a Substrate for Postoperative Atrial Fibrillation.
Heijman, Jordi; Muna, Azinwi Phina; Veleva, Tina; Molina, Cristina E; Sutanto, Henry; Tekook, Marcel; Wang, Qiongling; Abu-Taha, Issam H; Gorka, Marcel; Künzel, Stephan; El-Armouche, Ali; Reichenspurner, Hermann; Kamler, Markus; Nikolaev, Viacheslav; Ravens, Ursula; Li, Na; Nattel, Stanley; Wehrens, Xander H T; Dobrev, Dobromir.
Afiliação
  • Heijman J; 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.).
  • Muna AP; Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, The Netherlands (J.H., H.S.,).
  • Veleva T; 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.).
  • Molina CE; 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.).
  • Sutanto H; 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.).
  • Tekook M; 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.).
  • Wang Q; Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, The Netherlands (J.H., H.S.,).
  • Abu-Taha IH; 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.).
  • Gorka M; Cardiovascular Research Institute (Q.W., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.
  • Künzel S; Molecular Physiology and Biophysics, Medicine, Pediatrics, Neuroscience, and Center for Space Medicine (Q.W., X.H.T.W.), Baylor College of Medicine, Houston, TX.
  • El-Armouche A; 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.).
  • Reichenspurner H; 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.).
  • Kamler M; 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.).
  • Nikolaev V; Pharmacology and Toxicology, Medical Faculty, Technische Universität Dresden, Dresden, Germany (S.K., A.E.-A.).
  • Ravens U; Pharmacology and Toxicology, Medical Faculty, Technische Universität Dresden, Dresden, Germany (S.K., A.E.-A.).
  • Li N; Cardiovascular Surgery, University Heart Center Hamburg and DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (H.R.).
  • Nattel S; Thoracic and Cardiovascular Surgery, West German Heart and Vascular Center Essen, University Hospital Essen, Essen, Germany (M.K.).
  • Wehrens XHT; 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.).
  • Dobrev D; Institute of Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany (U.R.).
Circ Res ; 127(8): 1036-1055, 2020 09 25.
Article em En | MEDLINE | ID: mdl-32762493
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 [Ca2+]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 Ca2+-transient amplitude was smaller, although spontaneous sarcoplasmic reticulum (SR) Ca2+-release events and L-type Ca2+-current alternans occurred more frequently. CaMKII (Ca2+/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 Ca2+-content was unchanged in POAF despite greater SR Ca2+-leak, with a trend towards increased SR Ca2+-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-1ß 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 Ca2+-release events in both POAF cardiomyocytes and HL-1-cardiomyocytes. Computational modeling showed that RyR2 dysfunction and increased SR Ca2+-uptake are sufficient to reproduce the Ca2+-handling phenotype and indicated an increased risk of proarrhythmic delayed afterdepolarizations in POAF subjects in response to interleukin-1ß.

CONCLUSIONS:

Preexisting Ca2+-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 Ca2+-releases and arrhythmogenic afterdepolarizations, particularly upon exposure to inflammatory mediators. Our data reveal a potential cellular and molecular substrate for this important clinical problem.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Fibrilação Atrial / Miócitos Cardíacos / Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina / Inflamassomos / Proteína 3 que Contém Domínio de Pirina da Família NLR / Átrios do Coração / Frequência Cardíaca / Procedimentos Cirúrgicos Cardíacos Tipo de estudo: Observational_studies / Prognostic_studies / Risk_factors_studies Limite: Aged / Animals / Female / Humans / Male / Middle aged Idioma: En Revista: Circ Res Ano de publicação: 2020 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Fibrilação Atrial / Miócitos Cardíacos / Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina / Inflamassomos / Proteína 3 que Contém Domínio de Pirina da Família NLR / Átrios do Coração / Frequência Cardíaca / Procedimentos Cirúrgicos Cardíacos Tipo de estudo: Observational_studies / Prognostic_studies / Risk_factors_studies Limite: Aged / Animals / Female / Humans / Male / Middle aged Idioma: En Revista: Circ Res Ano de publicação: 2020 Tipo de documento: Article