1
|
Lin R, Peng X, Li Y, Wang X, Liu X, Jia X, Zhang C, Liu N, Dong J. Empagliflozin attenuates doxorubicin-impaired cardiac contractility by suppressing reactive oxygen species in isolated myocytes. Mol Cell Biochem 2024; 479:2105-2118. [PMID: 37648958 DOI: 10.1007/s11010-023-04830-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 08/14/2023] [Indexed: 09/01/2023]
Abstract
In animal studies, sodium-glucose co-transporter-2 inhibitors-such as empagliflozin-have been shown to improve heart failure and impaired cardiac contractility induced by anthracyclines-including doxorubicin-although the therapeutic mechanism remains unclear. Moreover, abnormalities in Ca2+ handling within ventricular myocytes are the predominant feature of heart failure. Accordingly, this study aimed to investigate whether empagliflozin can alleviate Ca2+ handling disorders induced by acute doxorubicin exposure and elucidate the underlying mechanisms. To this end, ventricular myocytes were isolated from C57BL/6 mice. Contraction function, Ca2+ handling, and mitochondrial reactive oxygen species (ROS) generation were then evaluated using IonOptix or confocal microscopy. Ca2+ handling proteins were detected by western blotting. Results show that incubation with 1 μmol/L of doxorubicin for 120-min impaired cardiac contractility in isolated myocytes, which was significantly alleviated by pretreatment with 1 μmol/L of empagliflozin. Doxorubicin also markedly induced Ca2+ handling disorders, including decreased Ca2+ transients, prolonged Ca2+ transient decay time, enhanced frequency of Ca2+ sparks, and decreased Ca2+ content in the sarcoplasmic reticulum. These dysregulations were improved by pretreatment with empagliflozin. Moreover, empagliflozin effectively inhibited doxorubicin-induced mitochondrial ROS production in isolated myocytes and rescued doxorubicin-induced oxidation of Ca2+/calmodulin-dependent protein kinase II (ox-CaMKII) and CaMKII-dependent phosphorylation of RyR2. Similarly, preincubation with 10 μmol/L Mito-TEMPO mimicked the protective effects of empagliflozin. Collectively, Empagliflozin ameliorated the doxorubicin-induced contraction malfunction and Ca2+-handling disorders. These findings suggest that empagliflozin alleviates Ca2+-handling disorders by improving ROS production in the mitochondria and alleviating the enhanced oxidative CaMKII signaling pathway induced by doxorubicin.
Collapse
Affiliation(s)
- Rong Lin
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No 2. Anzhen Road, Chaoyang, 100029, Beijing, China
- North China Medical & Health Group XingTai General Hospital, Xingtai, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing, China
| | - Xiaodong Peng
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No 2. Anzhen Road, Chaoyang, 100029, Beijing, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing, China
| | - Yukun Li
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No 2. Anzhen Road, Chaoyang, 100029, Beijing, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing, China
| | - Xuesi Wang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No 2. Anzhen Road, Chaoyang, 100029, Beijing, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing, China
| | - Xinmeng Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No 2. Anzhen Road, Chaoyang, 100029, Beijing, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing, China
| | - Xingze Jia
- North China Medical & Health Group XingTai General Hospital, Xingtai, China
| | - Chengjun Zhang
- North China Medical & Health Group XingTai General Hospital, Xingtai, China
| | - Nian Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No 2. Anzhen Road, Chaoyang, 100029, Beijing, China.
- National Clinical Research Center for Cardiovascular Diseases, Beijing, China.
| | - Jianzeng Dong
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No 2. Anzhen Road, Chaoyang, 100029, Beijing, China.
- National Clinical Research Center for Cardiovascular Diseases, Beijing, China.
| |
Collapse
|
2
|
Gissibl T, Stengel L, Tarnowski D, Maier LS, Wagner S, Feder AL, Sag CM. The inotropic and arrhythmogenic effects of acutely increased late I Na are associated with elevated ROS but not oxidation of PKARIα. Front Cardiovasc Med 2024; 11:1379930. [PMID: 39077112 PMCID: PMC11284163 DOI: 10.3389/fcvm.2024.1379930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/10/2024] [Indexed: 07/31/2024] Open
Abstract
Background Acute stimulation of the late sodium current (INaL) as pharmacologically induced by Anemonia toxin II (ATX-II) results in Na+-dependent Ca2+ overload and enhanced formation of reactive oxygen species (ROS). This is accompanied by an acute increase in the amplitude of the systolic Ca2+ transient. Ca2+ transient amplitude is determined by L-type Ca2+-mediated transsarcolemmal Ca2+ influx (ICa) into the cytosol and by systolic Ca2+ release from the sarcoplasmic reticulum (SR). Type-1 protein kinase A (PKARIα) becomes activated upon increased ROS and is capable of stimulating ICa, thereby sustaining the amplitude of the systolic Ca2+ transient upon oxidative stress. Objectives We aimed to investigate whether the increase of the systolic Ca2+ transient as acutely induced by INaL (by ATX-II) may involve stimulation of ICa through oxidized PKARIα. Methods We used a transgenic mouse model in which PKARIα was made resistant to oxidative activation by homozygous knock-in replacement of redox-sensitive Cysteine 17 with Serine within the regulatory subunits of PKARIα (KI). ATX-II (at 1 nmol/L) was used to acutely enhance INaL in freshly isolated ventricular myocytes from KI and wild-type (WT) control mice. Epifluorescence and confocal imaging were used to assess intracellular Ca2+ handling and ROS formation. A ruptured-patch whole-cell voltage-clamp was used to measure INaL and ICa. The impact of acutely enhanced INaL on RIα dimer formation and PKA target structures was studied using Western blot analysis. Results ATX-II increased INaL to a similar extent in KI and WT cells, which was associated with significant cytosolic and mitochondrial ROS formation in both genotypes. Acutely activated Ca2+ handling in terms of increased Ca2+ transient amplitudes and elevated SR Ca2+ load was equally present in KI and WT cells. Likewise, cellular arrhythmias as approximated by non-triggered Ca2+ elevations during Ca2+ transient decay and by diastolic SR Ca2+-spark frequency occurred in a comparable manner in both genotypes. Most importantly and in contrast to our initial hypothesis, ATX-II did not alter the magnitude or inactivation kinetics of ICa in neither WT nor KI cells and did not result in PKARIα dimerization (i.e., oxidation) despite a clear prooxidant intracellular environment. Conclusions The inotropic and arrhythmogenic effects of acutely increased INaL are associated with elevated ROS, but do not involve oxidation of PKARIα.
Collapse
|
3
|
Hegner P, Ofner F, Schaner B, Gugg M, Trum M, Lauerer AM, Maier LS, Arzt M, Lebek S, Wagner S. CaMKIIδ-dependent dysregulation of atrial Na + homeostasis promotes pro-arrhythmic activity in an obstructive sleep apnea mouse model. Front Pharmacol 2024; 15:1411822. [PMID: 38966545 PMCID: PMC11222670 DOI: 10.3389/fphar.2024.1411822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/16/2024] [Indexed: 07/06/2024] Open
Abstract
Background Obstructive sleep apnea (OSA) has been linked to various pathologies, including arrhythmias such as atrial fibrillation. Specific treatment options for OSA are mainly limited to symptomatic approaches. We previously showed that increased production of reactive oxygen species (ROS) stimulates late sodium current through the voltage-dependent Na+ channels via Ca2+/calmodulin-dependent protein kinase IIδ (CaMKIIδ), thereby increasing the propensity for arrhythmias. However, the impact on atrial intracellular Na+ homeostasis has never been demonstrated. Moreover, the patients often exhibit a broad range of comorbidities, making it difficult to ascertain the effects of OSA alone. Objective We analyzed the effects of OSA on ROS production, cytosolic Na+ level, and rate of spontaneous arrhythmia in atrial cardiomyocytes isolated from an OSA mouse model free from comorbidities. Methods OSA was induced in C57BL/6 wild-type and CaMKIIδ-knockout mice by polytetrafluorethylene (PTFE) injection into the tongue. After 8 weeks, their atrial cardiomyocytes were analyzed for cytosolic and mitochondrial ROS production via laser-scanning confocal microscopy. Quantifications of the cytosolic Na+ concentration and arrhythmia were performed by epifluorescence microscopy. Results PTFE treatment resulted in increased cytosolic and mitochondrial ROS production. Importantly, the cytosolic Na+ concentration was dramatically increased at various stimulation frequencies in the PTFE-treated mice, while the CaMKIIδ-knockout mice were protected. Accordingly, the rate of spontaneous Ca2+ release events increased in the wild-type PTFE mice while being impeded in the CaMKIIδ-knockout mice. Conclusion Atrial Na+ concentration and propensity for spontaneous Ca2+ release events were higher in an OSA mouse model in a CaMKIIδ-dependent manner, which could have therapeutic implications.
Collapse
Affiliation(s)
- Philipp Hegner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Florian Ofner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Benedikt Schaner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
- Department of Neurology and Clinical Neurophysiology, University Hospital Augsburg, Augsburg, Germany
| | - Mathias Gugg
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Maximilian Trum
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Anna-Maria Lauerer
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Lars Siegfried Maier
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Michael Arzt
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Simon Lebek
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Stefan Wagner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
4
|
Shi Q, Malik H, Crawford RM, Streeter J, Wang J, Huo R, Shih JC, Chen B, Hall D, Abel ED, Song LS, Anderson EJ. Cardiac monoamine oxidase-A inhibition protects against catecholamine-induced ventricular arrhythmias via enhanced diastolic calcium control. Cardiovasc Res 2024; 120:596-611. [PMID: 38198753 PMCID: PMC11074799 DOI: 10.1093/cvr/cvae012] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 11/01/2023] [Accepted: 11/22/2023] [Indexed: 01/12/2024] Open
Abstract
AIMS A mechanistic link between depression and risk of arrhythmias could be attributed to altered catecholamine metabolism in the heart. Monoamine oxidase-A (MAO-A), a key enzyme involved in catecholamine metabolism and longstanding antidepressant target, is highly expressed in the myocardium. The present study aimed to elucidate the functional significance and underlying mechanisms of cardiac MAO-A in arrhythmogenesis. METHODS AND RESULTS Analysis of the TriNetX database revealed that depressed patients treated with MAO inhibitors had a lower risk of arrhythmias compared with those treated with selective serotonin reuptake inhibitors. This effect was phenocopied in mice with cardiomyocyte-specific MAO-A deficiency (cMAO-Adef), which showed a significant reduction in both incidence and duration of catecholamine stress-induced ventricular tachycardia compared with wild-type mice. Additionally, cMAO-Adef cardiomyocytes exhibited altered Ca2+ handling under catecholamine stimulation, with increased diastolic Ca2+ reuptake, reduced diastolic Ca2+ leak, and diminished systolic Ca2+ release. Mechanistically, cMAO-Adef hearts had reduced catecholamine levels under sympathetic stress, along with reduced levels of reactive oxygen species and protein carbonylation, leading to decreased oxidation of Type II PKA and CaMKII. These changes potentiated phospholamban (PLB) phosphorylation, thereby enhancing diastolic Ca2+ reuptake, while reducing ryanodine receptor 2 (RyR2) phosphorylation to decrease diastolic Ca2+ leak. Consequently, cMAO-Adef hearts exhibited lower diastolic Ca2+ levels and fewer arrhythmogenic Ca2+ waves during sympathetic overstimulation. CONCLUSION Cardiac MAO-A inhibition exerts an anti-arrhythmic effect by enhancing diastolic Ca2+ handling under catecholamine stress.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Calcium/metabolism
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism
- Catecholamines/metabolism
- Cells, Cultured
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Diastole/drug effects
- Disease Models, Animal
- Heart Rate/drug effects
- Mice, Inbred C57BL
- Mice, Knockout
- Monoamine Oxidase/metabolism
- Monoamine Oxidase Inhibitors/pharmacology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Phosphorylation
- Reactive Oxygen Species/metabolism
- Ryanodine Receptor Calcium Release Channel/metabolism
- Tachycardia, Ventricular/enzymology
- Tachycardia, Ventricular/physiopathology
Collapse
Affiliation(s)
- Qian Shi
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
| | - Hamza Malik
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
| | - Rachel M Crawford
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, 180 S Grand Ave., Iowa City, IA 52242, USA
| | - Jennifer Streeter
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
| | - Jinxi Wang
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
| | - Ran Huo
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, 180 S Grand Ave., Iowa City, IA 52242, USA
| | - Jean C Shih
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, USA
| | - Biyi Chen
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
| | - Duane Hall
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, CBRB 2267285, Newton Rd, Iowa City, IA 52242, USA
| | - E Dale Abel
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, CBRB 2267285, Newton Rd, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, 169 Newton Rd, Iowa City, IA 52242, USA
| | - Long-Sheng Song
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, CBRB 2267285, Newton Rd, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, 169 Newton Rd, Iowa City, IA 52242, USA
| | - Ethan J Anderson
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, 180 S Grand Ave., Iowa City, IA 52242, USA
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, CBRB 2267285, Newton Rd, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, 169 Newton Rd, Iowa City, IA 52242, USA
| |
Collapse
|
5
|
Wang S, Zhang K, He M, Guo H, Cui H, Wang S, Lai Y. Effect of type 2 diabetes on cardiac arrhythmias in patients with obstructive hypertrophic cardiomyopathy. Diabetes Metab Syndr 2024; 18:102992. [PMID: 38560956 DOI: 10.1016/j.dsx.2024.102992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 04/04/2024]
Abstract
AIMS Type 2 diabetes (T2D), a prevalent cardiovascular disease, is linked with cardiac arrhythmias such as atrial fibrillation (AF) and ventricular arrhythmia. This study evaluated T2D's impact on these arrhythmias in patients with obstructive hypertrophic cardiomyopathy (OHCM). METHODS AND MATERIALS We retrospectively analyzed the data of 75 patients with OHCM and T2D from two medical centers in China from 2011 to 2020. A propensity score-matched cohort of 150 patients without T2D was also analyzed. RESULTS Altogether, 225 patients were included. The prevalence of supraventricular tachycardia (SVT), AF, and non-sustained ventricular tachycardia (NSVT) was higher in patients with HCM and T2D than in those without T2D. Multivariate logistic regression showed T2D as an independent risk factor for SVT (odds ratio [OR] = 1.90, 95% confidence interval [CI] = 1.01-3.58, P = 0.04), AF (OR = 2.68, 95% CI = 1.27-5.67, P = 0.01), and NSVT (OR = 2.18, 95% CI = 1.04-4.57, P = 0.04). Further analysis identified fasting glucose and glycosylated hemoglobin levels as independent risk factors for AF and NSVT in patients with T2D. CONCLUSIONS T2D independently increases the risk of cardiac arrhythmias (SVT, AF, NSVT) in OHCM patients. Furthermore, fasting glucose and glycosylated hemoglobin levels independently heighten AF and NSVT risk in OHCM patients with T2D.
Collapse
Affiliation(s)
- Shengwei Wang
- Department of Cardiovascular Surgery Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vascular Diseases, Beijing, China
| | - Ke Zhang
- Department of Cardiovascular Surgery Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vascular Diseases, Beijing, China
| | - Meng He
- Department of Cardiovascular Surgery Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vascular Diseases, Beijing, China
| | - Hongchang Guo
- Department of Cardiovascular Surgery Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vascular Diseases, Beijing, China
| | - Hao Cui
- Department of Cardiovascular Surgery Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vascular Diseases, Beijing, China
| | - Shuiyun Wang
- Department of Cardiovascular Surgery, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yongqiang Lai
- Department of Cardiovascular Surgery Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vascular Diseases, Beijing, China.
| |
Collapse
|
6
|
Yang PC, Rose A, DeMarco KR, Dawson JRD, Han Y, Jeng MT, Harvey RD, Santana LF, Ripplinger CM, Vorobyov I, Lewis TJ, Clancy CE. A multiscale predictive digital twin for neurocardiac modulation. J Physiol 2023; 601:3789-3812. [PMID: 37528537 PMCID: PMC10528740 DOI: 10.1113/jp284391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 07/11/2023] [Indexed: 08/03/2023] Open
Abstract
Cardiac function is tightly regulated by the autonomic nervous system (ANS). Activation of the sympathetic nervous system increases cardiac output by increasing heart rate and stroke volume, while parasympathetic nerve stimulation instantly slows heart rate. Importantly, imbalance in autonomic control of the heart has been implicated in the development of arrhythmias and heart failure. Understanding of the mechanisms and effects of autonomic stimulation is a major challenge because synapses in different regions of the heart result in multiple changes to heart function. For example, nerve synapses on the sinoatrial node (SAN) impact pacemaking, while synapses on contractile cells alter contraction and arrhythmia vulnerability. Here, we present a multiscale neurocardiac modelling and simulator tool that predicts the effect of efferent stimulation of the sympathetic and parasympathetic branches of the ANS on the cardiac SAN and ventricular myocardium. The model includes a layered representation of the ANS and reproduces firing properties measured experimentally. Model parameters are derived from experiments and atomistic simulations. The model is a first prototype of a digital twin that is applied to make predictions across all system scales, from subcellular signalling to pacemaker frequency to tissue level responses. We predict conditions under which autonomic imbalance induces proarrhythmia and can be modified to prevent or inhibit arrhythmia. In summary, the multiscale model constitutes a predictive digital twin framework to test and guide high-throughput prediction of novel neuromodulatory therapy. KEY POINTS: A multi-layered model representation of the autonomic nervous system that includes sympathetic and parasympathetic branches, each with sparse random intralayer connectivity, synaptic dynamics and conductance based integrate-and-fire neurons generates firing patterns in close agreement with experiment. A key feature of the neurocardiac computational model is the connection between the autonomic nervous system and both pacemaker and contractile cells, where modification to pacemaker frequency drives initiation of electrical signals in the contractile cells. We utilized atomic-scale molecular dynamics simulations to predict the association and dissociation rates of noradrenaline with the β-adrenergic receptor. Multiscale predictions demonstrate how autonomic imbalance may increase proclivity to arrhythmias or be used to terminate arrhythmias. The model serves as a first step towards a digital twin for predicting neuromodulation to prevent or reduce disease.
Collapse
Affiliation(s)
- Pei-Chi Yang
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA
| | - Adam Rose
- Department of Mathematics, University of California Davis, Davis, CA
| | - Kevin R. DeMarco
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA
| | - John R. D. Dawson
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA
| | - Yanxiao Han
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA
| | - Mao-Tsuen Jeng
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA
| | | | - L. Fernando Santana
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA
| | | | - Igor Vorobyov
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA
| | - Timothy J. Lewis
- Department of Mathematics, University of California Davis, Davis, CA
| | - Colleen E. Clancy
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA
- Center for Precision Medicine and Data Science, University of California Davis, Sacramento, CA
| |
Collapse
|
7
|
Popoiu TA, Maack C, Bertero E. Mitochondrial calcium signaling and redox homeostasis in cardiac health and disease. FRONTIERS IN MOLECULAR MEDICINE 2023; 3:1235188. [PMID: 39086688 PMCID: PMC11285591 DOI: 10.3389/fmmed.2023.1235188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/10/2023] [Indexed: 08/02/2024]
Abstract
The energy demand of cardiomyocytes changes continuously in response to variations in cardiac workload. Cardiac excitation-contraction coupling is fueled primarily by adenosine triphosphate (ATP) production by oxidative phosphorylation in mitochondria. The rate of mitochondrial oxidative metabolism is matched to the rate of ATP consumption in the cytosol by the parallel activation of oxidative phosphorylation by calcium (Ca2+) and adenosine diphosphate (ADP). During cardiac workload transitions, Ca2+ accumulates in the mitochondrial matrix, where it stimulates the activity of the tricarboxylic acid cycle. In this review, we describe how mitochondria internalize and extrude Ca2+, the relevance of this process for ATP production and redox homeostasis in the healthy heart, and how derangements in ion handling cause mitochondrial and cardiomyocyte dysfunction in heart failure.
Collapse
Affiliation(s)
- Tudor-Alexandru Popoiu
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
- “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Edoardo Bertero
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
- Chair of Cardiovascular Disease, Department of Internal Medicine and Specialties, University of Genoa, Genova, Italy
| |
Collapse
|
8
|
Xiao Z, Guan L, Shi H, Yu Y, Yu Z, Qin S, Su Y, Chen R, Li M, Ge J. Trimetazidine Affects Mitochondrial Calcium Uniporter Expression to Restore Ischemic Heart Function via Reactive Oxygen Species/NFκB Pathway Inhibition. J Cardiovasc Pharmacol 2023; 82:104-116. [PMID: 37163369 PMCID: PMC10402877 DOI: 10.1097/fjc.0000000000001434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 04/15/2023] [Indexed: 05/12/2023]
Abstract
ABSTRACT Studies have demonstrated the roles of trimetazidine beyond being an antianginal agent in ischemic heart disease (IHD) treatment associated with mechanisms of calcium regulation. Our recent studies revealed that mitochondrial calcium uniporter (MCU, the pore-forming unit responsible for mitochondrial calcium entrance) inhibition provided cardioprotective effects for failing hearts. Because trimetazidine and MCU are associated with calcium homeostasis, we hypothesized that trimetazidine may affect MCU to restore the failing heart function. In the present study, we tested this hypothesis in the context of cardiac ischemia in vivo and in vitro. The IHD model was established in male C57BL/6 mice followed by trimetazidine administration intraperitoneally at 20 mg/kg q.o.d for 8 weeks. In vitro studies were performed in a hypoxia model using primary rat neonate cardiomyocytes. The mice survival outcomes and heart function, pathohistologic, and biological changes were analyzed. The results demonstrated that trimetazidine treatment resulted in longer life spans and heart function improvement accompanied by restoration of mitochondrial calcium levels and increase in ATP production via MCU down-regulation. Studies in vitro further showed that trimetazidine treatment and MCU inhibition decreased reactive oxygen species (ROS) production, inhibited the NFκB pathway, and protected the cardiomyocytes from hypoxic injury, and vice versa. Thus, the present study unveils a unique mechanism in which trimetazidine is involved in ameliorating the ischemic failing heart via MCU down-regulation and the following mitochondrial calcium homeostasis restoration, ROS reduction, and cardiomyocyte protection through NFκB pathway inhibition. This mechanism provides a novel explanation for the treatment effects of trimetazidine on IHD.
Collapse
Affiliation(s)
- Zilong Xiao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Lichun Guan
- Department of Cardiovascular Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China; and
| | - Hui Shi
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Yong Yu
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
| | - Ziqing Yu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Shengmei Qin
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Yangang Su
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Ruizhen Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
| | - Minghui Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
| |
Collapse
|
9
|
Packer M. SGLT2 inhibitors: role in protective reprogramming of cardiac nutrient transport and metabolism. Nat Rev Cardiol 2023; 20:443-462. [PMID: 36609604 DOI: 10.1038/s41569-022-00824-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/29/2022] [Indexed: 01/09/2023]
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors reduce heart failure events by direct action on the failing heart that is independent of changes in renal tubular function. In the failing heart, nutrient transport into cardiomyocytes is increased, but nutrient utilization is impaired, leading to deficient ATP production and the cytosolic accumulation of deleterious glucose and lipid by-products. These by-products trigger downregulation of cytoprotective nutrient-deprivation pathways, thereby promoting cellular stress and undermining cellular survival. SGLT2 inhibitors restore cellular homeostasis through three complementary mechanisms: they might bind directly to nutrient-deprivation and nutrient-surplus sensors to promote their cytoprotective actions; they can increase the synthesis of ATP by promoting mitochondrial health (mediated by increasing autophagic flux) and potentially by alleviating the cytosolic deficiency in ferrous iron; and they might directly inhibit glucose transporter type 1, thereby diminishing the cytosolic accumulation of toxic metabolic by-products and promoting the oxidation of long-chain fatty acids. The increase in autophagic flux mediated by SGLT2 inhibitors also promotes the clearance of harmful glucose and lipid by-products and the disposal of dysfunctional mitochondria, allowing for mitochondrial renewal through mitochondrial biogenesis. This Review describes the orchestrated interplay between nutrient transport and metabolism and nutrient-deprivation and nutrient-surplus signalling, to explain how SGLT2 inhibitors reverse the profound nutrient, metabolic and cellular abnormalities observed in heart failure, thereby restoring the myocardium to a healthy molecular and cellular phenotype.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Dallas, TX, USA.
- Imperial College London, London, UK.
| |
Collapse
|
10
|
Uruski P, Matuszewska J, Leśniewska A, Rychlewski D, Niklas A, Mikuła-Pietrasik J, Tykarski A, Książek K. An integrative review of nonobvious puzzles of cellular and molecular cardiooncology. Cell Mol Biol Lett 2023; 28:44. [PMID: 37221467 DOI: 10.1186/s11658-023-00451-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/17/2023] [Indexed: 05/25/2023] Open
Abstract
Oncologic patients are subjected to four major treatment types: surgery, radiotherapy, chemotherapy, and immunotherapy. All nonsurgical forms of cancer management are known to potentially violate the structural and functional integrity of the cardiovascular system. The prevalence and severity of cardiotoxicity and vascular abnormalities led to the emergence of a clinical subdiscipline, called cardiooncology. This relatively new, but rapidly expanding area of knowledge, primarily focuses on clinical observations linking the adverse effects of cancer therapy with deteriorated quality of life of cancer survivors and their increased morbidity and mortality. Cellular and molecular determinants of these relations are far less understood, mainly because of several unsolved paths and contradicting findings in the literature. In this article, we provide a comprehensive view of the cellular and molecular etiology of cardiooncology. We pay particular attention to various intracellular processes that arise in cardiomyocytes, vascular endothelial cells, and smooth muscle cells treated in experimentally-controlled conditions in vitro and in vivo with ionizing radiation and drugs representing diverse modes of anti-cancer activity.
Collapse
Affiliation(s)
- Paweł Uruski
- Department of Hypertensiology, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Julia Matuszewska
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Aleksandra Leśniewska
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Daniel Rychlewski
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Arkadiusz Niklas
- Department of Hypertensiology, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Justyna Mikuła-Pietrasik
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Andrzej Tykarski
- Department of Hypertensiology, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Krzysztof Książek
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland.
| |
Collapse
|
11
|
Mitochondrial connexin43 and mitochondrial K ATP channels modulate triggered arrhythmias in mouse ventricular muscle. Pflugers Arch 2023; 475:477-488. [PMID: 36707457 DOI: 10.1007/s00424-023-02789-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/21/2022] [Accepted: 01/18/2023] [Indexed: 01/29/2023]
Abstract
Connexin43 (Cx43) exits as hemichannels in the inner mitochondrial membrane. We examined how mitochondrial Cx43 and mitochondrial KATP channels affect the occurrence of triggered arrhythmias. To generate cardiac-specific Cx43-deficient (cCx43-/-) mice, Cx43flox/flox mice were crossed with α-MHC (Myh6)-cre+/- mice. The resulting offspring, Cx43flox/flox/Myh6-cre+/- mice (cCx43-/- mice) and their littermates (cCx43+/+ mice), were used. Trabeculae were dissected from the right ventricles of mouse hearts. Cardiomyocytes were enzymatically isolated from the ventricles of mouse hearts. Force was measured with a strain gauge in trabeculae (22°C). To assess arrhythmia susceptibility, the minimal extracellular Ca2+ concentration ([Ca2+]o,min), at which arrhythmias were induced by electrical stimulation, was determined in trabeculae. ROS production was estimated with 2',7'-dichlorofluorescein (DCF), mitochondrial membrane potential with tetramethylrhodamine methyl ester (TMRM), and Ca2+ spark frequency with fluo-4 and confocal microscopy in cardiomyocytes. ROS production within the mitochondria was estimated with MitoSoxRed and mitochondrial Ca2+ with rhod-2 in trabeculae. Diazoxide was used to activate mitochondrial KATP. Most of cCx43-/- mice died suddenly within 8 weeks. Cx43 was present in the inner mitochondrial membrane in cCx43+/+ mice but not in cCx43-/- mice. In cCx43-/- mice, the [Ca2+]o,min was lower, and Ca2+ spark frequency, the slope of DCF fluorescence intensity, MitoSoxRed fluorescence, and rhod-2 fluorescence were higher. TMRM fluorescence was more decreased in cCx43-/- mice. Most of these changes were suppressed by diazoxide. In addition, in cCx43-/- mice, antioxidant peptide SS-31 and N-acetyl-L-cysteine increased the [Ca2+]o,min. These results suggest that Cx43 deficiency activates Ca2+ leak from the SR, probably due to depolarization of mitochondrial membrane potential, an increase in mitochondrial Ca2+, and an increase in ROS production, thereby causing triggered arrhythmias, and that Cx43 hemichannel deficiency may be compensated by activation of mitochondrial KATP channels in mouse hearts.
Collapse
|
12
|
Hegner P, Lebek S, Schaner B, Ofner F, Gugg M, Maier LS, Arzt M, Wagner S. CaMKII-Dependent Contractile Dysfunction and Pro-Arrhythmic Activity in a Mouse Model of Obstructive Sleep Apnea. Antioxidants (Basel) 2023; 12:antiox12020315. [PMID: 36829874 PMCID: PMC9952298 DOI: 10.3390/antiox12020315] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Left ventricular contractile dysfunction and arrhythmias frequently occur in patients with sleep-disordered breathing (SDB). The CaMKII-dependent dysregulation of cellular Ca homeostasis has recently been described in SDB patients, but these studies only partly explain the mechanism and are limited by the patients' heterogeneity. Here, we analyzed contractile function and Ca homeostasis in a mouse model of obstructive sleep apnea (OSA) that is not limited by confounding comorbidities. OSA was induced by artificial tongue enlargement with polytetrafluorethylene (PTFE) injection into the tongue of wildtype mice and mice with a genetic ablation of the oxidative activation sites of CaMKII (MMVV knock-in). After eight weeks, cardiac function was assessed with echocardiography. Reactive oxygen species (ROS) and Ca transients were measured using confocal and epifluorescence microscopy, respectively. Wildtype PTFE mice exhibited an impaired ejection fraction, while MMVV PTFE mice were fully protected. As expected, isolated cardiomyocytes from PTFE mice showed increased ROS production. We further observed decreased levels of steady-state Ca transients, decreased levels of caffeine-induced Ca transients, and increased pro-arrhythmic activity (defined as deviations from the diastolic Ca baseline) only in wildtype but not in MMVV PTFE mice. In summary, in the absence of any comorbidities, OSA was associated with contractile dysfunction and pro-arrhythmic activity and the inhibition of the oxidative activation of CaMKII conveyed cardioprotection, which may have therapeutic implications.
Collapse
Affiliation(s)
- Philipp Hegner
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Simon Lebek
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Benedikt Schaner
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Florian Ofner
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Mathias Gugg
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Lars Siegfried Maier
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Michael Arzt
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Stefan Wagner
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany
- Correspondence:
| |
Collapse
|
13
|
Linz B, Hesselkilde EM, Skarsfeldt MA, Hertel JN, Sattler SM, Yan Y, Tfelt-Hansen J, Diness JG, Bentzen BH, Linz D, Jespersen T. Pharmacological inhibition of SK-channels with AP14145 prevents atrial arrhythmogenic changes in a porcine model for obstructive respiratory events. J Cardiovasc Electrophysiol 2023; 34:126-134. [PMID: 36482155 PMCID: PMC10107889 DOI: 10.1111/jce.15769] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 11/02/2022] [Accepted: 11/07/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Obstructive sleep apnea (OSA) creates a complex substrate for atrial fibrillation (AF), which is refractory to many clinically available pharmacological interventions. We investigated atrial antiarrhythmogenic properties and ventricular electrophysiological safety of small-conductance Ca2+ -activated K+ (SK)-channel inhibition in a porcine model for obstructive respiratory events. METHODS In spontaneously breathing pigs, obstructive respiratory events were simulated by intermittent negative upper airway pressure (INAP) applied via a pressure device connected to the intubation tube. INAP was applied for 75 s, every 10 min, three times before and three times during infusion of the SK-channel inhibitor AP14145. Atrial effective refractory periods (AERP) were acquired before (pre-INAP), during (INAP) and after (post-) INAP. AF-inducibility was determined by a S1S2 atrial pacing protocol. Ventricular arrhythmicity was evaluated by heart rate adjusted QT-interval duration (QT-paced) and electromechanical window (EMW) shortening. RESULTS During vehicle infusion, INAP transiently shortened AERP (pre-INAP: 135 ± 10 ms vs. post-INAP 101 ± 11 ms; p = .008) and increased AF-inducibility. QT-paced prolonged during INAP (pre-INAP 270 ± 7 ms vs. INAP 275 ± 7 ms; p = .04) and EMW shortened progressively throughout INAP and post-INAP (pre-INAP 80 ± 4 ms; INAP 59 ± 6 ms, post-INAP 46 ± 10 ms). AP14145 prolonged baseline AERP, partially prevented INAP-induced AERP-shortening and reduced AF-susceptibility. AP14145 did not alter QT-paced at baseline (pre-AP14145 270 ± 7 ms vs. AP14145 268 ± 6 ms, p = .83) or QT-paced and EMW-shortening during INAP. CONCLUSION In a pig model for obstructive respiratory events, the SK-channel-inhibitor AP14145 prevented INAP-associated AERP-shortening and AF-susceptibility without impairing ventricular electrophysiology. Whether SK-channels represent a target for OSA-related AF in humans warrants further study.
Collapse
Affiliation(s)
- Benedikt Linz
- Faculty of Health and Medical Sciences, Department of Biomedical Sciences, Cardiac Physiology Laboratory, Panum Institutet, University of Copenhagen, Copenhagen, Denmark
| | - Eva M Hesselkilde
- Faculty of Health and Medical Sciences, Department of Biomedical Sciences, Cardiac Physiology Laboratory, Panum Institutet, University of Copenhagen, Copenhagen, Denmark
| | - Mark A Skarsfeldt
- Faculty of Health and Medical Sciences, Department of Biomedical Sciences, Cardiac Physiology Laboratory, Panum Institutet, University of Copenhagen, Copenhagen, Denmark.,Acesion Pharma, Copenhagen, Denmark
| | - Julie N Hertel
- Faculty of Health and Medical Sciences, Department of Biomedical Sciences, Cardiac Physiology Laboratory, Panum Institutet, University of Copenhagen, Copenhagen, Denmark
| | - Stefan M Sattler
- Faculty of Health and Medical Sciences, Department of Biomedical Sciences, Cardiac Physiology Laboratory, Panum Institutet, University of Copenhagen, Copenhagen, Denmark
| | - Yannan Yan
- Faculty of Health and Medical Sciences, Department of Biomedical Sciences, Cardiac Physiology Laboratory, Panum Institutet, University of Copenhagen, Copenhagen, Denmark
| | - Jacob Tfelt-Hansen
- The Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Department of Forensic Medicine, Faculty of Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Bo H Bentzen
- Faculty of Health and Medical Sciences, Department of Biomedical Sciences, Cardiac Physiology Laboratory, Panum Institutet, University of Copenhagen, Copenhagen, Denmark.,Acesion Pharma, Copenhagen, Denmark
| | - Dominik Linz
- Faculty of Health and Medical Sciences, Department of Biomedical Sciences, Cardiac Physiology Laboratory, Panum Institutet, University of Copenhagen, Copenhagen, Denmark.,Centre for Heart Rhythm Disorders, South Australian Health and Medical Research Institute, Royal Adelaide Hospital, University of Adelaide, Adelaide, Australia.,Department of Cardiology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Thomas Jespersen
- Faculty of Health and Medical Sciences, Department of Biomedical Sciences, Cardiac Physiology Laboratory, Panum Institutet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
14
|
Calmodulin-dependent protein kinase II activation promotes kidney mesangial expansion in streptozotocin-induced diabetic mice. Heliyon 2022; 8:e11653. [DOI: 10.1016/j.heliyon.2022.e11653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/30/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
|
15
|
Gao J, Xue G, Zhan G, Wang X, Li J, Yang X, Xia Y. Benefits of SGLT2 inhibitors in arrhythmias. Front Cardiovasc Med 2022; 9:1011429. [PMID: 36337862 PMCID: PMC9631490 DOI: 10.3389/fcvm.2022.1011429] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/04/2022] [Indexed: 09/25/2023] Open
Abstract
Some studies have shown that sodium-glucose cotransporter (SGLT) 2 inhibitors can definitively attenuate the occurrence of cardiovascular diseases such as heart failure (HF), dilated cardiomyopathy (DCM), and myocardial infarction. With the development of research, SGLT2 inhibitors can also reduce the risk of arrhythmias. So in this review, how SGLT2 inhibitors play a role in reducing the risk of arrhythmia from the perspective of electrical remodeling and structural remodeling are explored and then the possible mechanisms are discussed. Specifically, we focus on the role of SGLT2 inhibitors in Na+ and Ca2 + homeostasis and the transients of Na+ and Ca2 +, which could affect electrical remodeling and then lead to arrythmia. We also discuss the protective role of SGLT2 inhibitors in structural remodeling from the perspective of fibrosis, inflammation, oxidative stress, and apoptosis. Ultimately, it is clear that SGLT2 inhibitors have significant benefits on cardiovascular diseases such as HF, myocardial hypertrophy and myocardial infarction. It can be expected that SGLT2 inhibitors can reduce the risk of arrhythmia.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yunlong Xia
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
16
|
Mustroph J, Baier MJ, Pabel S, Stehle T, Trum M, Provaznik Z, Mohler PJ, Musa H, Hund TJ, Sossalla S, Maier LS, Wagner S. Empagliflozin Inhibits Cardiac Late Sodium Current by Ca/Calmodulin-Dependent Kinase II. Circulation 2022; 146:1259-1261. [PMID: 36251785 PMCID: PMC9586469 DOI: 10.1161/circulationaha.122.057364] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Julian Mustroph
- Department of Internal Medicine II (J.M., M.J.B., S.P., T.S., M.T., S.S., L.S.M., S.W.), University Medical Center Regensburg, Germany
| | - Maria J. Baier
- Department of Internal Medicine II (J.M., M.J.B., S.P., T.S., M.T., S.S., L.S.M., S.W.), University Medical Center Regensburg, Germany
| | - Steffen Pabel
- Department of Internal Medicine II (J.M., M.J.B., S.P., T.S., M.T., S.S., L.S.M., S.W.), University Medical Center Regensburg, Germany
| | - Thea Stehle
- Department of Internal Medicine II (J.M., M.J.B., S.P., T.S., M.T., S.S., L.S.M., S.W.), University Medical Center Regensburg, Germany
| | - Maximilian Trum
- Department of Internal Medicine II (J.M., M.J.B., S.P., T.S., M.T., S.S., L.S.M., S.W.), University Medical Center Regensburg, Germany
| | - Zdenek Provaznik
- Department of Cardiothoracic Surgery (Z.P.), University Medical Center Regensburg, Germany
| | - Peter J. Mohler
- Davis Heart and Lung Research Institute, Ohio State University, Columbus (P.J.M., H.M., T.J.H.)
| | - Hassan Musa
- Davis Heart and Lung Research Institute, Ohio State University, Columbus (P.J.M., H.M., T.J.H.)
| | - Thomas J. Hund
- Davis Heart and Lung Research Institute, Ohio State University, Columbus (P.J.M., H.M., T.J.H.)
| | - Samuel Sossalla
- Department of Internal Medicine II (J.M., M.J.B., S.P., T.S., M.T., S.S., L.S.M., S.W.), University Medical Center Regensburg, Germany
| | - Lars S. Maier
- Department of Internal Medicine II (J.M., M.J.B., S.P., T.S., M.T., S.S., L.S.M., S.W.), University Medical Center Regensburg, Germany
| | - Stefan Wagner
- Department of Internal Medicine II (J.M., M.J.B., S.P., T.S., M.T., S.S., L.S.M., S.W.), University Medical Center Regensburg, Germany
| |
Collapse
|
17
|
Abdelsayed M, Page D, Ruben PC. ARumenamides: A novel class of potential antiarrhythmic compounds. Front Pharmacol 2022; 13:976903. [PMID: 36249789 PMCID: PMC9554508 DOI: 10.3389/fphar.2022.976903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/02/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Most therapeutics targeting cardiac voltage-gated sodium channels (Nav1.5) attenuate the sodium current (INa) conducted through the pore of the protein. Whereas these drugs may be beneficial for disease states associated with gain-of-function (GoF) in Nav1.5, few attempts have been made to therapeutically treat loss-of-function (LoF) conditions. The primary impediment to designing efficacious therapies for LoF is a tendency for drugs to occlude the Nav1.5 central pore. We hypothesized that molecular candidates with a high affinity for the fenestrations would potentially reduce pore block.Methods and Results: Virtual docking was performed on 21 compounds, selected based on their affinity for the fenestrations in Nav1.5, which included a class of sulfonamides and carboxamides we identify as ARumenamide (AR). Six ARs, AR-051, AR-189, AR-674, AR-802, AR-807 and AR-811, were further docked against Nav1.5 built on NavAb and rNav1.5. Based on the virtual docking results, these particular ARs have a high affinity for Domain III-IV and Domain VI-I fenestrations. Upon functional characterization, a trend was observed in the effects of the six ARs on INa. An inverse correlation was established between the aromaticity of the AR’s functional moieties and compound block. Due to its aromaticity, AR-811 blocked INa the least compared with other aromatic ARs, which also decelerated fast inactivation onset. AR-674, with its aliphatic functional group, significantly suppresses INa and enhances use-dependence in Nav1.5. AR-802 and AR-811, in particular, decelerated fast inactivation kinetics in the most common Brugada Syndrome Type 1 and Long-QT Syndrome Type 3 mutant, E1784K, without affecting peak or persistent INa.Conclusion: Our hypothesis that LoF in Nav1.5 may be therapeutically treated was supported by the discovery of ARs, which appear to preferentially block the fenestrations. ARs with aromatic functional groups as opposed to aliphatic groups efficaciously maintained Nav1.5 availability. We predict that these bulkier side groups may have a higher affinity for the hydrophobic milieu of the fenestrations, remaining there rather than in the central pore of the channel. Future refinements of AR compound structures and additional validation by molecular dynamic simulations and screening against more Brugada variants will further support their potential benefits in treating certain LoF cardiac arrhythmias.
Collapse
Affiliation(s)
- Mena Abdelsayed
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
- Department of Medicine, Stanford University, Stanford, CA, United States
- *Correspondence: Mena Abdelsayed, ; Peter C. Ruben,
| | - Dana Page
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Peter C. Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- *Correspondence: Mena Abdelsayed, ; Peter C. Ruben,
| |
Collapse
|
18
|
Ziprasidone Induces Rabbit Atrium Arrhythmogenesis via Modification of Oxidative Stress and Sodium/Calcium Homeostasis. Biomedicines 2022; 10:biomedicines10050976. [PMID: 35625713 PMCID: PMC9138982 DOI: 10.3390/biomedicines10050976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/13/2022] [Accepted: 04/21/2022] [Indexed: 11/20/2022] Open
Abstract
Background: Atypical antipsychotics increase the risk of atrial arrhythmias and sudden cardiac death. This study investigated whether ziprasidone, a second-generation antipsychotic, affected intracellular Ca2+ and Na+ regulation and oxidative stress, providing proarrhythmogenic substrates in atriums. Methods: Electromechanical analyses of rabbit atrial tissues were conducted. Intracellular Ca2+ monitoring using Fluo-3, the patch-clamp method for ionic current recordings, and a fluorescence study for the detection of reactive oxygen species and intracellular Na+ levels were conducted in enzymatically dissociated atrial myocytes. Results: Ziprasidone-treated atriums showed sustained triggered activities after rapid pacing, which were inhibited by KN-93 and ranolazine. A reduced peak L-type Ca2+ channel current and enhanced late Na+ current were observed in ziprasidone-treated atrial myocytes, together with an increased cytosolic Na+ level. KN-93 suppressed the enhanced late Na+ current in ziprasidone-treated atrial myocytes. Atrial myocytes treated with ziprasidone showed reduced Ca2+ transient amplitudes and sarcoplasmic reticulum (SR) Ca2+ stores, and increased SR Ca2+ leakage. Cytosolic and mitochondrial reactive oxygen species production was increased in atrial myocytes treated with ziprasidone. TNF-α and NLRP3 were upregulated in ziprasidone-treated myocytes, and the level of phosphorylated calcium/calmodulin-dependent protein kinase II protein was increased. Conclusions: Our results suggest that ziprasidone increases the occurrence of atrial triggered activity and causes intracellular Ca2+ and Na+ dysregulation, which may result from enhanced oxidative stress and activation of the TNF-α/NLRP3 inflammasome pathway in ziprasidone-treated myocytes.
Collapse
|
19
|
Linz B, Hertel JN, Jespersen T, Linz D. Mechanisms and therapeutic opportunities in atrial fibrillation in relationship to alcohol use and abuse. Can J Cardiol 2022; 38:1352-1363. [DOI: 10.1016/j.cjca.2022.04.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/21/2022] [Accepted: 04/01/2022] [Indexed: 12/24/2022] Open
|
20
|
Chen S, Coronel R, Hollmann MW, Weber NC, Zuurbier CJ. Direct cardiac effects of SGLT2 inhibitors. Cardiovasc Diabetol 2022; 21:45. [PMID: 35303888 PMCID: PMC8933888 DOI: 10.1186/s12933-022-01480-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/09/2022] [Indexed: 12/17/2022] Open
Abstract
Sodium-glucose-cotransporter 2 inhibitors (SGLT2is) demonstrate large cardiovascular benefit in both diabetic and non-diabetic, acute and chronic heart failure patients. These inhibitors have on-target (SGLT2 inhibition in the kidney) and off-target effects that likely both contribute to the reported cardiovascular benefit. Here we review the literature on direct effects of SGLT2is on various cardiac cells and derive at an unifying working hypothesis. SGLT2is acutely and directly (1) inhibit cardiac sodium transporters and alter ion homeostasis, (2) reduce inflammation and oxidative stress, (3) influence metabolism, and (4) improve cardiac function. We postulate that cardiac benefit modulated by SGLT2i’s can be commonly attributed to their inhibition of sodium-loaders in the plasma membrane (NHE-1, Nav1.5, SGLT) affecting intracellular sodium-homeostasis (the sodium-interactome), thereby providing a unifying view on the various effects reported in separate studies. The SGLT2is effects are most apparent when cells or hearts are subjected to pathological conditions (reactive oxygen species, inflammation, acidosis, hypoxia, high saturated fatty acids, hypertension, hyperglycemia, and heart failure sympathetic stimulation) that are known to prime these plasmalemmal sodium-loaders. In conclusion, the cardiac sodium-interactome provides a unifying testable working hypothesis and a possible, at least partly, explanation to the clinical benefits of SGLT2is observed in the diseased patient.
Collapse
Affiliation(s)
- Sha Chen
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A.), Amsterdam UMC, Location Academic Medical Centre (AMC), Amsterdam, University of Amsterdam, Cardiovascular Sciences, Meibergdreef 11, Room M0-129, Amsterdam, Noord-Holland, 1105 AZ, The Netherlands
| | - Ruben Coronel
- Department of Experimental Cardiology, Amsterdam UMC, Location Academic Medical Centre (AMC), Amsterdam,, University of Amsterdam, Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Markus W Hollmann
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A.), Amsterdam UMC, Location Academic Medical Centre (AMC), Amsterdam, University of Amsterdam, Cardiovascular Sciences, Meibergdreef 11, Room M0-129, Amsterdam, Noord-Holland, 1105 AZ, The Netherlands
| | - Nina C Weber
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A.), Amsterdam UMC, Location Academic Medical Centre (AMC), Amsterdam, University of Amsterdam, Cardiovascular Sciences, Meibergdreef 11, Room M0-129, Amsterdam, Noord-Holland, 1105 AZ, The Netherlands
| | - Coert J Zuurbier
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A.), Amsterdam UMC, Location Academic Medical Centre (AMC), Amsterdam, University of Amsterdam, Cardiovascular Sciences, Meibergdreef 11, Room M0-129, Amsterdam, Noord-Holland, 1105 AZ, The Netherlands.
| |
Collapse
|
21
|
The Oxidative Balance Orchestrates the Main Keystones of the Functional Activity of Cardiomyocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7714542. [PMID: 35047109 PMCID: PMC8763515 DOI: 10.1155/2022/7714542] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 11/03/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022]
Abstract
This review is aimed at providing an overview of the key hallmarks of cardiomyocytes in physiological and pathological conditions. The main feature of cardiac tissue is the force generation through contraction. This process requires a conspicuous energy demand and therefore an active metabolism. The cardiac tissue is rich of mitochondria, the powerhouses in cells. These organelles, producing ATP, are also the main sources of ROS whose altered handling can cause their accumulation and therefore triggers detrimental effects on mitochondria themselves and other cell components thus leading to apoptosis and cardiac diseases. This review highlights the metabolic aspects of cardiomyocytes and wanders through the main systems of these cells: (a) the unique structural organization (such as different protein complexes represented by contractile, regulatory, and structural proteins); (b) the homeostasis of intracellular Ca2+ that represents a crucial ion for cardiac functions and E-C coupling; and (c) the balance of Zn2+, an ion with a crucial impact on the cardiovascular system. Although each system seems to be independent and finely controlled, the contractile proteins, intracellular Ca2+ homeostasis, and intracellular Zn2+ signals are strongly linked to each other by the intracellular ROS management in a fascinating way to form a "functional tetrad" which ensures the proper functioning of the myocardium. Nevertheless, if ROS balance is not properly handled, one or more of these components could be altered resulting in deleterious effects leading to an unbalance of this "tetrad" and promoting cardiovascular diseases. In conclusion, this "functional tetrad" is proposed as a complex network that communicates continuously in the cardiomyocytes and can drive the switch from physiological to pathological conditions in the heart.
Collapse
|
22
|
Enhanced Cardiac CaMKII Oxidation and CaMKII-Dependent SR Ca Leak in Patients with Sleep-Disordered Breathing. Antioxidants (Basel) 2022; 11:antiox11020331. [PMID: 35204213 PMCID: PMC8868143 DOI: 10.3390/antiox11020331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/26/2022] [Accepted: 02/07/2022] [Indexed: 02/04/2023] Open
Abstract
Background: Sleep-disordered breathing (SDB) is associated with increased oxidant generation. Oxidized Ca/calmodulin kinase II (CaMKII) can contribute to atrial arrhythmias by the stimulation of sarcoplasmic reticulum Ca release events, i.e., Ca sparks. Methods: We prospectively enrolled 39 patients undergoing cardiac surgery to screen for SDB and collected right atrial appendage biopsies. Results: SDB was diagnosed in 14 patients (36%). SDB patients had significantly increased levels of oxidized and activated CaMKII (assessed by Western blotting/specific pulldown). Moreover, SDB patients showed a significant increase in Ca spark frequency (CaSpF measured by confocal microscopy) compared with control subjects. CaSpF was 3.58 ± 0.75 (SDB) vs. 2.49 ± 0.84 (no SDB) 1/100 µm−1s−1 (p < 0.05). In linear multivariable regression models, SDB severity was independently associated with increased CaSpF (B [95%CI]: 0.05 [0.03; 0.07], p < 0.001) after adjusting for important comorbidities. Interestingly, 30 min exposure to the CaMKII inhibitor autocamtide-2 related autoinhibitory peptide normalized the increased CaSpF and eliminated the association between SDB and CaSpF (B [95%CI]: 0.01 [−0.1; 0.03], p = 0.387). Conclusions: Patients with SDB have increased CaMKII oxidation/activation and increased CaMKII-dependent CaSpF in the atrial myocardium, independent of major clinical confounders, which may be a novel target for treatment of atrial arrhythmias in SDB.
Collapse
|
23
|
Varma D, Almeida JFQ, DeSantiago J, Blatter LA, Banach K. Inositol 1,4,5-trisphosphate receptor - reactive oxygen signaling domain regulates excitation-contraction coupling in atrial myocytes. J Mol Cell Cardiol 2022; 163:147-155. [PMID: 34755642 PMCID: PMC8826595 DOI: 10.1016/j.yjmcc.2021.10.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 09/03/2021] [Accepted: 10/13/2021] [Indexed: 02/03/2023]
Abstract
The inositol 1,4,5-trisphosphate receptor (InsP3R) is up-regulated in patients with atrial fibrillation (AF) and InsP3-induced Ca2+ release (IICR) is linked to pro-arrhythmic spontaneous Ca2+ release events. Nevertheless, knowledge of the physiological relevance and regulation of InsP3Rs in atrial muscle is still limited. We hypothesize that InsP3R and NADPH oxidase 2 (NOX2) form a functional signaling domain where NOX2 derived reactive oxygen species (ROS) regulate InsP3R agonist affinity and thereby Ca2+ release. To quantitate the contribution of IICR to atrial excitation-contraction coupling (ECC) atrial myocytes (AMs) were isolated from wild type and NOX2 deficient (Nox2-/-) mice and changes in the cytoplasmic Ca2+ concentration ([Ca2+]i; fluo-4/AM, indo-1) or ROS (2',7'-dichlorofluorescein, DCF) were monitored by fluorescence microscopy. Superfusion of AMs with Angiotensin II (AngII: 1 μmol/L) significantly increased diastolic [Ca2+]i (F/F0, Ctrl: 1.00 ± 0.01, AngII: 1.20 ± 0.03; n = 7; p < 0.05), the field stimulation induced Ca2+ transient (CaT) amplitude (ΔF/F0, Ctrl: 2.00 ± 0.17, AngII: 2.39 ± 0.22, n = 7; p < 0.05), and let to the occurrence of spontaneous increases in [Ca2+]i. These changes in [Ca2+]i were suppressed by the InsP3R blocker 2-aminoethoxydiphenyl-borate (2-APB; 1 μmol/L). Concomitantly, AngII induced an increase in ROS production that was sensitive to the NOX2 specific inhibitor gp91ds-tat (1 μmol/L). In NOX2-/- AMs, AngII failed to increase diastolic [Ca2+]i, CaT amplitude, and the frequency of spontaneous Ca2+ increases. Furthermore, the enhancement of CaTs by exposure to membrane permeant InsP3 was abolished by NOX inhibition with apocynin (1 μM). AngII induced IICR in Nox2-/- AMs could be restored by addition of exogenous ROS (tert-butyl hydroperoxide, tBHP: 5 μmol/L). In saponin permeabilized AMs InsP3 (5 μmol/L) induced Ca2+ sparks that increased in frequency in the presence of ROS (InsP3: 9.65 ± 1.44 sparks*s-1*(100μm)-1; InsP3 + tBHP: 10.77 ± 1.5 sparks*s-1*(100μm)-1; n = 5; p < 0.05). The combined effect of InsP3 + tBHP was entirely suppressed by 2-APB and Xestospongine C (XeC). Changes in IICR due to InsP3R glutathionylation induced by diamide could be reversed by the reducing agent dithiothreitol (DTT: 1 mmol/L) and prevented by pretreatment with 2-APB, supporting that the ROS-dependent post-translational modification of the InsP3R plays a role in the regulation of ECC. Our data demonstrate that in AMs the InsP3R is under dual control of agonist induced InsP3 and ROS formation and suggest that InsP3 and NOX2-derived ROS co-regulate atrial IICR and ECC in a defined InsP3R/NOX2 signaling domain.
Collapse
Affiliation(s)
- Disha Varma
- Dept. of Internal Medicine/Cardiology, Rush University Medical Center, 1750 W. Harrison St, Chicago, IL 60612, USA.
| | - Jonathas F Q Almeida
- Dept. of Internal Medicine/Cardiology, Rush University Medical Center, 1750 W. Harrison St, Chicago, IL 60612, USA.
| | - Jaime DeSantiago
- Dept. of Physiology & Biophysics, Rush University Medical Center, 1750 W. Harrison St, Chicago, IL 60612, USA.
| | - Lothar A Blatter
- Dept. of Physiology & Biophysics, Rush University Medical Center, 1750 W. Harrison St, Chicago, IL 60612, USA.
| | - Kathrin Banach
- Dept. of Internal Medicine/Cardiology, Rush University Medical Center, 1750 W. Harrison St, Chicago, IL 60612, USA.
| |
Collapse
|
24
|
Role of ranolazine in heart failure: From cellular to clinic perspective. Eur J Pharmacol 2022; 919:174787. [PMID: 35114190 DOI: 10.1016/j.ejphar.2022.174787] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/25/2021] [Accepted: 01/25/2022] [Indexed: 12/17/2022]
Abstract
Ranolazine was approved by the US Food and Drug Administration as an antianginal drug in 2006, and has been used since in certain groups of patients with stable angina. The therapeutic action of ranolazine was initially attributed to inhibitory effects on fatty acids metabolism. As investigations went on, however, it developed that the main beneficial effects of ranolazine arise from its action on the late sodium current in the heart. Since late sodium currents were discovered to be involved in various heart pathologies such as ischemia, arrhythmias, systolic and diastolic dysfunctions, and all these conditions are associated with heart failure, ranolazine has in some way been tested either directly or indirectly on heart failure in numerous experimental and clinical studies. As the heart continuously remodels following any sort of severe injury, the inhibition by ranolazine of the underlying mechanisms of cardiac remodeling including ion disturbances, oxidative stress, inflammation, apoptosis, fibrosis, metabolic dysregulation, and neurohormonal impairment are discussed, along with unresolved issues. A projection of pathologies targeted by ranolazine from cellular level to clinical is provided in this review.
Collapse
|
25
|
Koyani CN, Scheruebel S, Jin G, Kolesnik E, Zorn-Pauly K, Mächler H, Hoefler G, von Lewinski D, Heinzel FR, Pelzmann B, Malle E. Hypochlorite-Modified LDL Induces Arrhythmia and Contractile Dysfunction in Cardiomyocytes. Antioxidants (Basel) 2021; 11:25. [PMID: 35052529 PMCID: PMC8772905 DOI: 10.3390/antiox11010025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/17/2021] [Accepted: 12/21/2021] [Indexed: 02/05/2023] Open
Abstract
Neutrophil-derived myeloperoxidase (MPO) and its potent oxidant, hypochlorous acid (HOCl), gained attention as important oxidative mediators in cardiac damage and dysfunction. As cardiomyocytes generate low-density lipoprotein (LDL)-like particles, we aimed to identify the footprints of proatherogenic HOCl-LDL, which adversely affects cellular signalling cascades in various cell types, in the human infarcted myocardium. We performed immunohistochemistry for MPO and HOCl-LDL in human myocardial tissue, investigated the impact of HOCl-LDL on electrophysiology and contractility in primary cardiomyocytes, and explored underlying mechanisms in HL-1 cardiomyocytes and human atrial appendages using immunoblot analysis, qPCR, and silencing experiments. HOCl-LDL reduced ICa,L and IK1, and increased INaL, leading to altered action potential characteristics and arrhythmic events including early- and delayed-afterdepolarizations. HOCl-LDL altered the expression and function of CaV1.2, RyR2, NCX1, and SERCA2a, resulting in impaired contractility and Ca2+ homeostasis. Elevated superoxide anion levels and oxidation of CaMKII were mediated via LOX-1 signaling in HL-1 cardiomyocytes. Furthermore, HOCl-LDL-mediated alterations of cardiac contractility and electrophysiology, including arrhythmic events, were ameliorated by the CaMKII inhibitor KN93 and the INaL blocker, ranolazine. This study provides an explanatory framework for the detrimental effects of HOCl-LDL compared to native LDL and cardiac remodeling in patients with high MPO levels during the progression of cardiovascular disease.
Collapse
Affiliation(s)
- Chintan N. Koyani
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria;
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, 8036 Graz, Austria; (G.J.); (E.K.); (D.v.L.)
| | - Susanne Scheruebel
- Division of Biophysics, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (S.S.); (K.Z.-P.)
| | - Ge Jin
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, 8036 Graz, Austria; (G.J.); (E.K.); (D.v.L.)
- The 2nd Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Ewald Kolesnik
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, 8036 Graz, Austria; (G.J.); (E.K.); (D.v.L.)
| | - Klaus Zorn-Pauly
- Division of Biophysics, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (S.S.); (K.Z.-P.)
| | - Heinrich Mächler
- Department of Surgery, Division of Cardiac Surgery, Medical University of Graz, 8036 Graz, Austria;
| | - Gerald Hoefler
- Diagnostic and Research Center for Molecular BioMedicine, Diagnostic and Research Institute of Pathology, Medical University of Graz, 8010 Graz, Austria;
| | - Dirk von Lewinski
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, 8036 Graz, Austria; (G.J.); (E.K.); (D.v.L.)
| | - Frank R. Heinzel
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, 13353 Berlin, Germany;
- Deutsches Zentrum für Herz-Kreislauf-Forschung (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Brigitte Pelzmann
- Division of Biophysics, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (S.S.); (K.Z.-P.)
| | - Ernst Malle
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria;
| |
Collapse
|
26
|
Robinson VM, Alsalahat I, Freeman S, Antzelevitch C, Barajas-Martinez H, Venetucci L. A Carvedilol Analogue, VK-II-86, Prevents Hypokalaemia-induced Ventricular Arrhythmia through Novel multi-Channel Effects. Br J Pharmacol 2021; 179:2713-2732. [PMID: 34877651 DOI: 10.1111/bph.15775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 11/07/2021] [Accepted: 11/23/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE QT prolongation and intracellular Ca2+ loading with diastolic Ca2+ release via ryanodine receptors (RyR2) are the predominant mechanisms underlying hypokalaemia-induced ventricular arrhythmia. We investigated the antiarrhythmic actions of two RyR2 inhibitors: dantrolene and VK-II-86, a carvedilol analogue with no β-blocking activity, in hypokalaemia. EXPERIMENTAL APPROACH Surface ECG and ventricular action potentials (APs) were recorded from whole-heart murine Langendorff preparations. Ventricular arrhythmia incidence was compared in hearts perfused with low [K+ ], and those pre-treated with dantrolene or VK-II-86. Whole-cell patch clamping was used in murine and canine ventricular cardiomyocytes to study the effects of dantrolene and VK-II-86 on AP parameters in low [K+ ] and the effects of VK-II-86 on the inward rectifier current (IK1 ), late sodium current (INa_L ) and the L-type Ca2+ current (ICa ). Effects of VK-II-86 on IKr were investigated in transfected HEK-293 cells. A fluorogenic probe quantified the effects of VK-II-86 on oxidative stress in hypokalaemia. KEY RESULTS Dantrolene reduced the incidence of ventricular arrhythmias induced by low [K+ ] in explanted murine hearts by 94%, whereas VK-II-86 prevented all arrhythmias. VK-II-86 prevented hypokalaemia-induced AP prolongation and depolarization, but did not alter AP parameters in normokalaemia. Hypokalaemia was associated with a significant reduction of IK1 and IKr , and increase in INa-L , and ICa . VK-II-86 prevented all hypokalaemia-induced changes in ion channel activity and oxidative stress. CONCLUSIONS AND IMPLICATIONS VK-II-86 prevents hypokalaemia-induced arrhythmogenesis by normalising calcium homeostasis and repolarization reserve. VK-II-86 may provide an exciting treatment in hypokalaemia and other arrhythmias caused by delayed repolarization or Ca2+ overload.
Collapse
Affiliation(s)
- Victoria M Robinson
- The University of Manchester, UK.,Lankenau Institute for Medical Research, Wynnewood, PA, USA
| | | | | | - Charles Antzelevitch
- Lankenau Institute for Medical Research, Wynnewood, PA, USA.,Sidney Kimmel College of Medicine, Thomas Jefferson University, Philadelphia, PA, USA.,Lankenau Heart Institute, Wynnewood, PA, USA
| | | | | |
Collapse
|
27
|
Angelini M, Pezhouman A, Savalli N, Chang MG, Steccanella F, Scranton K, Calmettes G, Ottolia M, Pantazis A, Karagueuzian HS, Weiss JN, Olcese R. Suppression of ventricular arrhythmias by targeting late L-type Ca2+ current. J Gen Physiol 2021; 153:212725. [PMID: 34698805 PMCID: PMC8552156 DOI: 10.1085/jgp.202012584] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 07/15/2021] [Accepted: 09/02/2021] [Indexed: 12/15/2022] Open
Abstract
Ventricular arrhythmias, a leading cause of sudden cardiac death, can be triggered by cardiomyocyte early afterdepolarizations (EADs). EADs can result from an abnormal late activation of L-type Ca2+ channels (LTCCs). Current LTCC blockers (class IV antiarrhythmics), while effective at suppressing EADs, block both early and late components of ICa,L, compromising inotropy. However, computational studies have recently demonstrated that selective reduction of late ICa,L (Ca2+ influx during late phases of the action potential) is sufficient to potently suppress EADs, suggesting that effective antiarrhythmic action can be achieved without blocking the early peak ICa,L, which is essential for proper excitation–contraction coupling. We tested this new strategy using a purine analogue, roscovitine, which reduces late ICa,L with minimal effect on peak current. Scaling our investigation from a human CaV1.2 channel clone to rabbit ventricular myocytes and rat and rabbit perfused hearts, we demonstrate that (1) roscovitine selectively reduces ICa,L noninactivating component in a human CaV1.2 channel clone and in ventricular myocytes native current, (2) the pharmacological reduction of late ICa,L suppresses EADs and EATs (early after Ca2+ transients) induced by oxidative stress and hypokalemia in isolated myocytes, largely preserving cell shortening and normal Ca2+ transient, and (3) late ICa,L reduction prevents/suppresses ventricular tachycardia/fibrillation in ex vivo rabbit and rat hearts subjected to hypokalemia and/or oxidative stress. These results support the value of an antiarrhythmic strategy based on the selective reduction of late ICa,L to suppress EAD-mediated arrhythmias. Antiarrhythmic therapies based on this idea would modify the gating properties of CaV1.2 channels rather than blocking their pore, largely preserving contractility.
Collapse
Affiliation(s)
- Marina Angelini
- Division of Molecular Medicine, Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Arash Pezhouman
- Department of Medicine (Cardiology), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Nicoletta Savalli
- Division of Molecular Medicine, Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Marvin G Chang
- Department of Anesthesia and Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Federica Steccanella
- Division of Molecular Medicine, Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Kyle Scranton
- Division of Molecular Medicine, Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Guillaume Calmettes
- Department of Medicine (Cardiology), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Michela Ottolia
- Division of Molecular Medicine, Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA.,University of California, Los Angeles Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Antonios Pantazis
- Division of Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Wallenberg Center for Molecular Medicine, Linköping University, Linköping, Sweden
| | - Hrayr S Karagueuzian
- Department of Medicine (Cardiology), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA.,Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - James N Weiss
- Department of Medicine (Cardiology), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA.,Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA.,Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Riccardo Olcese
- Division of Molecular Medicine, Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA.,Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA.,University of California, Los Angeles Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA.,Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
28
|
Central Sleep Apnea Is Associated with an Abnormal P-Wave Terminal Force in Lead V 1 in Patients with Acute Myocardial Infarction Independent from Ventricular Function. J Clin Med 2021; 10:jcm10235555. [PMID: 34884253 PMCID: PMC8658572 DOI: 10.3390/jcm10235555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/19/2021] [Accepted: 11/25/2021] [Indexed: 11/17/2022] Open
Abstract
Sleep-disordered breathing (SDB) is highly prevalent in patients with cardiovascular disease. We have recently shown that an elevation of the electrocardiographic (ECG) parameter P wave terminal force in lead V1 (PTFV1) is linked to atrial proarrhythmic activity by stimulation of reactive oxygen species (ROS)-dependent pathways. Since SDB leads to increased ROS generation, we aimed to investigate the relationship between SDB-related hypoxia and PTFV1 in patients with first-time acute myocardial infarction (AMI). We examined 56 patients with first-time AMI. PTFV1 was analyzed in 12-lead ECGs and defined as abnormal when ≥4000 µV*ms. Polysomnography (PSG) to assess SDB was performed within 3–5 days after AMI. SDB was defined by an apnea-hypopnea-index (AHI) >15/h. The multivariable regression analysis showed a significant association between SDB-related hypoxia and the magnitude of PTFV1 independent from other relevant clinical co-factors. Interestingly, this association was mainly driven by central but not obstructive apnea events. Additionally, abnormal PTFV1 was associated with SDB severity (as measured by AHI, B 21.495; CI [10.872 to 32.118]; p < 0.001), suggesting that ECG may help identify patients suitable for SDB screening. Hypoxia as a consequence of central sleep apnea may result in atrial electrical remodeling measured by abnormal PTFV1 in patients with first-time AMI independent of ventricular function. The PTFV1 may be used as a clinical marker for increased SDB risk in cardiovascular patients.
Collapse
|
29
|
Hegner P, Lebek S, Maier LS, Arzt M, Wagner S. The Effect of Gender and Sex Hormones on Cardiovascular Disease, Heart Failure, Diabetes, and Atrial Fibrillation in Sleep Apnea. Front Physiol 2021; 12:741896. [PMID: 34744785 PMCID: PMC8564381 DOI: 10.3389/fphys.2021.741896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/27/2021] [Indexed: 12/29/2022] Open
Abstract
Sleep apnea is a highly prevalent disorder with increasing impact on healthcare systems worldwide. Previous studies have been conducted primarily with male subjects, and prevalence and severity of sleep apnea in women are underestimated. Recent clinical and basic science evidence increasingly points to different mechanisms in men and women with sleep-disordered breathing (SDB). SDB is associated with a variety of comorbidities, including cardiovascular disease, heart failure, diabetes, and atrial fibrillation. In this review, we discuss sex-dependent mechanisms of SDB in select associated conditions to sharpen our clinical understanding of these sex-dependent inherent differences.
Collapse
Affiliation(s)
- Philipp Hegner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Simon Lebek
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Lars Siegfried Maier
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Michael Arzt
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Stefan Wagner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
30
|
Islam MMT, Tarnowski D, Zhang M, Trum M, Lebek S, Mustroph J, Daniel H, Moellencamp J, Pabel S, Sossalla S, El‐Armouche A, Nikolaev VO, Shah AM, Eaton P, Maier LS, Sag CM, Wagner S. Enhanced Heart Failure in Redox-Dead Cys17Ser PKARIα Knock-In Mice. J Am Heart Assoc 2021; 10:e021985. [PMID: 34583520 PMCID: PMC8649132 DOI: 10.1161/jaha.121.021985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Background PKARIα (protein kinase A type I-α regulatory subunit) is redox-active independent of its physiologic agonist cAMP. However, it is unknown whether this alternative mechanism of PKARIα activation may be of relevance to cardiac excitation-contraction coupling. Methods and Results We used a redox-dead transgenic mouse model with homozygous knock-in replacement of redox-sensitive cysteine 17 with serine within the regulatory subunits of PKARIα (KI). Reactive oxygen species were acutely evoked by exposure of isolated cardiac myocytes to AngII (angiotensin II, 1 µmol/L). The long-term relevance of oxidized PKARIα was investigated in KI mice and their wild-type (WT) littermates following transverse aortic constriction (TAC). AngII increased reactive oxygen species in both groups but with RIα dimer formation in WT only. AngII induced translocation of PKARI to the cell membrane and resulted in protein kinase A-dependent stimulation of ICa (L-type Ca current) in WT with no effect in KI myocytes. Consequently, Ca transients were reduced in KI myocytes as compared with WT cells following acute AngII exposure. Transverse aortic constriction-related reactive oxygen species formation resulted in RIα oxidation in WT but not in KI mice. Within 6 weeks after TAC, KI mice showed an enhanced deterioration of contractile function and impaired survival compared with WT. In accordance, compared with WT, ventricular myocytes from failing KI mice displayed significantly reduced Ca transient amplitudes and lack of ICa stimulation. Conversely, direct pharmacological stimulation of ICa using Bay K8644 rescued Ca transients in AngII-treated KI myocytes and contractile function in failing KI mice in vivo. Conclusions Oxidative activation of PKARIα with subsequent stimulation of ICa preserves cardiac function in the setting of acute and chronic oxidative stress.
Collapse
Affiliation(s)
- M. M. Towhidul Islam
- Department of Internal Medicine IIUniversity Medical Center RegensburgRegensburgGermany
- Department of Biochemistry and Molecular BiologyUniversity of DhakaBangladesh
| | - Daniel Tarnowski
- Department of Internal Medicine IIUniversity Medical Center RegensburgRegensburgGermany
| | - Min Zhang
- School of Cardiovascular Medicine & SciencesKings College London British Heart Foundation Centre of ExcellenceLondonUnited Kingdom
| | - Maximilian Trum
- Department of Internal Medicine IIUniversity Medical Center RegensburgRegensburgGermany
| | - Simon Lebek
- Department of Internal Medicine IIUniversity Medical Center RegensburgRegensburgGermany
| | - Julian Mustroph
- Department of Internal Medicine IIUniversity Medical Center RegensburgRegensburgGermany
| | - Henriette Daniel
- Department of Internal Medicine IIUniversity Medical Center RegensburgRegensburgGermany
| | - Johanna Moellencamp
- Department of Internal Medicine IIUniversity Medical Center RegensburgRegensburgGermany
| | - Steffen Pabel
- Department of Internal Medicine IIUniversity Medical Center RegensburgRegensburgGermany
| | - Samuel Sossalla
- Department of Internal Medicine IIUniversity Medical Center RegensburgRegensburgGermany
| | - Ali El‐Armouche
- Department of Pharmacology and ToxicologyTechnical University DresdenDresdenGermany
| | - Viacheslav O. Nikolaev
- Institute of Experimental Cardiovascular ResearchUniversity Medical Center Hamburg‐EppendorfEppendorfGermany
| | - Ajay M. Shah
- School of Cardiovascular Medicine & SciencesKings College London British Heart Foundation Centre of ExcellenceLondonUnited Kingdom
| | - Philip Eaton
- The William Harvey Research InstituteBarts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUnited Kingdom
| | - Lars S. Maier
- Department of Internal Medicine IIUniversity Medical Center RegensburgRegensburgGermany
| | - Can Martin Sag
- Department of Internal Medicine IIUniversity Medical Center RegensburgRegensburgGermany
| | - Stefan Wagner
- Department of Internal Medicine IIUniversity Medical Center RegensburgRegensburgGermany
| |
Collapse
|
31
|
Trum M, Riechel J, Wagner S. Cardioprotection by SGLT2 Inhibitors-Does It All Come Down to Na +? Int J Mol Sci 2021; 22:ijms22157976. [PMID: 34360742 PMCID: PMC8347698 DOI: 10.3390/ijms22157976] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/22/2021] [Accepted: 07/22/2021] [Indexed: 12/15/2022] Open
Abstract
Sodium-glucose co-transporter 2 inhibitors (SGLT2i) are emerging as a new treatment strategy for heart failure with reduced ejection fraction (HFrEF) and—depending on the wistfully awaited results of two clinical trials (DELIVER and EMPEROR-Preserved)—may be the first drug class to improve cardiovascular outcomes in patients suffering from heart failure with preserved ejection fraction (HFpEF). Proposed mechanisms of action of this class of drugs are diverse and include metabolic and hemodynamic effects as well as effects on inflammation, neurohumoral activation, and intracellular ion homeostasis. In this review we focus on the growing body of evidence for SGLT2i-mediated effects on cardiac intracellular Na+ as an upstream mechanism. Therefore, we will first give a short overview of physiological cardiomyocyte Na+ handling and its deterioration in heart failure. On this basis we discuss the salutary effects of SGLT2i on Na+ homeostasis by influencing NHE1 activity, late INa as well as CaMKII activity. Finally, we highlight the potential relevance of these effects for systolic and diastolic dysfunction as well as arrhythmogenesis.
Collapse
|
32
|
Veitch CR, Power AS, Erickson JR. CaMKII Inhibition is a Novel Therapeutic Strategy to Prevent Diabetic Cardiomyopathy. Front Pharmacol 2021; 12:695401. [PMID: 34381362 PMCID: PMC8350113 DOI: 10.3389/fphar.2021.695401] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/14/2021] [Indexed: 11/24/2022] Open
Abstract
Increasing prevalence of diabetes mellitus worldwide has pushed the complex disease state to the foreground of biomedical research, especially concerning its multifaceted impacts on the cardiovascular system. Current therapies for diabetic cardiomyopathy have had a positive impact, but with diabetic patients still suffering from a significantly greater burden of cardiac pathology compared to the general population, the need for novel therapeutic approaches is great. A new therapeutic target, calcium/calmodulin-dependent kinase II (CaMKII), has emerged as a potential treatment option for preventing cardiac dysfunction in the setting of diabetes. Within the last 10 years, new evidence has emerged describing the pathophysiological consequences of CaMKII activation in the diabetic heart, the mechanisms that underlie persistent CaMKII activation, and the protective effects of CaMKII inhibition to prevent diabetic cardiomyopathy. This review will examine recent evidence tying cardiac dysfunction in diabetes to CaMKII activation. It will then discuss the current understanding of the mechanisms by which CaMKII activity is enhanced during diabetes. Finally, it will examine the benefits of CaMKII inhibition to treat diabetic cardiomyopathy, including contractile dysfunction, heart failure with preserved ejection fraction, and arrhythmogenesis. We intend this review to serve as a critical examination of CaMKII inhibition as a therapeutic strategy, including potential drawbacks of this approach.
Collapse
Affiliation(s)
- Christopher R Veitch
- Department of Physiology and HeartOtago, University of Otago, Dunedin, New Zealand
| | - Amelia S Power
- Department of Physiology and HeartOtago, University of Otago, Dunedin, New Zealand
| | - Jeffrey R Erickson
- Department of Physiology and HeartOtago, University of Otago, Dunedin, New Zealand
| |
Collapse
|
33
|
Ca 2+/calmodulin kinase II-dependent regulation of β IV-spectrin modulates cardiac fibroblast gene expression, proliferation, and contractility. J Biol Chem 2021; 297:100893. [PMID: 34153319 PMCID: PMC8294584 DOI: 10.1016/j.jbc.2021.100893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 06/10/2021] [Accepted: 06/17/2021] [Indexed: 01/26/2023] Open
Abstract
Fibrosis is a pronounced feature of heart disease and the result of dysregulated activation of resident cardiac fibroblasts (CFs). Recent work identified stress-induced degradation of the cytoskeletal protein βIV-spectrin as an important step in CF activation and cardiac fibrosis. Furthermore, loss of βIV-spectrin was found to depend on Ca2+/calmodulin-dependent kinase II (CaMKII). Therefore, we sought to determine the mechanism for CaMKII-dependent regulation of βIV-spectrin and CF activity. Computational screening and MS revealed a critical serine residue (S2250 in mouse and S2254 in human) in βIV-spectrin phosphorylated by CaMKII. Disruption of βIV-spectrin/CaMKII interaction or alanine substitution of βIV-spectrin Ser2250 (βIV-S2254A) prevented CaMKII-induced degradation, whereas a phosphomimetic construct (βIV-spectrin with glutamic acid substitution at serine 2254 [βIV-S2254E]) showed accelerated degradation in the absence of CaMKII. To assess the physiological significance of this phosphorylation event, we expressed exogenous βIV-S2254A and βIV-S2254E constructs in βIV-spectrin-deficient CFs, which have increased proliferation and fibrotic gene expression compared with WT CFs. βIV-S2254A but not βIV-S2254E normalized CF proliferation, gene expression, and contractility. Pathophysiological targeting of βIV-spectrin phosphorylation and subsequent degradation was identified in CFs activated with the profibrotic ligand angiotensin II, resulting in increased proliferation and signal transducer and activation of transcription 3 nuclear accumulation. While therapeutic delivery of exogenous WT βIV-spectrin partially reversed these trends, βIV-S2254A completely negated increased CF proliferation and signal transducer and activation of transcription 3 translocation. Moreover, we observed βIV-spectrin phosphorylation and associated loss in total protein within human heart tissue following heart failure. Together, these data illustrate a considerable role for the βIV-spectrin/CaMKII interaction in activating profibrotic signaling.
Collapse
|
34
|
Liu Y, Shao Q, Cheng HJ, Li T, Zhang X, Callahan MF, Herrington D, Kitzman D, Zhao D, Cheng CP. Chronic Ca 2+/Calmodulin-Dependent Protein Kinase II Inhibition Rescues Advanced Heart Failure. J Pharmacol Exp Ther 2021; 377:316-325. [PMID: 33722881 PMCID: PMC8140392 DOI: 10.1124/jpet.120.000361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/11/2021] [Indexed: 11/22/2022] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) is upregulated in congestive heart failure (CHF), contributing to electrical, structural, and functional remodeling. CaMKII inhibition is known to improve CHF, but its direct cardiac effects in CHF remain unclear. We hypothesized that CaMKII inhibition improves cardiomyocyte function, [Ca2+]i regulation, and β-adrenergic reserve, thus improving advanced CHF. In a 16-week study, we compared plasma neurohormonal levels and left ventricular (LV)- and myocyte-functional and calcium transient ([Ca2+]iT) responses in male Sprague-Dawley rats (10/group) with CHF induced by isoproterenol (170 mg/kg sq for 2 days). In rats with CHF, we studied the effects of the CaMKII inhibitor KN-93 or its inactive analog KN-92 (n = 4) (70 µg/kg per day, mini-pump) for 4 weeks. Compared with controls, isoproterenol-treated rats had severe CHF with 5-fold-increased plasma norepinephrine and about 50% decreases in ejection fraction (EF) and LV contractility [slope of LV end-systolic pressure-LV end-systolic volume relation (EES)] but increased time constant of LV relaxation (τ). They also showed significantly reduced myocyte contraction [maximum rate of myocyte shortening (dL/dtmax)], relaxation (dL/dtmax), and [Ca2+]iT Isoproterenol superfusion caused significantly fewer increases in dL/dtmax and [Ca2+]iT KN-93 treatment prevented plasma norepinephrine elevation, with increased basal and acute isoproterenol-stimulated increases in EF and EES and decreased τ in CHF. KN-93 treatment preserved normal myocyte contraction, relaxation, [Ca2+]iT, and β-adrenergic reserve, whereas KN-92 treatment failed to improve LV and myocyte function, and plasma norepinephrine remained high in CHF. Thus, chronic CaMKII inhibition prevented CHF-induced activation of the sympathetic nervous system, restoring normal LV and cardiomyocyte basal and β-adrenergic-stimulated contraction, relaxation, and [Ca2+]iT, thereby playing a rescue role in advanced CHF. SIGNIFICANCE STATEMENT: We investigated the therapeutic efficacy of late initiation of chronic Ca2+/calmodulin-dependent protein kinase II (CaMKII) inhibition on progression of advanced congestive heart failure (CHF). Chronic CaMKII inhibition prevented CHF-induced activation of the sympathetic nervous system and restored normal intrinsic cardiomyocyte basal and β-adrenergic receptor-stimulated relaxation, contraction, and [Ca2+]i regulation, leading to reversal of CHF progression. These data provide new evidence that CaMKII inhibition is able and sufficient to rescue a failing heart, and thus cardiac CaMKII inhibition is a promising target for improving CHF treatment.
Collapse
Affiliation(s)
- Yixi Liu
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - Qun Shao
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - Heng-Jie Cheng
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - Tiankai Li
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - Xiaowei Zhang
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - Michael F Callahan
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - David Herrington
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - Dalane Kitzman
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - David Zhao
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - Che-Ping Cheng
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| |
Collapse
|
35
|
Kolodziej F, O’Halloran KD. Re-Evaluating the Oxidative Phenotype: Can Endurance Exercise Save the Western World? Antioxidants (Basel) 2021; 10:609. [PMID: 33921022 PMCID: PMC8071436 DOI: 10.3390/antiox10040609] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/06/2021] [Accepted: 04/10/2021] [Indexed: 01/16/2023] Open
Abstract
Mitochondria are popularly called the "powerhouses" of the cell. They promote energy metabolism through the tricarboxylic acid (TCA) cycle and oxidative phosphorylation, which in contrast to cytosolic glycolysis are oxygen-dependent and significantly more substrate efficient. That is, mitochondrial metabolism provides substantially more cellular energy currency (ATP) per macronutrient metabolised. Enhancement of mitochondrial density and metabolism are associated with endurance training, which allows for the attainment of high relative VO2 max values. However, the sedentary lifestyle and diet currently predominant in the Western world lead to mitochondrial dysfunction. Underdeveloped mitochondrial metabolism leads to nutrient-induced reducing pressure caused by energy surplus, as reduced nicotinamide adenine dinucleotide (NADH)-mediated high electron flow at rest leads to "electron leak" and a chronic generation of superoxide radicals (O2-). Chronic overload of these reactive oxygen species (ROS) damages cell components such as DNA, cell membranes, and proteins. Counterintuitively, transiently generated ROS during exercise contributes to adaptive reduction-oxidation (REDOX) signalling through the process of cellular hormesis or "oxidative eustress" defined by Helmut Sies. However, the unaccustomed, chronic oxidative stress is central to the leading causes of mortality in the 21st century-metabolic syndrome and the associated cardiovascular comorbidities. The endurance exercise training that improves mitochondrial capacity and the protective antioxidant cellular system emerges as a universal intervention for mitochondrial dysfunction and resultant comorbidities. Furthermore, exercise might also be a solution to prevent ageing-related degenerative diseases, which are caused by impaired mitochondrial recycling. This review aims to break down the metabolic components of exercise and how they translate to athletic versus metabolically diseased phenotypes. We outline a reciprocal relationship between oxidative metabolism and inflammation, as well as hypoxia. We highlight the importance of oxidative stress for metabolic and antioxidant adaptation. We discuss the relevance of lactate as an indicator of critical exercise intensity, and inferring from its relationship with hypoxia, we suggest the most appropriate mode of exercise for the case of a lost oxidative identity in metabolically inflexible patients. Finally, we propose a reciprocal signalling model that establishes a healthy balance between the glycolytic/proliferative and oxidative/prolonged-ageing phenotypes. This model is malleable to adaptation with oxidative stress in exercise but is also susceptible to maladaptation associated with chronic oxidative stress in disease. Furthermore, mutations of components involved in the transcriptional regulatory mechanisms of mitochondrial metabolism may lead to the development of a cancerous phenotype, which progressively presents as one of the main causes of death, alongside the metabolic syndrome.
Collapse
Affiliation(s)
- Filip Kolodziej
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, T12 XF62 Cork, Ireland;
| | | |
Collapse
|
36
|
Shen F, Xu X, Yu Z, Li H, Shen H, Li X, Shen M, Chen G. Rbfox-1 contributes to CaMKIIα expression and intracerebral hemorrhage-induced secondary brain injury via blocking micro-RNA-124. J Cereb Blood Flow Metab 2021; 41:530-545. [PMID: 32248729 PMCID: PMC7922744 DOI: 10.1177/0271678x20916860] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RNA-binding protein fox-1 homolog 1 (Rbfox-1), an RNA-binding protein in neurons, is thought to be associated with many neurological diseases. To date, the mechanism on which Rbfox-1 worsens secondary cell death in ICH remains poorly understood. In this study, we aimed to explore the role of Rbfox-1 in intracerebral hemorrhage (ICH)-induced secondary brain injury (SBI) and to identify its underlying mechanisms. We found that the expression of Rbfox-1 in neurons was significantly increased after ICH, which was accompanied by increases in the binding of Rbfox-1 to Ca2+/calmodulin-dependent protein kinase II (CaMKIIα) mRNA and the protein level of CaMKIIα. In addition, when exposed to exogenous upregulation or downregulation of Rbfox-1, the protein level of CaMKIIα showed a concomitant trend in brain tissue, which further suggested that CaMKIIα is a downstream-target protein of Rbfox-1. The upregulation of both proteins caused intracellular-Ca2+ overload and neuronal degeneration, which exacerbated brain damage. Furthermore, we found that Rbfox-1 promoted the expression of CaMKIIα via blocking the binding of micro-RNA-124 to CaMKIIα mRNA. Thus, Rbfox-1 is expected to be a promising therapeutic target for SBI after ICH.
Collapse
Affiliation(s)
- Fang Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China.,School of Nursing, Medical College of Soochow University, Suzhou, China
| | - Xiang Xu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhengquan Yu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meifen Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China.,School of Nursing, Medical College of Soochow University, Suzhou, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
37
|
Mustroph J, Sag CM, Bähr F, Schmidtmann AL, Gupta SN, Dietz A, Islam MMT, Lücht C, Beuthner BE, Pabel S, Baier MJ, El-Armouche A, Sossalla S, Anderson ME, Möllmann J, Lehrke M, Marx N, Mohler PJ, Bers DM, Unsöld B, He T, Dewenter M, Backs J, Maier LS, Wagner S. Loss of CASK Accelerates Heart Failure Development. Circ Res 2021; 128:1139-1155. [PMID: 33593074 DOI: 10.1161/circresaha.120.318170] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Julian Mustroph
- Department of Internal Medicine II, University Medical Center Regensburg, Germany (J. Mustroph, C.M.S., C.L., S.P., M.J.B., S.S., B.U., L.S.M., S.W.)
| | - Can M Sag
- Department of Internal Medicine II, University Medical Center Regensburg, Germany (J. Mustroph, C.M.S., C.L., S.P., M.J.B., S.S., B.U., L.S.M., S.W.)
| | - Felix Bähr
- Cardiology & Pneumology, University Medical Center Göttingen, Germany (F.B., A.-L.S., S.N.G., A.D., M.M.T.I., B.E.B., S.S.)
| | - Anna-Lena Schmidtmann
- Cardiology & Pneumology, University Medical Center Göttingen, Germany (F.B., A.-L.S., S.N.G., A.D., M.M.T.I., B.E.B., S.S.)
| | - Shamindra N Gupta
- Cardiology & Pneumology, University Medical Center Göttingen, Germany (F.B., A.-L.S., S.N.G., A.D., M.M.T.I., B.E.B., S.S.)
| | - Alexander Dietz
- Cardiology & Pneumology, University Medical Center Göttingen, Germany (F.B., A.-L.S., S.N.G., A.D., M.M.T.I., B.E.B., S.S.)
| | - M M Towhidul Islam
- Cardiology & Pneumology, University Medical Center Göttingen, Germany (F.B., A.-L.S., S.N.G., A.D., M.M.T.I., B.E.B., S.S.)
| | - Charlotte Lücht
- Department of Internal Medicine II, University Medical Center Regensburg, Germany (J. Mustroph, C.M.S., C.L., S.P., M.J.B., S.S., B.U., L.S.M., S.W.)
| | - Bo Eric Beuthner
- Cardiology & Pneumology, University Medical Center Göttingen, Germany (F.B., A.-L.S., S.N.G., A.D., M.M.T.I., B.E.B., S.S.)
| | - Steffen Pabel
- Department of Internal Medicine II, University Medical Center Regensburg, Germany (J. Mustroph, C.M.S., C.L., S.P., M.J.B., S.S., B.U., L.S.M., S.W.)
| | - Maria J Baier
- Department of Internal Medicine II, University Medical Center Regensburg, Germany (J. Mustroph, C.M.S., C.L., S.P., M.J.B., S.S., B.U., L.S.M., S.W.)
| | - Ali El-Armouche
- Department of Pharmacology and Toxicology, Technical University Dresden, Germany (A.E.-A.)
| | - Samuel Sossalla
- Department of Internal Medicine II, University Medical Center Regensburg, Germany (J. Mustroph, C.M.S., C.L., S.P., M.J.B., S.S., B.U., L.S.M., S.W.).,Cardiology & Pneumology, University Medical Center Göttingen, Germany (F.B., A.-L.S., S.N.G., A.D., M.M.T.I., B.E.B., S.S.)
| | | | - Julia Möllmann
- Clinic for Cardiology, Angiology, and Internal Intensive Care, University Clinic Aachen, Germany (J. Möllmann, M.L., N.M.)
| | - Michael Lehrke
- Clinic for Cardiology, Angiology, and Internal Intensive Care, University Clinic Aachen, Germany (J. Möllmann, M.L., N.M.)
| | - Nikolaus Marx
- Clinic for Cardiology, Angiology, and Internal Intensive Care, University Clinic Aachen, Germany (J. Möllmann, M.L., N.M.)
| | - Peter J Mohler
- College of Medicine, the Ohio State University Wexner Medical Center, Columbus (P.J.M.)
| | - Donald M Bers
- College of Biological Sciences, University of California at Davis (D.M.B.)
| | - Bernhard Unsöld
- Department of Internal Medicine II, University Medical Center Regensburg, Germany (J. Mustroph, C.M.S., C.L., S.P., M.J.B., S.S., B.U., L.S.M., S.W.)
| | - Tao He
- Department of Molecular Cardiology and Epigenetics, University Clinic Heidelberg, Germany (T.H., M.D., J.B.)
| | - Matthias Dewenter
- Department of Molecular Cardiology and Epigenetics, University Clinic Heidelberg, Germany (T.H., M.D., J.B.)
| | - Johannes Backs
- Department of Molecular Cardiology and Epigenetics, University Clinic Heidelberg, Germany (T.H., M.D., J.B.)
| | - Lars S Maier
- Department of Internal Medicine II, University Medical Center Regensburg, Germany (J. Mustroph, C.M.S., C.L., S.P., M.J.B., S.S., B.U., L.S.M., S.W.)
| | - Stefan Wagner
- Department of Internal Medicine II, University Medical Center Regensburg, Germany (J. Mustroph, C.M.S., C.L., S.P., M.J.B., S.S., B.U., L.S.M., S.W.)
| |
Collapse
|
38
|
Saadeh K, Fazmin IT. Mitochondrial Dysfunction Increases Arrhythmic Triggers and Substrates; Potential Anti-arrhythmic Pharmacological Targets. Front Cardiovasc Med 2021; 8:646932. [PMID: 33659284 PMCID: PMC7917191 DOI: 10.3389/fcvm.2021.646932] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 01/26/2021] [Indexed: 12/31/2022] Open
Abstract
Incidence of cardiac arrhythmias increases significantly with age. In order to effectively stratify arrhythmic risk in the aging population it is crucial to elucidate the relevant underlying molecular mechanisms. The changes underlying age-related electrophysiological disruption appear to be closely associated with mitochondrial dysfunction. Thus, the present review examines the mechanisms by which age-related mitochondrial dysfunction promotes arrhythmic triggers and substrate. Namely, via alterations in plasmalemmal ionic currents (both sodium and potassium), gap junctions, cellular Ca2+ homeostasis, and cardiac fibrosis. Stratification of patients' mitochondrial function status permits application of appropriate anti-arrhythmic therapies. Here, we discuss novel potential anti-arrhythmic pharmacological interventions that specifically target upstream mitochondrial function and hence ameliorates the need for therapies targeting downstream changes which have constituted traditional antiarrhythmic therapy.
Collapse
Affiliation(s)
- Khalil Saadeh
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom.,Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Ibrahim Talal Fazmin
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom.,Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom.,Royal Papworth Hospital NHS Foundation Trust, Cambridge, United Kingdom
| |
Collapse
|
39
|
Protective Role of Polyphenols in Heart Failure: Molecular Targets and Cellular Mechanisms Underlying Their Therapeutic Potential. Int J Mol Sci 2021; 22:ijms22041668. [PMID: 33562294 PMCID: PMC7914665 DOI: 10.3390/ijms22041668] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) is a leading cause of death in the United States, with a 5-year mortality rate of 50% despite modern pharmacological therapies. Plant-based diets are comprised of a diverse polyphenol profile, which lends to their association with reduced cardiovascular disease risk. Whether a polyphenol-rich diet can slow the progression of or reverse HF in humans is not known. To date, in vitro and in vivo studies have reported on the protective role of polyphenols in HF. In this review, we will discuss the major mechanisms by which polyphenols mitigate HF in vitro and in vivo, including (1) reduced cardiac inflammation and oxidative stress, (2) reduced mitochondrial dysfunction, (3) improved Ca2+ homeostasis, (4) increased survival signaling, and (5) increased sirtuin 1 activity.
Collapse
|
40
|
Mesubi OO, Rokita AG, Abrol N, Wu Y, Chen B, Wang Q, Granger JM, Tucker-Bartley A, Luczak ED, Murphy KR, Umapathi P, Banerjee PS, Boronina TN, Cole RN, Maier LS, Wehrens XH, Pomerantz JL, Song LS, Ahima RS, Hart GW, Zachara NE, Anderson ME. Oxidized CaMKII and O-GlcNAcylation cause increased atrial fibrillation in diabetic mice by distinct mechanisms. J Clin Invest 2021; 131:95747. [PMID: 33151911 PMCID: PMC7810480 DOI: 10.1172/jci95747] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 11/03/2020] [Indexed: 12/21/2022] Open
Abstract
Diabetes mellitus (DM) and atrial fibrillation (AF) are major unsolved public health problems, and diabetes is an independent risk factor for AF. However, the mechanism(s) underlying this clinical association is unknown. ROS and protein O-GlcNAcylation (OGN) are increased in diabetic hearts, and calmodulin kinase II (CaMKII) is a proarrhythmic signal that may be activated by ROS (oxidized CaMKII, ox-CaMKII) and OGN (OGN-CaMKII). We induced type 1 (T1D) and type 2 DM (T2D) in a portfolio of genetic mouse models capable of dissecting the role of ROS and OGN at CaMKII and global OGN in diabetic AF. Here, we showed that T1D and T2D significantly increased AF, and this increase required CaMKII and OGN. T1D and T2D both required ox-CaMKII to increase AF; however, we did not detect OGN-CaMKII or a role for OGN-CaMKII in diabetic AF. Collectively, our data affirm CaMKII as a critical proarrhythmic signal in diabetic AF and suggest ROS primarily promotes AF by ox-CaMKII, while OGN promotes AF by a CaMKII-independent mechanism(s). These results provide insights into the mechanisms for increased AF in DM and suggest potential benefits for future CaMKII and OGN targeted therapies.
Collapse
Affiliation(s)
- Olurotimi O. Mesubi
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Adam G. Rokita
- Division of Cardiovascular Medicine and Cardiovascular Research Center, Carver College of Medicine, Iowa City, Iowa, USA
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Neha Abrol
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yuejin Wu
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Biyi Chen
- Division of Cardiovascular Medicine and Cardiovascular Research Center, Carver College of Medicine, Iowa City, Iowa, USA
| | - Qinchuan Wang
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jonathan M. Granger
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Anthony Tucker-Bartley
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Elizabeth D. Luczak
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kevin R. Murphy
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Priya Umapathi
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Partha S. Banerjee
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tatiana N. Boronina
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Robert N. Cole
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lars S. Maier
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Xander H. Wehrens
- Department of Molecular Physiology and Biophysics, Department of Medicine (Cardiology), Department of Pediatrics, and Center for Space Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Joel L. Pomerantz
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Institute for Cell Engineering
| | - Long-Sheng Song
- Division of Cardiovascular Medicine and Cardiovascular Research Center, Carver College of Medicine, Iowa City, Iowa, USA
| | - Rexford S. Ahima
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine
| | - Gerald W. Hart
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Natasha E. Zachara
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mark E. Anderson
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, and
- Department of Physiology and Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
41
|
Wang W, Shen W, Zhang S, Luo G, Wang K, Xu Y, Zhang H. The Role of CaMKII Overexpression and Oxidation in Atrial Fibrillation-A Simulation Study. Front Physiol 2021; 11:607809. [PMID: 33391023 PMCID: PMC7775483 DOI: 10.3389/fphys.2020.607809] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/19/2020] [Indexed: 11/13/2022] Open
Abstract
This simulation study aims to investigate how the Calcium/calmodulin-dependent protein kinase II (CaMKII) overexpression and oxidation would influence the cardiac electrophysiological behavior and its arrhythmogenic mechanism in atria. A new-built CaMKII oxidation module and a refitted CaMKII overexpression module were integrated into a mouse atrial cell model for analyzing cardiac electrophysiological variations in action potential (AP) characteristics and intracellular Ca2+ cycling under different conditions. Simulation results showed that CaMKII overexpression significantly increased the phosphorylation level of its downstream target proteins, resulting in prolonged AP and smaller calcium transient amplitude, and impaired the Ca2+ cycling stability. These effects were exacerbated by extra reactive oxygen species, which oxidized CaMKII and led to continuous high CaMKII activation in both systolic and diastolic phases. Intracellular Ca2+ depletion and sustained delayed afterdepolarizations (DADs) were observed under co-existing CaMKII overexpression and oxidation, which could be effectively reversed by clamping the phosphorylation level of ryanodine receptor (RyR). We also found that the stability of RyR release highly depended on a delicate balance between the level of RyR phosphorylation and sarcoplasmic reticulum Ca2+ concentration, which was closely related to the genesis of DADs. We concluded that the CaMKII overexpression and oxidation have a synergistic role in increasing the activity of CaMKII, and the unstable RyR may be the key downstream target in the CaMKII arrhythmogenic mechanism. Our simulation provides detailed mechanistic insights into the arrhythmogenic effect of CaMKII overexpression and oxidation, which suggests CaMKII as a promising target in the therapy of atrial fibrillation.
Collapse
Affiliation(s)
- Wei Wang
- Shenzhen Key Laboratory of Visual Object Detection and Recognition, Harbin Institute of Technology, Shenzhen, China.,Peng Cheng Lab, Shenzhen, China
| | - Weijian Shen
- Biological Physics Group, School of Physics and Astronomy, The University of Manchester, Manchester, United Kingdom
| | - Shanzhuo Zhang
- Department of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Gongning Luo
- Department of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Kuanquan Wang
- Department of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Yong Xu
- Shenzhen Key Laboratory of Visual Object Detection and Recognition, Harbin Institute of Technology, Shenzhen, China
| | - Henggui Zhang
- Peng Cheng Lab, Shenzhen, China.,Biological Physics Group, School of Physics and Astronomy, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
42
|
Liu Q, Sun J, Zhang L, Xu Y, Wu B, Cao J. The Agonist of Inward Rectifier Potassium Channel (I K1) Attenuates Rat Reperfusion Arrhythmias Linked to CaMKII Signaling. Int Heart J 2021; 62:1348-1357. [PMID: 34853227 DOI: 10.1536/ihj.21-379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Inward rectifier potassium channels (IK1, Kir) are known to play critical roles in arrhythmogenesis. Thus, how IK1 agonist affects reperfusion arrhythmias needs to be clarified, and its underlying mechanisms should be determined. Reperfusion arrhythmias were modeled by coronary ligation (ischemia, 15 minutes) and release (reperfusion, 15 minutes). Zacopride (1.5-50 μg/kg in vivo, or 0.1-10 μmol/Lex vivo) was applied in the settings of pretreatment (3 minutes before coronary ligation) and posttreatment (5 minutes after coronary ligation). Hypoxia (45 minutes) /reoxygenation (30 minutes) model was established in cultured H9c2 (2-1) cardiomyocytes. Zacopride or KN93 was applied before hypoxia (pretreatment). In the setting of pre- or posttreatment, zacopride at 15 μg/kg in vivo or 1 μmol/Lin vitro exhibited superlative protections on reperfusion arrhythmias or intracellular calcium overload. Western blot data from ex vivo hearts or H9c2 (2-1) cardiomyocytes showed that I/R (H/R) induced the inhibition of Kir2.1 (the dominant subunit of IK1 channel in ventricle), phosphorylation and oxidation of CaMKII, downregulation of SERCA2, phosphorylation of phospholamban (at Thr17), and activation of caspase-3. Zacopride treatment (1 μmol/L) was noted to strikingly restore the expression of Kir2.1 and SERCA2 and decrease the activity of CaMKII, phospholamban, and caspase-3. These effects were largely eliminated by co-application of IK1 blocker BaCl2. CaMKII inhibitor KN93 attenuated calcium overload and p-PLB (Thr17) in an IK1-independent manner. IK1-depedent inhibition of CaMKII activity is found to be a key cardiac salvage signaling under Ca2+ dyshomeostasis and reactive oxygen species (ROS) stress. IK1 might be a novel target for pharmacological conditioning of reperfusion arrhythmia, especially for the application after unpredictable ischemia.
Collapse
Affiliation(s)
- Qinghua Liu
- Department of Pathophysiology, Shanxi Medical University
| | - Jiaxing Sun
- Department of Pathophysiology, Shanxi Medical University
| | - Lijun Zhang
- Department of Pathophysiology, Shanxi Medical University
| | - Yanwu Xu
- Department of Biochemistry, Shanghai University of Traditional Chinese Medicine
| | - Bowei Wu
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of Physiology, Shanxi Medical University
| | - Jimin Cao
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of Physiology, Shanxi Medical University
| |
Collapse
|
43
|
Hegyi B, Pölönen RP, Hellgren KT, Ko CY, Ginsburg KS, Bossuyt J, Mercola M, Bers DM. Cardiomyocyte Na + and Ca 2+ mishandling drives vicious cycle involving CaMKII, ROS, and ryanodine receptors. Basic Res Cardiol 2021; 116:58. [PMID: 34648073 PMCID: PMC8516771 DOI: 10.1007/s00395-021-00900-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/08/2021] [Accepted: 09/30/2021] [Indexed: 12/19/2022]
Abstract
Cardiomyocyte Na+ and Ca2+ mishandling, upregulated Ca2+/calmodulin-dependent kinase II (CaMKII), and increased reactive oxygen species (ROS) are characteristics of various heart diseases, including heart failure (HF), long QT (LQT) syndrome, and catecholaminergic polymorphic ventricular tachycardia (CPVT). These changes may form a vicious cycle of positive feedback to promote cardiac dysfunction and arrhythmias. In HF rabbit cardiomyocytes investigated in this study, the inhibition of CaMKII, late Na+ current (INaL), and leaky ryanodine receptors (RyRs) all attenuated the prolongation and increased short-term variability (STV) of action potential duration (APD), but in age-matched controls these inhibitors had no or minimal effects. In control cardiomyocytes, we enhanced RyR leak (by low [caffeine] plus isoproterenol mimicking CPVT) which markedly increased STV and delayed afterdepolarizations (DADs). These proarrhythmic changes were significantly attenuated by both CaMKII inhibition and mitochondrial ROS scavenging, with a slight synergy with INaL inhibition. Inducing LQT by elevating INaL (by Anemone toxin II, ATX-II) caused markedly prolonged APD, increased STV, and early afterdepolarizations (EADs). Those proarrhythmic ATX-II effects were largely attenuated by mitochondrial ROS scavenging, and partially reduced by inhibition of CaMKII and pathological leaky RyRs using dantrolene. In human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) bearing LQT3 mutation SCN5A N406K, dantrolene significantly attenuated cell arrhythmias and APD prolongation. Targeting critical components of the Na+-Ca2+-CaMKII-ROS-INaL arrhythmogenic vicious cycle may exhibit important on-target and also trans-target effects (e.g., INaL and RyR inhibition can alter INaL-mediated LQT3 effects). Incorporating this vicious cycle into therapeutic strategies provides novel integrated insight for treating cardiac arrhythmias and diseases.
Collapse
Affiliation(s)
- Bence Hegyi
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| | - Risto-Pekka Pölönen
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA ,grid.168010.e0000000419368956Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305 USA
| | - Kim T. Hellgren
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| | - Christopher Y. Ko
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| | - Kenneth S. Ginsburg
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| | - Julie Bossuyt
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| | - Mark Mercola
- grid.168010.e0000000419368956Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305 USA
| | - Donald M. Bers
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| |
Collapse
|
44
|
A novel mouse model of obstructive sleep apnea by bulking agent-induced tongue enlargement results in left ventricular contractile dysfunction. PLoS One 2020; 15:e0243844. [PMID: 33301470 PMCID: PMC7728202 DOI: 10.1371/journal.pone.0243844] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 11/28/2020] [Indexed: 01/24/2023] Open
Abstract
AIMS Obstructive sleep apnea (OSA) is a widespread disease with high global socio-economic impact. However, detailed pathomechanisms are still unclear, partly because current animal models of OSA do not simulate spontaneous airway obstruction. We tested whether polytetrafluoroethylene (PTFE) injection into the tongue induces spontaneous obstructive apneas. METHODS AND RESULTS PTFE (100 μl) was injected into the tongue of 31 male C57BL/6 mice and 28 mice were used as control. Spontaneous apneas and inspiratory flow limitations were recorded by whole-body plethysmography and mRNA expression of the hypoxia marker KDM6A was quantified by qPCR. Left ventricular function was assessed by echocardiography and ventricular CaMKII expression was measured by Western blotting. After PTFE injection, mice showed features of OSA such as significantly increased tongue diameters that were associated with significantly and sustained increased frequencies of inspiratory flow limitations and apneas. Decreased KDM6A mRNA levels indicated chronic hypoxemia. 8 weeks after surgery, PTFE-treated mice showed a significantly reduced left ventricular ejection fraction. Moreover, the severity of diastolic dysfunction (measured as E/e') correlated significantly with the frequency of apneas. Accordingly, CaMKII expression was significantly increased in PTFE mice and correlated significantly with the frequency of apneas. CONCLUSIONS We describe here the first mouse model of spontaneous inspiratory flow limitations, obstructive apneas, and hypoxia by tongue enlargement due to PTFE injection. These mice develop systolic and diastolic dysfunction and increased CaMKII expression. This mouse model offers great opportunities to investigate the effects of obstructive apneas.
Collapse
|
45
|
Trum M, Islam MMT, Lebek S, Baier M, Hegner P, Eaton P, Maier LS, Wagner S. Inhibition of cardiac potassium currents by oxidation-activated protein kinase A contributes to early afterdepolarizations in the heart. Am J Physiol Heart Circ Physiol 2020; 319:H1347-H1357. [PMID: 33035439 PMCID: PMC7792712 DOI: 10.1152/ajpheart.00182.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) have been shown to prolong cardiac action potential duration resulting in afterdepolarizations, the cellular basis of triggered arrhythmias. As previously shown, protein kinase A type I (PKA I) is readily activated by oxidation of its regulatory subunits. However, the relevance of this mechanism of activation for cardiac pathophysiology is still elusive. In this study, we investigated the effects of oxidation-activated PKA I on cardiac electrophysiology. Ventricular cardiomyocytes were isolated from redox-dead PKA-RI Cys17Ser knock-in (KI) and wild-type (WT) mice and exposed to H2O2 (200 µmol/L) or vehicle (Veh) solution. In WT myocytes, exposure to H2O2 significantly increased oxidation of the regulatory subunit I (RI) and thus its dimerization (threefold increase in PKA RI dimer). Whole cell current clamp and voltage clamp were used to measure cardiac action potentials (APs), transient outward potassium current (Ito) and inward rectifying potassium current (IK1), respectively. In WT myocytes, H2O2 exposure significantly prolonged AP duration due to significantly decreased Ito and IK1 resulting in frequent early afterdepolarizations (EADs). Preincubation with the PKA-specific inhibitor Rp-8-Br-cAMPS (10 µmol/L) completely abolished the H2O2-dependent decrease in Ito and IK1 in WT myocytes. Intriguingly, H2O2 exposure did not prolong AP duration, nor did it decrease Ito, and only slightly enhanced EAD frequency in KI myocytes. Treatment of WT and KI cardiomyocytes with the late INa inhibitor TTX (1 µmol/L) completely abolished EAD formation. Our results suggest that redox-activated PKA may be important for H2O2-dependent arrhythmias and could be important for the development of specific antiarrhythmic drugs.NEW & NOTEWORTHY Oxidation-activated PKA type I inhibits transient outward potassium current (Ito) and inward rectifying potassium current (IK1) and contributes to ROS-induced APD prolongation as well as generation of early afterdepolarizations in murine ventricular cardiomyocytes.
Collapse
Affiliation(s)
- M. Trum
- 1Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - M. M. T. Islam
- 2Department of Biochemistry and Molecular Biology, University of Dhaka, Bangladesh
- 3Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - S. Lebek
- 1Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - M. Baier
- 1Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - P. Hegner
- 1Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - P. Eaton
- 4The William Harvey Research Institute, Charterhouse Square, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - L. S. Maier
- 1Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - S. Wagner
- 1Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
46
|
Song L, Zhang ZF, Hu LK, Zhang PH, Cao ZZ, Liu ZP, Zhang PP, Ma JH. Curcumin, a Multi-Ion Channel Blocker That Preferentially Blocks Late Na + Current and Prevents I/R-Induced Arrhythmias. Front Physiol 2020; 11:978. [PMID: 32973546 PMCID: PMC7472421 DOI: 10.3389/fphys.2020.00978] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 07/20/2020] [Indexed: 11/13/2022] Open
Abstract
Increasing evidence shows that Curcumin (Cur) has a protective effect against cardiovascular diseases. However, the role of Cur in the electrophysiology of cardiomyocytes is currently not entirely understood. Therefore, the present study was conducted to investigate the effects of Cur on the action potential and transmembrane ion currents in rabbit ventricular myocytes to explore its antiarrhythmic property. The whole-cell patch clamp was used to record the action potential and ion currents, while the multichannel acquisition and analysis system was used to synchronously record the electrocardiogram and monophasic action potential. The results showed that 30 μmol/L Cur shortened the 50 and 90% repolarization of action potential by 17 and 7%, respectively. In addition, Cur concentration dependently inhibited the Late-sodium current (I Na.L), Transient-sodium current (I Na.T), L-type calcium current (I Ca.L), and Rapidly delayed rectifying potassium current (I Kr), with IC50 values of 7.53, 398.88, 16.66, and 9.96 μmol/L, respectively. Importantly, the inhibitory effect of Cur on I Na.L was 52.97-fold higher than that of I Na.T. Moreover, Cur decreased ATX II-prolonged APD, suppressed the ATX II-induced early afterdepolarization (EAD) and Ca2+-induced delayed afterdepolarization (DAD) in ventricular myocytes, and reduced the occurrence and average duration of ventricular tachycardias and ventricular fibrillations induced by ischemia-reperfusion injury. In conclusion, Cur inhibited I Na.L, I Na.T, I Ca.L, and I Kr; shortened APD; significantly suppressed EAD and DAD-like arrhythmogenic activities at the cellular level; and exhibited antiarrhythmic effect at the organ level. It is first revealed that Cur is a multi-ion channel blocker that preferentially blocks I Na.L and may have potential antiarrhythmic property.
Collapse
Affiliation(s)
- Lv Song
- Cardio-Electrophysiological Research Laboratory, Medical College of Wuhan University of Science and Technology, Wuhan, China.,College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College of Wuhan University of Science and Technology, Wuhan, China
| | - Ze-Fu Zhang
- Cardio-Electrophysiological Research Laboratory, Medical College of Wuhan University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College of Wuhan University of Science and Technology, Wuhan, China
| | - Liang-Kun Hu
- Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Pei-Hua Zhang
- Cardio-Electrophysiological Research Laboratory, Medical College of Wuhan University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College of Wuhan University of Science and Technology, Wuhan, China
| | - Zhen-Zhen Cao
- Cardio-Electrophysiological Research Laboratory, Medical College of Wuhan University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College of Wuhan University of Science and Technology, Wuhan, China
| | - Zhi-Pei Liu
- Cardio-Electrophysiological Research Laboratory, Medical College of Wuhan University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College of Wuhan University of Science and Technology, Wuhan, China
| | - Pei-Pei Zhang
- Cardio-Electrophysiological Research Laboratory, Medical College of Wuhan University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College of Wuhan University of Science and Technology, Wuhan, China
| | - Ji-Hua Ma
- Cardio-Electrophysiological Research Laboratory, Medical College of Wuhan University of Science and Technology, Wuhan, China.,College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College of Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
47
|
Imran H, Nester W, Elgendy IY, Saad M. Role of sodium glucose co-transporter 2 inhibitors in patients with heart failure: an elusive mechanism. Ann Med 2020; 52:178-190. [PMID: 32393068 PMCID: PMC7877993 DOI: 10.1080/07853890.2020.1767298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) is a major cause of morbidity and mortality worldwide, and the burden of HF continues to rise. There has been an interest in sodium-glucose co-transporter-2 (SGLT2) inhibitors for their role in reducing HF hospitalizations in pivotal trials. Since these agents were approved by the Food and Drug Administration for the management of diabetes mellitus, multiple small trials and analyses have tried to explain the underlying beneficial mechanisms in HF . In this review, we discuss different mechanisms by which SGLT2 inhibitors play hemodynamic, metabolic, and cellular roles in different HF phenotypes. We also address issues pertaining to the safety of these relatively newer agents.KEY MESSAGESSGLT2 inhibitors are associated with a reduction in HF hospitalizations in both diabetics and non-diabetics.The beneficial role of SGLT2 inhibitors in reducing HF hospitalization is observed among participants with established cardiovascular disease/HF and at-risk population.SGLT2 inhibitors pose an important role in renal protection, another mechanism by which these medications can be helpful in HF patients.
Collapse
Affiliation(s)
- Hafiz Imran
- Cardiovascular Institute, Warren Alpert Medical School at Brown University, Providence, RI, USA
| | - William Nester
- Cardiovascular Institute, Warren Alpert Medical School at Brown University, Providence, RI, USA
| | - Islam Y Elgendy
- Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Marwan Saad
- Cardiovascular Institute, Warren Alpert Medical School at Brown University, Providence, RI, USA
| |
Collapse
|
48
|
Zhao Y, James NA, Beshay AR, Chang EE, Lin A, Bashar F, Wassily A, Nguyen B, Nguyen TP. Adult zebrafish ventricular electrical gradients as tissue mechanisms of ECG patterns under baseline vs. oxidative stress. Cardiovasc Res 2020; 117:1891-1907. [PMID: 32735330 DOI: 10.1093/cvr/cvaa238] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 06/29/2020] [Accepted: 07/24/2020] [Indexed: 12/27/2022] Open
Abstract
AIMS In mammalian ventricles, electrical gradients establish electrical heterogeneities as essential tissue mechanisms to optimize mechanical efficiency and safeguard electrical stability. Electrical gradients shape mammalian electrocardiographic patterns; disturbance of electrical gradients is proarrhythmic. The zebrafish heart is a popular surrogate model for human cardiac electrophysiology thanks to its remarkable recapitulation of human electrocardiogram and ventricular action potential features. Yet, zebrafish ventricular electrical gradients are largely unexplored. The goal of this study is to define the zebrafish ventricular electrical gradients that shape the QRS complex and T wave patterns at baseline and under oxidative stress. METHODS AND RESULTS We performed in vivo electrocardiography and ex vivo voltage-sensitive fluorescent epicardial and transmural optical mapping of adult zebrafish hearts at baseline and during acute H2O2 exposure. At baseline, apicobasal activation and basoapical repolarization gradients accounted for the polarity concordance between the QRS complex and T wave. During H2O2 exposure, differential regional impairment of activation and repolarization at the apex and base disrupted prior to baseline electrical gradients, resulting in either reversal or loss of polarity concordance between the QRS complex and T wave. KN-93, a specific calcium/calmodulin-dependent protein kinase II inhibitor (CaMKII), protected zebrafish hearts from H2O2 disruption of electrical gradients. The protection was complete if administered prior to oxidative stress exposure. CONCLUSIONS Despite remarkable apparent similarities, zebrafish and human ventricular electrocardiographic patterns are mirror images supported by opposite electrical gradients. Like mammalian ventricles, zebrafish ventricles are also susceptible to H2O2 proarrhythmic perturbation via CaMKII activation. Our findings suggest that the adult zebrafish heart may constitute a clinically relevant model to investigate ventricular arrhythmias induced by oxidative stress. However, the fundamental ventricular activation and repolarization differences between the two species that we demonstrated in this study highlight the potential limitations when extrapolating results from zebrafish experiments to human cardiac electrophysiology, arrhythmias, and drug toxicities.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Thao P Nguyen
- The Cardiovascular Research Laboratory, Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
49
|
Mustroph J, Drzymalski M, Baier M, Pabel S, Biedermann A, Memmel B, Durczok M, Neef S, Sag CM, Floerchinger B, Rupprecht L, Schmid C, Zausig Y, Bégis G, Briand V, Ozoux ML, Tamarelle D, Ballet V, Janiak P, Beauverger P, Maier LS, Wagner S. The oral Ca/calmodulin-dependent kinase II inhibitor RA608 improves contractile function and prevents arrhythmias in heart failure. ESC Heart Fail 2020; 7:2871-2883. [PMID: 32691522 PMCID: PMC7524064 DOI: 10.1002/ehf2.12895] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 05/29/2020] [Accepted: 06/24/2020] [Indexed: 01/15/2023] Open
Abstract
Aims Excessive activation of Ca/calmodulin‐dependent kinase II (CaMKII) is of critical importance in heart failure (HF) and atrial fibrillation. Unfortunately, lack of selectivity, specificity, and bioavailability have slowed down development of inhibitors for clinical use. We investigated a novel CaMKIIδ/CaMKIIɣ‐selective, ATP‐competitive, orally available CaMKII inhibitor (RA608) on right atrial biopsies of 119 patients undergoing heart surgery. Furthermore, we evaluated its oral efficacy to prevent deterioration of HF in mice after transverse aortic constriction (TAC). Methods and results In human atrial cardiomyocytes and trabeculae, respectively, RA608 significantly reduced sarcoplasmic reticulum Ca leak, reduced diastolic tension, and increased sarcoplasmic reticulum Ca content. Patch‐clamp recordings confirmed the safety of RA608 in human cardiomyocytes. C57BL6/J mice were subjected to TAC, and left ventricular function was monitored by echocardiography. Two weeks after TAC, RA608 was administered by oral gavage for 7 days. Oral RA608 treatment prevented deterioration of ejection fraction. At 3 weeks after TAC, ejection fraction was 46.1 ± 3.7% (RA608) vs. 34.9 ± 2.6% (vehicle), n = 9 vs. n = 12, P < 0.05, ANOVA, which correlated with significantly less CaMKII autophosphorylation at threonine 287. Moreover, a single oral dose significantly reduced inducibility of atrial and ventricular arrhythmias in CaMKIIδ transgenic mice 4 h after administration. Atrial fibrillation was induced in 6/6 mice for vehicle vs. 1/7 for RA608, P < 0.05, 'n − 1' χ2 test. Ventricular tachycardia was induced in 6/7 for vehicle vs. 2/7 for RA608, P < 0.05, 'n − 1' χ2 test. Conclusions RA608 is the first orally administrable CaMKII inhibitor with potent efficacy in human myocytes. Moreover, oral administration potently inhibits arrhythmogenesis and attenuates HF development in mice in vivo.
Collapse
Affiliation(s)
- Julian Mustroph
- Department of Internal Medicine II, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, Germany
| | - Marzena Drzymalski
- Department of Internal Medicine II, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, Germany
| | - Maria Baier
- Department of Internal Medicine II, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, Germany
| | - Steffen Pabel
- Department of Internal Medicine II, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, Germany
| | - Alexander Biedermann
- Department of Internal Medicine II, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, Germany
| | - Bernadette Memmel
- Department of Internal Medicine II, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, Germany
| | - Melanie Durczok
- Department of Internal Medicine II, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, Germany
| | - Stefan Neef
- Department of Internal Medicine II, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, Germany
| | - Can Martin Sag
- Department of Internal Medicine II, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, Germany
| | - Bernhard Floerchinger
- Department of Cardiothoracic Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Leopold Rupprecht
- Department of Cardiothoracic Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Christof Schmid
- Department of Cardiothoracic Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - York Zausig
- Department of Anesthesiology, University Medical Center Regensburg, Regensburg, Germany
| | | | | | | | | | | | - Philip Janiak
- Sanofi Research & Development (R&D), Chilly-Mazarin, France
| | | | - Lars S Maier
- Department of Internal Medicine II, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, Germany
| | - Stefan Wagner
- Department of Internal Medicine II, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, Germany
| |
Collapse
|
50
|
Role of Oxidation-Dependent CaMKII Activation in the Genesis of Abnormal Action Potentials in Atrial Cardiomyocytes: A Simulation Study. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1597012. [PMID: 32685443 PMCID: PMC7327560 DOI: 10.1155/2020/1597012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 05/20/2020] [Accepted: 06/02/2020] [Indexed: 01/04/2023]
Abstract
Atrial fibrillation is a common cardiac arrhythmia with an increasing incidence rate. Particularly for the aging population, understanding the underlying mechanisms of atrial arrhythmia is important in designing clinical treatment. Recently, experiments have shown that atrial arrhythmia is associated with oxidative stress. In this study, an atrial cell model including oxidative-dependent Ca2+/calmodulin- (CaM-) dependent protein kinase II (CaMKII) activation was developed to explore the intrinsic mechanisms of atrial arrhythmia induced by oxidative stress. The simulation results showed that oxidative stress caused early afterdepolarizations (EADs) of action potentials by altering the dynamics of transmembrane currents and intracellular calcium cycling. Oxidative stress gradually elevated the concentration of calcium ions in the cytoplasm by enhancing the L-type Ca2+ current and sarcoplasmic reticulum (SR) calcium release. Owing to increased intracellular calcium concentration, the inward Na+/Ca2+ exchange current was elevated which slowed down the repolarization of the action potential. Thus, the action potential was prolonged and the L-type Ca2+ current was reactivated, resulting in the genesis of EAD. Furthermore, based on the atrial single-cell model, a two-dimensional (2D) ideal tissue model was developed to explore the effect of oxidative stress on the electrical excitation wave conduction in 2D tissue. Simulation results demonstrated that, under oxidative stress conditions, EAD hindered the conduction of electrical excitation and caused an unstable spiral wave, which could disrupt normal cardiac rhythm and cause atrial arrhythmia. This study showed the effects of excess reactive oxygen species on calcium cycling and action potential in atrial myocytes and provided insights regarding atrial arrhythmia induced by oxidative stress.
Collapse
|