1
|
Habecker BA, Bers DM, Birren SJ, Chang R, Herring N, Kay MW, Li D, Mendelowitz D, Mongillo M, Montgomery JM, Ripplinger CM, Tampakakis E, Winbo A, Zaglia T, Zeltner N, Paterson DJ. Molecular and cellular neurocardiology in heart disease. J Physiol 2025; 603:1689-1728. [PMID: 38778747 PMCID: PMC11582088 DOI: 10.1113/jp284739] [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: 02/23/2024] [Accepted: 04/16/2024] [Indexed: 05/25/2024] Open
Abstract
This paper updates and builds on a previous White Paper in this journal that some of us contributed to concerning the molecular and cellular basis of cardiac neurobiology of heart disease. Here we focus on recent findings that underpin cardiac autonomic development, novel intracellular pathways and neuroplasticity. Throughout we highlight unanswered questions and areas of controversy. Whilst some neurochemical pathways are already demonstrating prognostic viability in patients with heart failure, we also discuss the opportunity to better understand sympathetic impairment by using patient specific stem cells that provides pathophysiological contextualization to study 'disease in a dish'. Novel imaging techniques and spatial transcriptomics are also facilitating a road map for target discovery of molecular pathways that may form a therapeutic opportunity to treat cardiac dysautonomia.
Collapse
Affiliation(s)
- Beth A Habecker
- Department of Chemical Physiology & Biochemistry, Department of Medicine Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis School of Medicine, Davis, CA, USA
| | - Susan J Birren
- Department of Biology, Volen Center for Complex Systems, Brandeis University, Waltham, MA, USA
| | - Rui Chang
- Department of Neuroscience, Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Neil Herring
- Burdon Sanderson Cardiac Science Centre and BHF Centre of Research Excellence, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Matthew W Kay
- Department of Biomedical Engineering, George Washington University, Washington, DC, USA
| | - Dan Li
- Burdon Sanderson Cardiac Science Centre and BHF Centre of Research Excellence, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - David Mendelowitz
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
| | - Marco Mongillo
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Johanna M Montgomery
- Department of Physiology and Manaaki Manawa Centre for Heart Research, University of Auckland, Auckland, New Zealand
| | - Crystal M Ripplinger
- Department of Pharmacology, University of California, Davis School of Medicine, Davis, CA, USA
| | | | - Annika Winbo
- Department of Physiology and Manaaki Manawa Centre for Heart Research, University of Auckland, Auckland, New Zealand
| | - Tania Zaglia
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Nadja Zeltner
- Departments of Biochemistry and Molecular Biology, Cell Biology, and Center for Molecular Medicine, University of Georgia, Athens, GA, USA
| | - David J Paterson
- Burdon Sanderson Cardiac Science Centre and BHF Centre of Research Excellence, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
2
|
Zhou J, Chen Z, Zhang M, Ye Y, Shen Y, Wu X. Exploration of the potential association between newer antiseizure medications and arrhythmias: Integrating pharmacovigilance and bioinformatics evidence. Seizure 2024; 123:26-33. [PMID: 39454529 DOI: 10.1016/j.seizure.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
INTRODUCTION Arrhythmias resulting from newer antiseizure medications (ASMs) may significantly impact the safety and quality of life of patients with epilepsy. This study investigated the potential association between new first-line or second-line ASMs and arrhythmias. METHODS Pharmacovigilance analysis was conducted using data from the Food and Drug Administration Adverse Event Reporting System (FAERS) from 2004 to 2023. A disproportionality analysis was performed to compare newer ASMs with other drugs, using carbamazepine and valproate as positive controls. Newer ASMs were categorized into sodium channel (SCN) main mechanism, SCN possible mechanism, and non-SCN group. The bioinformatics analysis involved retrieving therapeutic gene targets for ASMs from the DrugBank and OMIM databases, as well as identifying arrhythmia disease targets from the GeneCards database. Additionally, enrichment analysis of gene ontology functions and KEGG pathways was conducted. RESULTS A total of 3,457 cases of arrhythmias associated with newer ASMs were identified in the FAERS database. Disproportionality analysis indicates that brivaracetam (IC025 = 0.08), zonisamide (IC025 = 0.13), eslicarbazepine (IC025 = 0.39), and lacosamide (IC025 = 0.84) exhibited a positive signal for arrhythmias, with signals predominantly observed in the SCN main mechanism group. Furthermore, bioinformatics analysis revealed the involvement of adrenergic signaling in cardiac myocytes, as well as the participation of sodium channel genes in ASM-induced arrhythmias. CONCLUSION Our findings suggest a potential association between SCN-ASMs and arrhythmias, highlighting the importance of monitoring and evaluating the safety profiles of newer ASMs in clinical practice. Further research is necessary to elucidate the underlying mechanisms and inform patient care strategies.
Collapse
Affiliation(s)
- Jianxing Zhou
- Department of Pharmacy, Fujian Medical University Union Hospital, 29 Xinquan Rd. Gulou District, Fuzhou, Fujian 350001, China; School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China.
| | - Zhenhui Chen
- Department of Pharmacy, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, China
| | - Mengjun Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yanrong Ye
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun Shen
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xuemei Wu
- Department of Pharmacy, Fujian Medical University Union Hospital, 29 Xinquan Rd. Gulou District, Fuzhou, Fujian 350001, China; School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
3
|
Carvajal C, Yan J, Nani A, DeSantiago J, Wan X, Deschenes I, Ai X, Fill M. Isolated Cardiac Ryanodine Receptor Function Varies Between Mammals. J Membr Biol 2024; 257:25-36. [PMID: 38285125 PMCID: PMC11299243 DOI: 10.1007/s00232-023-00301-0] [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: 08/03/2023] [Accepted: 11/24/2023] [Indexed: 01/30/2024]
Abstract
Concerted robust opening of cardiac ryanodine receptors' (RyR2) Ca2+ release 1oplasmic reticulum (SR) is fundamental for normal systolic cardiac function. During diastole, infrequent spontaneous RyR2 openings mediate the SR Ca2+ leak that normally constrains SR Ca2+ load. Abnormal large diastolic RyR2-mediated Ca2+ leak events can cause delayed after depolarizations (DADs) and arrhythmias. The RyR2-associated mechanisms underlying these processes are being extensively studied at multiple levels utilizing various model animals. Since there are well-described species-specific differences in cardiac intracellular Ca2+ handing in situ, we tested whether or not single RyR2 function in vitro retains this species specificity. We isolated RyR2-rich heavy SR microsomes from mouse, rat, rabbit, and human ventricular muscle and quantified RyR2 function using identical solutions and methods. The single RyR2 cytosolic Ca2+ sensitivity was similar across these species. However, there were significant species differences in single RyR2 mean open times in both systole and diastole-like solutions. In diastole-like solutions, single rat/mouse RyR2 open probability and frequency of long openings (> 6 ms) were similar, but these values were significantly greater than those of either single rabbit or human RyR2s. We propose these in vitro single RyR2 functional differences across species stem from the species-specific RyR2 regulatory environment present in the source tissue. Our results show the single rabbit RyR2 functional attributes, particularly in diastole-like conditions, replicate those of single human RyR2 best among the species tested.
Collapse
Affiliation(s)
- Catherine Carvajal
- Department of Physiology & Biophysics, Section of Cellular Signaling, Rush University Medical Center, 1750 W. Harrison Avenue, Chicago, IL, 60612, USA
| | - Jiajie Yan
- Department of Physiology & Biophysics, Section of Cellular Signaling, Rush University Medical Center, 1750 W. Harrison Avenue, Chicago, IL, 60612, USA
- Department of Physiology & Cell Biology, College of Medicine, The Ohio State University, 333 W. 10Th Avenue, Columbus, OH, 43210, USA
| | - Alma Nani
- Department of Physiology & Biophysics, Section of Cellular Signaling, Rush University Medical Center, 1750 W. Harrison Avenue, Chicago, IL, 60612, USA
| | - Jaime DeSantiago
- Department of Physiology & Biophysics, Section of Cellular Signaling, Rush University Medical Center, 1750 W. Harrison Avenue, Chicago, IL, 60612, USA
| | - Xiaoping Wan
- Department of Physiology & Cell Biology, College of Medicine, The Ohio State University, 333 W. 10Th Avenue, Columbus, OH, 43210, USA
| | - Isabelle Deschenes
- Department of Physiology & Cell Biology, College of Medicine, The Ohio State University, 333 W. 10Th Avenue, Columbus, OH, 43210, USA
| | - Xun Ai
- Department of Physiology & Biophysics, Section of Cellular Signaling, Rush University Medical Center, 1750 W. Harrison Avenue, Chicago, IL, 60612, USA.
- Department of Physiology & Cell Biology, College of Medicine, The Ohio State University, 333 W. 10Th Avenue, Columbus, OH, 43210, USA.
| | - Michael Fill
- Department of Physiology & Biophysics, Section of Cellular Signaling, Rush University Medical Center, 1750 W. Harrison Avenue, Chicago, IL, 60612, USA.
- Department of Molecular Biophysics & Physiology, Rush University Medical Center, 1750 West Harrison Street, Columbus, OH, 43210, USA.
| |
Collapse
|
4
|
Wong YW, Haqqani H, Molenaar P. Roles of β-adrenoceptor Subtypes and Therapeutics in Human Cardiovascular Disease: Heart Failure, Tachyarrhythmias and Other Cardiovascular Disorders. Handb Exp Pharmacol 2024; 285:247-295. [PMID: 38844580 DOI: 10.1007/164_2024_720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
β-Adrenoceptors (β-ARs) provide an important therapeutic target for the treatment of cardiovascular disease. Three β-ARs, β1-AR, β2-AR, β3-AR are localized to the human heart. Activation of β1-AR and β2-ARs increases heart rate, force of contraction (inotropy) and consequently cardiac output to meet physiological demand. However, in disease, chronic over-activation of β1-AR is responsible for the progression of disease (e.g. heart failure) mediated by pathological hypertrophy, adverse remodelling and premature cell death. Furthermore, activation of β1-AR is critical in the pathogenesis of cardiac arrhythmias while activation of β2-AR directly influences blood pressure haemostasis. There is an increasing awareness of the contribution of β2-AR in cardiovascular disease, particularly arrhythmia generation. All β-blockers used therapeutically to treat cardiovascular disease block β1-AR with variable blockade of β2-AR depending on relative affinity for β1-AR vs β2-AR. Since the introduction of β-blockers into clinical practice in 1965, β-blockers with different properties have been trialled, used and evaluated, leading to better understanding of their therapeutic effects and tolerability in various cardiovascular conditions. β-Blockers with the property of intrinsic sympathomimetic activity (ISA), i.e. β-blockers that also activate the receptor, were used in the past for post-treatment of myocardial infarction and had limited use in heart failure. The β-blocker carvedilol continues to intrigue due to numerous properties that differentiate it from other β-blockers and is used successfully in the treatment of heart failure. The discovery of β3-AR in human heart created interest in the role of β3-AR in heart failure but has not resulted in therapeutics at this stage.
Collapse
Affiliation(s)
- Yee Weng Wong
- Cardiovascular Molecular & Therapeutics Translational Research Group, Northside Clinical School of Medicine, University of Queensland, The Prince Charles Hospital, Chermside, QLD, Australia
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Haris Haqqani
- Cardiovascular Molecular & Therapeutics Translational Research Group, Northside Clinical School of Medicine, University of Queensland, The Prince Charles Hospital, Chermside, QLD, Australia
- Department of Cardiology, The Prince Charles Hospital, Chermside, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Peter Molenaar
- Cardiovascular Molecular & Therapeutics Translational Research Group, Northside Clinical School of Medicine, University of Queensland, The Prince Charles Hospital, Chermside, QLD, Australia.
- Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia.
| |
Collapse
|
5
|
Sharma AK, Singh S, Bhat M, Gill K, Zaid M, Kumar S, Shakya A, Tantray J, Jose D, Gupta R, Yangzom T, Sharma RK, Sahu SK, Rathore G, Chandolia P, Singh M, Mishra A, Raj S, Gupta A, Agarwal M, Kifayat S, Gupta A, Gupta P, Vashist A, Vaibhav P, Kathuria N, Yadav V, Singh RP, Garg A. New drug discovery of cardiac anti-arrhythmic drugs: insights in animal models. Sci Rep 2023; 13:16420. [PMID: 37775650 PMCID: PMC10541452 DOI: 10.1038/s41598-023-41942-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 09/04/2023] [Indexed: 10/01/2023] Open
Abstract
Cardiac rhythm regulated by micro-macroscopic structures of heart. Pacemaker abnormalities or disruptions in electrical conduction, lead to arrhythmic disorders may be benign, typical, threatening, ultimately fatal, occurs in clinical practice, patients on digitalis, anaesthesia or acute myocardial infarction. Both traditional and genetic animal models are: In-vitro: Isolated ventricular Myocytes, Guinea pig papillary muscles, Patch-Clamp Experiments, Porcine Atrial Myocytes, Guinea pig ventricular myocytes, Guinea pig papillary muscle: action potential and refractory period, Langendorff technique, Arrhythmia by acetylcholine or potassium. Acquired arrhythmia disorders: Transverse Aortic Constriction, Myocardial Ischemia, Complete Heart Block and AV Node Ablation, Chronic Tachypacing, Inflammation, Metabolic and Drug-Induced Arrhythmia. In-Vivo: Chemically induced arrhythmia: Aconitine antagonism, Digoxin-induced arrhythmia, Strophanthin/ouabain-induced arrhythmia, Adrenaline-induced arrhythmia, and Calcium-induced arrhythmia. Electrically induced arrhythmia: Ventricular fibrillation electrical threshold, Arrhythmia through programmed electrical stimulation, sudden coronary death in dogs, Exercise ventricular fibrillation. Genetic Arrhythmia: Channelopathies, Calcium Release Deficiency Syndrome, Long QT Syndrome, Short QT Syndrome, Brugada Syndrome. Genetic with Structural Heart Disease: Arrhythmogenic Right Ventricular Cardiomyopathy/Dysplasia, Dilated Cardiomyopathy, Hypertrophic Cardiomyopathy, Atrial Fibrillation, Sick Sinus Syndrome, Atrioventricular Block, Preexcitation Syndrome. Arrhythmia in Pluripotent Stem Cell Cardiomyocytes. Conclusion: Both traditional and genetic, experimental models of cardiac arrhythmias' characteristics and significance help in development of new antiarrhythmic drugs.
Collapse
Affiliation(s)
- Ashish Kumar Sharma
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India.
| | - Shivam Singh
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Mehvish Bhat
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Kartik Gill
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Mohammad Zaid
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Anjali Shakya
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Junaid Tantray
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Divyamol Jose
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Rashmi Gupta
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Tsering Yangzom
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Rajesh Kumar Sharma
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | | | - Gulshan Rathore
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Priyanka Chandolia
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Mithilesh Singh
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Anurag Mishra
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Shobhit Raj
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Archita Gupta
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Mohit Agarwal
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Sumaiya Kifayat
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Anamika Gupta
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Prashant Gupta
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Ankit Vashist
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Parth Vaibhav
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Nancy Kathuria
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Vipin Yadav
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Ravindra Pal Singh
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Arun Garg
- MVN University, Palwal, Haryana, India
| |
Collapse
|
6
|
van Weperen VYH, Ripplinger CM, Vaseghi M. Autonomic control of ventricular function in health and disease: current state of the art. Clin Auton Res 2023; 33:491-517. [PMID: 37166736 PMCID: PMC10173946 DOI: 10.1007/s10286-023-00948-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/20/2023] [Indexed: 05/12/2023]
Abstract
PURPOSE Cardiac autonomic dysfunction is one of the main pillars of cardiovascular pathophysiology. The purpose of this review is to provide an overview of the current state of the art on the pathological remodeling that occurs within the autonomic nervous system with cardiac injury and available neuromodulatory therapies for autonomic dysfunction in heart failure. METHODS Data from peer-reviewed publications on autonomic function in health and after cardiac injury are reviewed. The role of and evidence behind various neuromodulatory therapies both in preclinical investigation and in-use in clinical practice are summarized. RESULTS A harmonic interplay between the heart and the autonomic nervous system exists at multiple levels of the neuraxis. This interplay becomes disrupted in the setting of cardiovascular disease, resulting in pathological changes at multiple levels, from subcellular cardiac signaling of neurotransmitters to extra-cardiac, extra-thoracic remodeling. The subsequent detrimental cycle of sympathovagal imbalance, characterized by sympathoexcitation and parasympathetic withdrawal, predisposes to ventricular arrhythmias, progression of heart failure, and cardiac mortality. Knowledge on the etiology and pathophysiology of this condition has increased exponentially over the past few decades, resulting in a number of different neuromodulatory approaches. However, significant knowledge gaps in both sympathetic and parasympathetic interactions and causal factors that mediate progressive sympathoexcitation and parasympathetic dysfunction remain. CONCLUSIONS Although our understanding of autonomic imbalance in cardiovascular diseases has significantly increased, specific, pivotal mediators of this imbalance and the recognition and implementation of available autonomic parameters and neuromodulatory therapies are still lagging.
Collapse
Affiliation(s)
- Valerie Y H van Weperen
- Division of Cardiology, Department of Medicine, UCLA Cardiac Arrythmia Center, University of California, 100 Medical Plaza, Suite 660, Los Angeles, CA, 90095, USA
| | | | - Marmar Vaseghi
- Division of Cardiology, Department of Medicine, UCLA Cardiac Arrythmia Center, University of California, 100 Medical Plaza, Suite 660, Los Angeles, CA, 90095, USA.
| |
Collapse
|
7
|
Fang C, Zuo K, Jiao K, Zhu X, Fu Y, Zhong J, Xu L, Yang X. PAGln, an Atrial Fibrillation-Linked Gut Microbial Metabolite, Acts as a Promoter of Atrial Myocyte Injury. Biomolecules 2022; 12:biom12081120. [PMID: 36009014 PMCID: PMC9405855 DOI: 10.3390/biom12081120] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/05/2022] [Accepted: 08/11/2022] [Indexed: 11/25/2022] Open
Abstract
Phenylacetylglutamine (PAGln), a gut microbiota (GM)-derived metabolite, is associated with cardiovascular disease. Studies have shown that disordered GM participated in the progression of atrial fibrillation (AF), but the relationship between PAGln and AF is unclear. This study investigated the characteristics of PAGln in AF patients and its impact on atrial myocytes. Based on our previous metagenomic data, the relative abundance of porA, a critical bacterial enzyme for PAGln synthesis, exhibited an increased tendency in AF. In an independent cohort consisting of 42 controls without AF and 92 AF patients, plasma PAGln levels were higher in AF patients than in controls (p < 0.001) by immunoassay. Notably, PAGln exerted a predictive potential of AF with an AUC of 0.774 (p < 0.001), and a predictive model constructed based on the PAGln and Taiwan AF score further improved the predictive potential. Furthermore, a positive correlation was determined between PAGln and LA diameter. Subsequently, the effect of PAGln intervention was examined on HL-1 cells in vitro, revealing that PAGln increased apoptosis, reactive oxygen species (ROS) production, CaMKII and RyR2 activation and decreased cell viability. In conclusion, increased PAGln was associated with AF, and PAGln might contribute to the AF pathogenesis by promoting oxidative stress and apoptosis in atrial myocytes.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Li Xu
- Correspondence: (L.X.); (X.Y.); Tel.: +86-10-85231937 (X.Y.)
| | - Xinchun Yang
- Correspondence: (L.X.); (X.Y.); Tel.: +86-10-85231937 (X.Y.)
| |
Collapse
|
8
|
Bartolucci C, Forouzandehmehr M, Severi S, Paci M. A Novel In Silico Electromechanical Model of Human Ventricular Cardiomyocyte. Front Physiol 2022; 13:906146. [PMID: 35721558 PMCID: PMC9198403 DOI: 10.3389/fphys.2022.906146] [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: 03/28/2022] [Accepted: 05/10/2022] [Indexed: 11/25/2022] Open
Abstract
Contractility has become one of the main readouts in computational and experimental studies on cardiomyocytes. Following this trend, we propose a novel mathematical model of human ventricular cardiomyocytes electromechanics, BPSLand, by coupling a recent human contractile element to the BPS2020 model of electrophysiology. BPSLand is the result of a hybrid optimization process and it reproduces all the electrophysiology experimental indices captured by its predecessor BPS2020, simultaneously enabling the simulation of realistic human active tension and its potential abnormalities. The transmural heterogeneity in both electrophysiology and contractility departments was simulated consistent with previous computational and in vitro studies. Furthermore, our model could capture delayed afterdepolarizations (DADs), early afterdepolarizations (EADs), and contraction abnormalities in terms of aftercontractions triggered by either drug action or special pacing modes. Finally, we further validated the mechanical results of the model against previous experimental and in silico studies, e.g., the contractility dependence on pacing rate. Adding a new level of applicability to the normative models of human cardiomyocytes, BPSLand represents a robust, fully-human in silico model with promising capabilities for translational cardiology.
Collapse
Affiliation(s)
- Chiara Bartolucci
- Computational Physiopathology Unit, Department of Electrical, Electronic and Information Engineering "Guglielmo Marconi", University of Bologna, Bologna, Italy
| | | | - Stefano Severi
- Computational Physiopathology Unit, Department of Electrical, Electronic and Information Engineering "Guglielmo Marconi", University of Bologna, Bologna, Italy
| | - Michelangelo Paci
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
9
|
Abstract
Cardiac arrhythmias are a significant cause of morbidity and mortality worldwide, accounting for 10% to 15% of all deaths. Although most arrhythmias are due to acquired heart disease, inherited channelopathies and cardiomyopathies disproportionately affect children and young adults. Arrhythmogenesis is complex, involving anatomic structure, ion channels and regulatory proteins, and the interplay between cells in the conduction system, cardiomyocytes, fibroblasts, and the immune system. Animal models of arrhythmia are powerful tools for studying not only molecular and cellular mechanism of arrhythmogenesis but also more complex mechanisms at the whole heart level, and for testing therapeutic interventions. This review summarizes basic and clinical arrhythmia mechanisms followed by an in-depth review of published animal models of genetic and acquired arrhythmia disorders.
Collapse
Affiliation(s)
- Daniel J Blackwell
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| | - Jeffrey Schmeckpeper
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| | - Bjorn C Knollmann
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
10
|
Amoni M, Dries E, Ingelaere S, Vermoortele D, Roderick HL, Claus P, Willems R, Sipido KR. Ventricular Arrhythmias in Ischemic Cardiomyopathy-New Avenues for Mechanism-Guided Treatment. Cells 2021; 10:2629. [PMID: 34685609 PMCID: PMC8534043 DOI: 10.3390/cells10102629] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/20/2021] [Accepted: 09/23/2021] [Indexed: 12/13/2022] Open
Abstract
Ischemic heart disease is the most common cause of lethal ventricular arrhythmias and sudden cardiac death (SCD). In patients who are at high risk after myocardial infarction, implantable cardioverter defibrillators are the most effective treatment to reduce incidence of SCD and ablation therapy can be effective for ventricular arrhythmias with identifiable culprit lesions. Yet, these approaches are not always successful and come with a considerable cost, while pharmacological management is often poor and ineffective, and occasionally proarrhythmic. Advances in mechanistic insights of arrhythmias and technological innovation have led to improved interventional approaches that are being evaluated clinically, yet pharmacological advancement has remained behind. We review the mechanistic basis for current management and provide a perspective for gaining new insights that centre on the complex tissue architecture of the arrhythmogenic infarct and border zone with surviving cardiac myocytes as the source of triggers and central players in re-entry circuits. Identification of the arrhythmia critical sites and characterisation of the molecular signature unique to these sites can open avenues for targeted therapy and reduce off-target effects that have hampered systemic pharmacotherapy. Such advances are in line with precision medicine and a patient-tailored therapy.
Collapse
Affiliation(s)
- Matthew Amoni
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (M.A.); (E.D.); (S.I.); (H.L.R.); (R.W.)
- Division of Cardiology, University Hospitals Leuven, 3000 Leuven, Belgium
- Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7935, South Africa
| | - Eef Dries
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (M.A.); (E.D.); (S.I.); (H.L.R.); (R.W.)
| | - Sebastian Ingelaere
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (M.A.); (E.D.); (S.I.); (H.L.R.); (R.W.)
- Division of Cardiology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Dylan Vermoortele
- Imaging and Cardiovascular Dynamics, Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (D.V.); (P.C.)
| | - H. Llewelyn Roderick
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (M.A.); (E.D.); (S.I.); (H.L.R.); (R.W.)
| | - Piet Claus
- Imaging and Cardiovascular Dynamics, Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (D.V.); (P.C.)
| | - Rik Willems
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (M.A.); (E.D.); (S.I.); (H.L.R.); (R.W.)
- Division of Cardiology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Karin R. Sipido
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (M.A.); (E.D.); (S.I.); (H.L.R.); (R.W.)
| |
Collapse
|
11
|
Du X. Sympatho-adrenergic mechanisms in heart failure: new insights into pathophysiology. MEDICAL REVIEW (BERLIN, GERMANY) 2021; 1:47-77. [PMID: 37724075 PMCID: PMC10388789 DOI: 10.1515/mr-2021-0007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/02/2021] [Indexed: 09/20/2023]
Abstract
The sympathetic nervous system is activated in the setting of heart failure (HF) to compensate for hemodynamic instability. However, acute sympathetic surge or sustained high neuronal firing rates activates β-adrenergic receptor (βAR) signaling contributing to myocardial remodeling, dysfunction and electrical instability. Thus, sympatho-βAR activation is regarded as a hallmark of HF and forms pathophysiological basis for β-blocking therapy. Building upon earlier research findings, studies conducted in the recent decades have significantly advanced our understanding on the sympatho-adrenergic mechanism in HF, which forms the focus of this article. This review notes recent research progress regarding the roles of cardiac β2AR or α1AR in the failing heart, significance of β1AR-autoantibodies, and βAR signaling through G-protein independent signaling pathways. Sympatho-βAR regulation of immune cells or fibroblasts is specifically discussed. On the neuronal aspects, knowledge is assembled on the remodeling of sympathetic nerves of the failing heart, regulation by presynaptic α2AR of NE release, and findings on device-based neuromodulation of the sympathetic nervous system. The review ends with highlighting areas where significant knowledge gaps exist but hold promise for new breakthroughs.
Collapse
Affiliation(s)
- Xiaojun Du
- Faculty of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, 76 West Yanta Road, Xi’an710061, Shaanxi, China
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC3004, Australia
| |
Collapse
|
12
|
Ghigo A, Harvey RD. Illuminating cAMP Dynamics at Ryanodine Receptors in Arrhythmias. Circ Res 2021; 129:95-97. [PMID: 34166076 DOI: 10.1161/circresaha.121.319449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Italy (A.G.)
| | - Robert D Harvey
- Department of Pharmacology, University of Nevada, Reno (R.D.H.)
| |
Collapse
|
13
|
Fisher ME, Bovo E, Aguayo-Ortiz R, Cho EE, Pribadi MP, Dalton MP, Rathod N, Lemieux MJ, Espinoza-Fonseca LM, Robia SL, Zima AV, Young HS. Dwarf open reading frame (DWORF) is a direct activator of the sarcoplasmic reticulum calcium pump SERCA. eLife 2021; 10:65545. [PMID: 34075877 PMCID: PMC8203291 DOI: 10.7554/elife.65545] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 06/01/2021] [Indexed: 01/05/2023] Open
Abstract
The sarco-plasmic reticulum calcium pump (SERCA) plays a critical role in the contraction-relaxation cycle of muscle. In cardiac muscle, SERCA is regulated by the inhibitor phospholamban. A new regulator, dwarf open reading frame (DWORF), has been reported to displace phospholamban from SERCA. Here, we show that DWORF is a direct activator of SERCA, increasing its turnover rate in the absence of phospholamban. Measurement of in-cell calcium dynamics supports this observation and demonstrates that DWORF increases SERCA-dependent calcium reuptake. These functional observations reveal opposing effects of DWORF activation and phospholamban inhibition of SERCA. To gain mechanistic insight into SERCA activation, fluorescence resonance energy transfer experiments revealed that DWORF has a higher affinity for SERCA in the presence of calcium. Molecular modeling and molecular dynamics simulations provide a model for DWORF activation of SERCA, where DWORF modulates the membrane bilayer and stabilizes the conformations of SERCA that predominate during elevated cytosolic calcium.
Collapse
Affiliation(s)
- M'Lynn E Fisher
- Department of Biochemistry, University of Alberta, Edmonton, Canada
| | - Elisa Bovo
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, United States
| | - Rodrigo Aguayo-Ortiz
- Center for Arrhythmia Research, Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, United States
| | - Ellen E Cho
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, United States
| | - Marsha P Pribadi
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, United States
| | - Michael P Dalton
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, United States
| | - Nishadh Rathod
- Department of Biochemistry, University of Alberta, Edmonton, Canada
| | - M Joanne Lemieux
- Department of Biochemistry, University of Alberta, Edmonton, Canada
| | - L Michel Espinoza-Fonseca
- Center for Arrhythmia Research, Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, United States
| | - Seth L Robia
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, United States
| | - Aleksey V Zima
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, United States
| | - Howard S Young
- Department of Biochemistry, University of Alberta, Edmonton, Canada
| |
Collapse
|
14
|
Berisha F, Götz KR, Wegener JW, Brandenburg S, Subramanian H, Molina CE, Rüffer A, Petersen J, Bernhardt A, Girdauskas E, Jungen C, Pape U, Kraft AE, Warnke S, Lindner D, Westermann D, Blankenberg S, Meyer C, Hasenfuß G, Lehnart SE, Nikolaev VO. cAMP Imaging at Ryanodine Receptors Reveals β 2-Adrenoceptor Driven Arrhythmias. Circ Res 2021; 129:81-94. [PMID: 33902292 DOI: 10.1161/circresaha.120.318234] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Filip Berisha
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (F.B., H.S., C.E.M., A.E.K., V.O.N.).,Department of Cardiology (F.B., C.J., U.P., S.W., D.L., D.W., S. Blankenberg, C.M.), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (F.B., H.S., C.E.M., A.E., S.W., D.L., D.W., S. Blankenberg, V.O.N.)
| | - Konrad R Götz
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, Georg August University Medical Center, Germany (K.R.G., J.W.W., S. Brandenburg, G.H., S.E.L.)
| | - Jörg W Wegener
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, Georg August University Medical Center, Germany (K.R.G., J.W.W., S. Brandenburg, G.H., S.E.L.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany (J.W.W., S. Brandenburg, G.H., S.E.L.)
| | - Sören Brandenburg
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, Georg August University Medical Center, Germany (K.R.G., J.W.W., S. Brandenburg, G.H., S.E.L.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany (J.W.W., S. Brandenburg, G.H., S.E.L.)
| | - Hariharan Subramanian
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (F.B., H.S., C.E.M., A.E.K., V.O.N.).,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (F.B., H.S., C.E.M., A.E., S.W., D.L., D.W., S. Blankenberg, V.O.N.)
| | - Cristina E Molina
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (F.B., H.S., C.E.M., A.E.K., V.O.N.).,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (F.B., H.S., C.E.M., A.E., S.W., D.L., D.W., S. Blankenberg, V.O.N.)
| | - André Rüffer
- Department of Cardiovascular Surgery, University Heart and Vascular Center Hamburg, Germany (A.R., J.P., A.B., E.G.)
| | - Johannes Petersen
- Department of Cardiovascular Surgery, University Heart and Vascular Center Hamburg, Germany (A.R., J.P., A.B., E.G.)
| | - Alexander Bernhardt
- Department of Cardiovascular Surgery, University Heart and Vascular Center Hamburg, Germany (A.R., J.P., A.B., E.G.)
| | - Evaldas Girdauskas
- Department of Cardiovascular Surgery, University Heart and Vascular Center Hamburg, Germany (A.R., J.P., A.B., E.G.)
| | - Christiane Jungen
- Department of Cardiology (F.B., C.J., U.P., S.W., D.L., D.W., S. Blankenberg, C.M.), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Cardiology-Electrophysiology, cNEP (Cardiac Neuro- and Electrophysiology Research Group) (C.J., U.P., C.M.), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrike Pape
- Department of Cardiology (F.B., C.J., U.P., S.W., D.L., D.W., S. Blankenberg, C.M.), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Cardiology-Electrophysiology, cNEP (Cardiac Neuro- and Electrophysiology Research Group) (C.J., U.P., C.M.), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Axel E Kraft
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (F.B., H.S., C.E.M., A.E.K., V.O.N.).,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (F.B., H.S., C.E.M., A.E., S.W., D.L., D.W., S. Blankenberg, V.O.N.)
| | - Svenja Warnke
- Department of Cardiology (F.B., C.J., U.P., S.W., D.L., D.W., S. Blankenberg, C.M.), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (F.B., H.S., C.E.M., A.E., S.W., D.L., D.W., S. Blankenberg, V.O.N.)
| | - Diana Lindner
- Department of Cardiology (F.B., C.J., U.P., S.W., D.L., D.W., S. Blankenberg, C.M.), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (F.B., H.S., C.E.M., A.E., S.W., D.L., D.W., S. Blankenberg, V.O.N.)
| | - Dirk Westermann
- Department of Cardiology (F.B., C.J., U.P., S.W., D.L., D.W., S. Blankenberg, C.M.), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (F.B., H.S., C.E.M., A.E., S.W., D.L., D.W., S. Blankenberg, V.O.N.)
| | - Stefan Blankenberg
- Department of Cardiology (F.B., C.J., U.P., S.W., D.L., D.W., S. Blankenberg, C.M.), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (F.B., H.S., C.E.M., A.E., S.W., D.L., D.W., S. Blankenberg, V.O.N.)
| | - Christian Meyer
- Department of Cardiology (F.B., C.J., U.P., S.W., D.L., D.W., S. Blankenberg, C.M.), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Cardiology-Electrophysiology, cNEP (Cardiac Neuro- and Electrophysiology Research Group) (C.J., U.P., C.M.), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gerd Hasenfuß
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, Georg August University Medical Center, Germany (K.R.G., J.W.W., S. Brandenburg, G.H., S.E.L.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany (J.W.W., S. Brandenburg, G.H., S.E.L.)
| | - Stephan E Lehnart
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany (J.W.W., S. Brandenburg, G.H., S.E.L.)
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (F.B., H.S., C.E.M., A.E.K., V.O.N.).,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (F.B., H.S., C.E.M., A.E., S.W., D.L., D.W., S. Blankenberg, V.O.N.)
| |
Collapse
|
15
|
Stress-driven cardiac calcium mishandling via a kinase-to-kinase crosstalk. Pflugers Arch 2021; 473:363-375. [PMID: 33590296 PMCID: PMC7940337 DOI: 10.1007/s00424-021-02533-2] [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: 11/11/2020] [Revised: 01/19/2021] [Accepted: 02/02/2021] [Indexed: 01/25/2023]
Abstract
Calcium homeostasis in the cardiomyocyte is critical to the regulation of normal cardiac function. Abnormal calcium dynamics such as altered uptake by the sarcoplasmic reticulum (SR) Ca2+-ATPase and increased diastolic SR calcium leak are involved in the development of maladaptive cardiac remodeling under pathological conditions. Ca2+/calmodulin-dependent protein kinase II-δ (CaMKIIδ) is a well-recognized key molecule in calcium dysregulation in cardiomyocytes. Elevated cellular stress is known as a common feature during pathological remodeling, and c-jun N-terminal kinase (JNK) is an important stress kinase that is activated in response to intrinsic and extrinsic stress stimuli. Our lab recently identified specific actions of JNK isoform 2 (JNK2) in CaMKIIδ expression, activation, and CaMKIIδ-dependent SR Ca2+ mishandling in the stressed heart. This review focuses on the current understanding of cardiac SR calcium handling under physiological and pathological conditions as well as the newly identified contribution of the stress kinase JNK2 in CaMKIIδ-dependent SR Ca2+ abnormal mishandling. The new findings identifying dual roles of JNK2 in CaMKIIδ expression and activation are also discussed in this review.
Collapse
|
16
|
CaMKII and PKA-dependent phosphorylation co-regulate nuclear localization of HDAC4 in adult cardiomyocytes. Basic Res Cardiol 2021; 116:11. [PMID: 33590335 PMCID: PMC7884572 DOI: 10.1007/s00395-021-00850-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023]
Abstract
Nuclear histone deacetylase 4 (HDAC4) represses MEF2-mediated transcription, implicated in the development of heart failure. CaMKII-dependent phosphorylation drives nucleus-to-cytoplasm HDAC4 shuttling, but protein kinase A (PKA) is also linked to HDAC4 translocation. However, the interplay of CaMKII and PKA in regulating adult cardiomyocyte HDAC4 translocation is unclear. Here we sought to determine the interplay of PKA- and CaMKII-dependent HDAC4 phosphorylation and translocation in adult mouse, rabbit and human ventricular myocytes. Confocal imaging and protein analyses revealed that inhibition of CaMKII-but not PKA, PKC or PKD-raised nucleo-to-cytoplasmic HDAC4 fluorescence ratio (FNuc/FCyto) by ~ 50%, indicating baseline CaMKII activity that limits HDAC4 nuclear localization. Further CaMKII activation (via increased extracellular [Ca2+], high pacing frequencies, angiotensin II or overexpression of CaM or CaMKIIδC) led to significant HDAC4 nuclear export. In contrast, PKA activation by isoproterenol or forskolin drove HDAC4 into the nucleus (raising FNuc/FCyto by > 60%). These PKA-mediated effects were abolished in cells pretreated with PKA inhibitors and in cells expressing mutant HDAC4 in S265/266A mutant. In physiological conditions where both kinases are active, PKA-dependent nuclear accumulation of HDAC4 was predominant in the very early response, while CaMKII-dependent HDAC4 export prevailed upon prolonged stimuli. This orchestrated co-regulation was shifted in failing cardiomyocytes, where CaMKII-dependent effects predominated over PKA-dependent response. Importantly, human cardiomyocytes showed similar CaMKII- and PKA-dependent HDAC4 shifts. Collectively, CaMKII limits nuclear localization of HDAC4, while PKA favors HDAC4 nuclear retention and S265/266 is essential for PKA-mediated regulation. These pathways thus compete in HDAC4 nuclear localization and transcriptional regulation in cardiac signaling.
Collapse
|
17
|
Mora MT, Gong JQX, Sobie EA, Trenor B. The role of β-adrenergic system remodeling in human heart failure: A mechanistic investigation. J Mol Cell Cardiol 2020; 153:14-25. [PMID: 33326834 DOI: 10.1016/j.yjmcc.2020.12.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 12/02/2020] [Accepted: 12/08/2020] [Indexed: 01/01/2023]
Abstract
β-adrenergic receptor antagonists (β-blockers) are extensively used to improve cardiac performance in heart failure (HF), but the electrical improvements with these clinical treatments are not fully understood. The aim of this study was to analyze the electrophysiological effects of β-adrenergic system remodeling in heart failure with reduced ejection fraction and the underlying mechanisms. We used a combined mathematical model that integrated β-adrenergic signaling with electrophysiology and calcium cycling in human ventricular myocytes. HF remodeling, both in the electrophysiological and signaling systems, was introduced to quantitatively analyze changes in electrophysiological properties due to the stimulation of β-adrenergic receptors in failing myocytes. We found that the inotropic effect of β-adrenergic stimulation was reduced in HF due to the altered Ca2+ dynamics resulting from the combination of structural, electrophysiological and signaling remodeling. Isolated cells showed proarrhythmic risk after sympathetic stimulation because early afterdepolarizations appeared, and the vulnerability was greater in failing myocytes. When analyzing coupled cells, β-adrenergic stimulation reduced transmural repolarization gradients between endocardium and epicardium in normal tissue, but was less effective at reducing these gradients after HF remodeling. The comparison of the selective activation of β-adrenergic isoforms revealed that the response to β2-adrenergic receptors stimulation was blunted in HF while β1-adrenergic receptors downstream effectors regulated most of the changes observed after sympathetic stimulation. In conclusion, this study was able to reproduce an altered β-adrenergic activity on failing myocytes and to explain the mechanisms involved. The derived predictions could help in the treatment of HF and guide in the design of future experiments.
Collapse
Affiliation(s)
- Maria T Mora
- Centro de Investigación e Innovación en Bioingeniería, Universitat Politècnica de València, Valencia, Spain
| | - Jingqi Q X Gong
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric A Sobie
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Beatriz Trenor
- Centro de Investigación e Innovación en Bioingeniería, Universitat Politècnica de València, Valencia, Spain.
| |
Collapse
|
18
|
Werley CA, Boccardo S, Rigamonti A, Hansson EM, Cohen AE. Multiplexed Optical Sensors in Arrayed Islands of Cells for multimodal recordings of cellular physiology. Nat Commun 2020; 11:3881. [PMID: 32753572 PMCID: PMC7403318 DOI: 10.1038/s41467-020-17607-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 07/07/2020] [Indexed: 11/29/2022] Open
Abstract
Cells typically respond to chemical or physical perturbations via complex signaling cascades which can simultaneously affect multiple physiological parameters, such as membrane voltage, calcium, pH, and redox potential. Protein-based fluorescent sensors can report many of these parameters, but spectral overlap prevents more than ~4 modalities from being recorded in parallel. Here we introduce the technique, MOSAIC, Multiplexed Optical Sensors in Arrayed Islands of Cells, where patterning of fluorescent sensor-encoding lentiviral vectors with a microarray printer enables parallel recording of multiple modalities. We demonstrate simultaneous recordings from 20 sensors in parallel in human embryonic kidney (HEK293) cells and in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), and we describe responses to metabolic and pharmacological perturbations. Together, these results show that MOSAIC can provide rich multi-modal data on complex physiological responses in multiple cell types.
Collapse
Affiliation(s)
- Christopher A Werley
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, 02138, USA
- Q-State Biosciences, Cambridge, MA, 02139, USA
| | - Stefano Boccardo
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, 02138, USA
- Nobel Biocare AG, Kloten, Switzerland
| | - Alessandra Rigamonti
- Integrated Cardio Metabolic Centre, Department of Medicine Huddinge, Karolinska Institute, Huddinge, Sweden
| | - Emil M Hansson
- Integrated Cardio Metabolic Centre, Department of Medicine Huddinge, Karolinska Institute, Huddinge, Sweden
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Adam E Cohen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, 02138, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA.
| |
Collapse
|
19
|
He M, Zhao WB, Nguyen MN, Kiriazis H, Li YQ, Hu H, Du XJ. Association between heart rate variability indices and features of spontaneous ventricular tachyarrhythmias in mice. Clin Exp Pharmacol Physiol 2020; 47:1193-1202. [PMID: 32027390 DOI: 10.1111/1440-1681.13275] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 02/02/2020] [Accepted: 02/03/2020] [Indexed: 12/20/2022]
Abstract
Direct evidence is limited for the association between heart rate variability (HRV) indices and ventricular tachyarrhythmias (VTAs). While galectin-3 (Gal-3) is regarded as a causal factor for cardiac remodelling and a biomarker for arrhythmias, its regulation on VTAs and HVR is unknown. Using aged transgenic (TG) mice with cardiac overexpression of β2 -adrenoceptors and spontaneous VTAs, we studied whether changes in HRV indices correlated with the severity of VTAs, and whether Gal-3 gene knockout (KO) in TG mice might limit VTA. Body-surface ECG was recorded (10-minute period) in 9- to 10-month-old mice of non-transgenic (nTG), TG and TG × Gal-3 knockout (TG/KO). Time-domain, frequency-domain and nonlinear-domain HRV indices were calculated using the R-R intervals extracted from ECG signals and compared with frequency of VTAs. TG and TG/KO mice developed frequent VTAs and showed significant changes in certain time-domain and nonlinear-domain HRV indices relative to nTG mice. The severity of VTAs in TG and TG/KO mice in combination, estimated by VTA counts and arrhythmia score, was significantly correlated with certain time-domain and nonlinear-domain HRV indices. In conclusion, significant changes in HRV indices were evident and correlated with the severity of spontaneous VTAs in TG mice. The frequency of VTA and HRV indices were largely comparable between TG and TG/KO mice. Deletion of Gal-3 in TG mice altered certain HRV indices implying influence by neuronally localized Gal-3 on autonomic nervous activity.
Collapse
Affiliation(s)
- Mi He
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- School of Biomedical Engineering and Imaging Medicine, Third Military Medical University (Army Medical University), Chongqing, China
| | - Wei-Bo Zhao
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - My-Nhan Nguyen
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Vic, Australia
| | - Helen Kiriazis
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Vic, Australia
| | - Yong-Qin Li
- School of Biomedical Engineering and Imaging Medicine, Third Military Medical University (Army Medical University), Chongqing, China
| | - Houyuan Hu
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiao-Jun Du
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Vic, Australia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University (Health Science Center), Xi'an, China
| |
Collapse
|
20
|
Zhang Y, Hou MC, Li JJ, Qi Y, Zhang Y, She G, Ren YJ, Wu W, Pang ZD, Xie W, Deng XL, Du XJ. Cardiac β-adrenergic receptor activation mediates distinct and cell type-dependent changes in the expression and distribution of connexin 43. J Cell Mol Med 2020; 24:8505-8517. [PMID: 32578931 PMCID: PMC7412418 DOI: 10.1111/jcmm.15469] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/27/2020] [Accepted: 05/12/2020] [Indexed: 12/14/2022] Open
Abstract
Activation of the sympatho-β-adrenergic receptors (β-ARs) system is a hallmark of heart failure, leading to fibrosis and arrhythmias. Connexin 43 (Cx43) is the most abundant gap junctional protein in the myocardium. Current knowledge is limited regarding Cx43 remodelling in diverse cell types in the diseased myocardium and the underlying mechanism. We studied cell type-dependent changes in Cx43 remodelling due to β-AR overactivation and molecular mechanisms involved. Mouse models of isoproterenol stimulation or transgenic cardiomyocyte overexpression of β2 -AR were used, which exhibited cardiac fibrosis and up-regulated total Cx43 abundance. In both models, whereas Cx43 expression in cardiomyocytes was reduced and more laterally distributed, fibroblasts exhibited elevated Cx43 expression and enhanced gap junction communication. Mechanistically, activation of β2 -AR in fibroblasts in vitro elevated Cx43 expression, which was abolished by the β2 -antagonist ICI-118551 or protein kinase A inhibitor H-89, but simulated by the adenylyl cyclase activator forskolin. Our in vitro and in vivo data showed that β-AR activation-induced production of IL-18 sequentially stimulated Cx43 expression in fibroblasts in a paracrine fashion. In summary, our findings demonstrate a pivotal role of β-AR in mediating distinct and cell type-dependent changes in the expression and distribution of Cx43, leading to pathological gap junction remodelling in the myocardium.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Meng-Chen Hou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Department of Pathology, Xi'an Guangren Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Jing-Jing Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Sciences and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Ying Qi
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Sciences and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Yu Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Gang She
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yu-Jie Ren
- Department of Pathology, Xi'an Guangren Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Wei Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Zheng-Da Pang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Wenjun Xie
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Sciences and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xiu-Ling Deng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, Shaanxi, China
| | - Xiao-Jun Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
| |
Collapse
|
21
|
Dries E, Amoni M, Vandenberk B, Johnson DM, Gilbert G, Nagaraju CK, Puertas RD, Abdesselem M, Santiago DJ, Roderick HL, Claus P, Willems R, Sipido KR. Altered adrenergic response in myocytes bordering a chronic myocardial infarction underlies in vivo triggered activity and repolarization instability. J Physiol 2020; 598:2875-2895. [PMID: 31900932 PMCID: PMC7496440 DOI: 10.1113/jp278839] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 01/01/2020] [Indexed: 01/24/2023] Open
Abstract
Key points Ventricular arrhythmias are a major complication after myocardial infarction (MI), associated with sympathetic activation. The structurally heterogeneous peri‐infarct zone is a known substrate, but the functional role of the myocytes is less well known. Recordings of monophasic action potentials in vivo reveal that the peri‐infarct zone is a source of delayed afterdepolarizations (DADs) and has a high beat‐to‐beat variability of repolarization (BVR) during adrenergic stimulation (isoproterenol, ISO). Myocytes isolated from the peri‐infarct region have more DADs and spontaneous action potentials, with spontaneous Ca2+ release, under ISO. These myocytes also have reduced repolarization reserve and increased BVR. Other properties of post‐MI remodelling are present in both peri‐infarct and remote myocytes. These data highlight the importance of altered myocyte adrenergic responses in the peri‐infarct region as source and substrate of post‐MI arrhythmias.
Abstract Ventricular arrhythmias are a major early complication after myocardial infarction (MI). The heterogeneous peri‐infarct zone forms a substrate for re‐entry while arrhythmia initiation is often associated with sympathetic activation. We studied the mechanisms triggering these post‐MI arrhythmias in vivo and their relation to regional myocyte remodelling. In pigs with chronic MI (6 weeks), in vivo monophasic action potentials were simultaneously recorded in the peri‐infarct and remote regions during adrenergic stimulation with isoproterenol (isoprenaline; ISO). Sham animals served as controls. During infusion of ISO in vivo, the incidence of delayed afterdepolarizations (DADs) and beat‐to‐beat variability of repolarization (BVR) was higher in the peri‐infarct than in the remote region. Myocytes isolated from the peri‐infarct region, in comparison to myocytes from the remote region, had more DADs, associated with spontaneous Ca2+ release, and a higher incidence of spontaneous action potentials (APs) when exposed to ISO (9.99 ± 4.2 vs. 0.16 ± 0.05 APs/min, p = 0.004); these were suppressed by CaMKII inhibition. Peri‐infarct myocytes also had reduced repolarization reserve and increased BVR (26 ± 10 ms vs. 9 ± 7 ms, P < 0.001), correlating with DAD activity. In contrast to these regional distinctions under ISO, alterations in Ca2+ handling at baseline and myocyte hypertrophy were present throughout the left ventricle (LV). Expression of some of the related genes was, however, different between the regions. In conclusion, altered myocyte adrenergic responses in the peri‐infarct but not the remote region provide a source of triggered activity in vivo and of repolarization instability amplifying the substrate for re‐entry. These findings stimulate further exploration of region‐specific therapies targeting myocytes and autonomic modulation. Ventricular arrhythmias are a major complication after myocardial infarction (MI), associated with sympathetic activation. The structurally heterogeneous peri‐infarct zone is a known substrate, but the functional role of the myocytes is less well known. Recordings of monophasic action potentials in vivo reveal that the peri‐infarct zone is a source of delayed afterdepolarizations (DADs) and has a high beat‐to‐beat variability of repolarization (BVR) during adrenergic stimulation (isoproterenol, ISO). Myocytes isolated from the peri‐infarct region have more DADs and spontaneous action potentials, with spontaneous Ca2+ release, under ISO. These myocytes also have reduced repolarization reserve and increased BVR. Other properties of post‐MI remodelling are present in both peri‐infarct and remote myocytes. These data highlight the importance of altered myocyte adrenergic responses in the peri‐infarct region as source and substrate of post‐MI arrhythmias.
Collapse
Affiliation(s)
- Eef Dries
- Experimental Cardiology, University of Leuven, Herestraat 49 box 911, Leuven, Belgium
| | - Matthew Amoni
- Experimental Cardiology, University of Leuven, Herestraat 49 box 911, Leuven, Belgium
| | - Bert Vandenberk
- Experimental Cardiology, University of Leuven, Herestraat 49 box 911, Leuven, Belgium
| | - Daniel M Johnson
- Experimental Cardiology, University of Leuven, Herestraat 49 box 911, Leuven, Belgium.,Institute of Cardiovascular Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Guillaume Gilbert
- Experimental Cardiology, University of Leuven, Herestraat 49 box 911, Leuven, Belgium
| | - Chandan K Nagaraju
- Experimental Cardiology, University of Leuven, Herestraat 49 box 911, Leuven, Belgium
| | - Rosa Doñate Puertas
- Experimental Cardiology, University of Leuven, Herestraat 49 box 911, Leuven, Belgium
| | - Mouna Abdesselem
- Experimental Cardiology, University of Leuven, Herestraat 49 box 911, Leuven, Belgium
| | - Demetrio J Santiago
- Experimental Cardiology, University of Leuven, Herestraat 49 box 911, Leuven, Belgium.,Laboratory of Molecular Cardiology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), C. Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - H Llewelyn Roderick
- Experimental Cardiology, University of Leuven, Herestraat 49 box 911, Leuven, Belgium
| | - Piet Claus
- Experimental Cardiology, University of Leuven, Herestraat 49 box 911, Leuven, Belgium
| | - Rik Willems
- Experimental Cardiology, University of Leuven, Herestraat 49 box 911, Leuven, Belgium
| | - Karin R Sipido
- Experimental Cardiology, University of Leuven, Herestraat 49 box 911, Leuven, Belgium
| |
Collapse
|
22
|
Clauss S, Bleyer C, Schüttler D, Tomsits P, Renner S, Klymiuk N, Wakili R, Massberg S, Wolf E, Kääb S. Animal models of arrhythmia: classic electrophysiology to genetically modified large animals. Nat Rev Cardiol 2020; 16:457-475. [PMID: 30894679 DOI: 10.1038/s41569-019-0179-0] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Arrhythmias are common and contribute substantially to cardiovascular morbidity and mortality. The underlying pathophysiology of arrhythmias is complex and remains incompletely understood, which explains why mostly only symptomatic therapy is available. The evaluation of the complex interplay between various cell types in the heart, including cardiomyocytes from the conduction system and the working myocardium, fibroblasts and cardiac immune cells, remains a major challenge in arrhythmia research because it can be investigated only in vivo. Various animal species have been used, and several disease models have been developed to study arrhythmias. Although every species is useful and might be ideal to study a specific hypothesis, we suggest a practical trio of animal models for future use: mice for genetic investigations, mechanistic evaluations or early studies to identify potential drug targets; rabbits for studies on ion channel function, repolarization or re-entrant arrhythmias; and pigs for preclinical translational studies to validate previous findings. In this Review, we provide a comprehensive overview of different models and currently used species for arrhythmia research, discuss their advantages and disadvantages and provide guidance for researchers who are considering performing in vivo studies.
Collapse
Affiliation(s)
- Sebastian Clauss
- Department of Medicine I, University Hospital Munich, Campus Grosshadern, Ludwig-Maximilians University Munich (LMU), Munich, Germany. .,DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany.
| | - Christina Bleyer
- Department of Medicine I, University Hospital Munich, Campus Grosshadern, Ludwig-Maximilians University Munich (LMU), Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
| | - Dominik Schüttler
- Department of Medicine I, University Hospital Munich, Campus Grosshadern, Ludwig-Maximilians University Munich (LMU), Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
| | - Philipp Tomsits
- Department of Medicine I, University Hospital Munich, Campus Grosshadern, Ludwig-Maximilians University Munich (LMU), Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
| | - Simone Renner
- Institute of Molecular Animal Breeding and Biotechnology, Ludwig-Maximilians University Munich (LMU), Munich, Germany.,DZD (German Centre for Diabetes Research), Neuherberg, Germany
| | - Nikolai Klymiuk
- Institute of Molecular Animal Breeding and Biotechnology, Ludwig-Maximilians University Munich (LMU), Munich, Germany
| | - Reza Wakili
- Universitätsklinikum Essen, Westdeutsches Herz- und Gefäßzentrum Essen, Essen, Germany
| | - Steffen Massberg
- Department of Medicine I, University Hospital Munich, Campus Grosshadern, Ludwig-Maximilians University Munich (LMU), Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
| | - Eckhard Wolf
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany.,Institute of Molecular Animal Breeding and Biotechnology, Ludwig-Maximilians University Munich (LMU), Munich, Germany.,DZD (German Centre for Diabetes Research), Neuherberg, Germany
| | - Stefan Kääb
- Department of Medicine I, University Hospital Munich, Campus Grosshadern, Ludwig-Maximilians University Munich (LMU), Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
| |
Collapse
|
23
|
West TM, Wang Q, Deng B, Zhang Y, Barbagallo F, Reddy GR, Chen D, Phan KS, Xu B, Isidori A, Xiang YK. Phosphodiesterase 5 Associates With β2 Adrenergic Receptor to Modulate Cardiac Function in Type 2 Diabetic Hearts. J Am Heart Assoc 2019; 8:e012273. [PMID: 31311394 PMCID: PMC6761630 DOI: 10.1161/jaha.119.012273] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background In murine heart failure models and in humans with diabetic‐related heart hypertrophy, inhibition of phosphodiesterase 5 (PDE5) by sildenafil improves cardiac outcomes. However, the mechanism by which sildenafil improves cardiac function is unclear. We have observed a relationship between PDE5 and β2 adrenergic receptor (β2AR), which is characterized here as a novel mechanistic axis by which sildenafil improves symptoms of diabetic cardiomyopathy. Methods and Results Wild‐type and β2AR knockout mice fed a high fat diet (HFD) were treated with sildenafil, and echocardiogram analysis was performed. Cardiomyocytes were isolated for excitation‐contraction (E‐C) coupling, fluorescence resonant energy transfer, and proximity ligation assays; while heart tissues were implemented for biochemical and histological analyses. PDE5 selectively associates with β2AR, but not β1 adrenergic receptor, and inhibition of PDE5 with sildenafil restores the impaired response to adrenergic stimulation in HFD mice and isolated ventriculomyocytes. Sildenafil enhances β adrenergic receptor (βAR)‐stimulated cGMP and cAMP signals in HFD myocytes. Consequently, inhibition of PDE5 leads to protein kinase G–, and to a lesser extent, calcium/calmodulin‐dependent kinase II–dependent improvements in adrenergically stimulated E‐C coupling. Deletion of β2AR abolishes sildenafil's effect. Although the PDE5‐β2AR association is not altered in HFD, phosphodiesterase 3 displays an increased association with the β2AR‐PDE5 complex in HFD myocytes. Conclusions This study elucidates mechanisms by which the β2AR‐PDE5 axis can be targeted for treating diabetic cardiomyopathy. Inhibition of PDE5 enhances β2AR stimulation of cGMP and cAMP signals, as well as protein kinase G–dependent E‐C coupling in HFD myocytes.
Collapse
Affiliation(s)
- Toni M West
- Department of Pharmacology University of California Davis School of Medicine Davis CA
| | - Qingtong Wang
- Department of Pharmacology University of California Davis School of Medicine Davis CA
| | - Bingqing Deng
- Department of Pharmacology University of California Davis School of Medicine Davis CA.,Sun-Yet Sen Memorial hospital Sun-Yet Sen University Guangzhou China
| | - Yu Zhang
- Department of Pharmacology University of California Davis School of Medicine Davis CA.,College of Pharmacy Guangzhou Medical University Guangzhou China
| | - Federica Barbagallo
- Department of Pharmacology University of California Davis School of Medicine Davis CA.,Department of Experimental Medicine Sapienza University of Rome Rome Italy
| | - Gopireddy R Reddy
- Department of Pharmacology University of California Davis School of Medicine Davis CA
| | - Dana Chen
- Department of Pharmacology University of California Davis School of Medicine Davis CA
| | - Kyle S Phan
- Department of Pharmacology University of California Davis School of Medicine Davis CA
| | - Bing Xu
- Department of Pharmacology University of California Davis School of Medicine Davis CA.,College of Pharmacy Guangzhou Medical University Guangzhou China
| | - Andres Isidori
- Department of Experimental Medicine Sapienza University of Rome Rome Italy
| | - Yang K Xiang
- Department of Pharmacology University of California Davis School of Medicine Davis CA.,VA Northern California Health Care System Mather CA
| |
Collapse
|
24
|
Modeling of LMNA-Related Dilated Cardiomyopathy Using Human Induced Pluripotent Stem Cells. Cells 2019; 8:cells8060594. [PMID: 31208058 PMCID: PMC6627421 DOI: 10.3390/cells8060594] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/12/2019] [Accepted: 06/13/2019] [Indexed: 12/31/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is one of the leading causes of heart failure and heart transplantation. A portion of familial DCM is due to mutations in the LMNA gene encoding the nuclear lamina proteins lamin A and C and without adequate treatment these patients have a poor prognosis. To get better insights into pathobiology behind this disease, we focused on modeling LMNA-related DCM using human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CM). Primary skin fibroblasts from DCM patients carrying the most prevalent Finnish founder mutation (p.S143P) in LMNA were reprogrammed into hiPSCs and further differentiated into cardiomyocytes (CMs). The cellular structure, functionality as well as gene and protein expression were assessed in detail. While mutant hiPSC-CMs presented virtually normal sarcomere structure under normoxia, dramatic sarcomere damage and an increased sensitivity to cellular stress was observed after hypoxia. A detailed electrophysiological evaluation revealed bradyarrhythmia and increased occurrence of arrhythmias in mutant hiPSC-CMs on β-adrenergic stimulation. Mutant hiPSC-CMs also showed increased sensitivity to hypoxia on microelectrode array and altered Ca2+ dynamics. Taken together, p.S143P hiPSC-CM model mimics hallmarks of LMNA-related DCM and provides a useful tool to study the underlying cellular mechanisms of accelerated cardiac degeneration in this disease.
Collapse
|
25
|
Hegyi B, Morotti S, Liu C, Ginsburg KS, Bossuyt J, Belardinelli L, Izu LT, Chen-Izu Y, Bányász T, Grandi E, Bers DM. Enhanced Depolarization Drive in Failing Rabbit Ventricular Myocytes: Calcium-Dependent and β-Adrenergic Effects on Late Sodium, L-Type Calcium, and Sodium-Calcium Exchange Currents. Circ Arrhythm Electrophysiol 2019; 12:e007061. [PMID: 30879336 PMCID: PMC6720130 DOI: 10.1161/circep.118.007061] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 02/13/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Heart failure (HF) is characterized by electrophysiological remodeling resulting in increased risk of cardiac arrhythmias. Previous reports suggest that elevated inward ionic currents in HF promote action potential (AP) prolongation, increased short-term variability of AP repolarization, and delayed afterdepolarizations. However, the underlying changes in late Na+ current (INaL), L-type Ca2+ current, and NCX (Na+/Ca2+ exchanger) current are often measured in nonphysiological conditions (square-pulse voltage clamp, slow pacing rates, exogenous Ca2+ buffers). METHODS We measured the major inward currents and their Ca2+- and β-adrenergic dependence under physiological AP clamp in rabbit ventricular myocytes in chronic pressure/volume overload-induced HF (versus age-matched control). RESULTS AP duration and short-term variability of AP repolarization were increased in HF, and importantly, inhibition of INaL decreased both parameters to the control level. INaL was slightly increased in HF versus control even when intracellular Ca2+ was strongly buffered. But under physiological AP clamp with normal Ca2+ cycling, INaL was markedly upregulated in HF versus control (dependent largely on CaMKII [Ca2+/calmodulin-dependent protein kinase II] activity). β-Adrenergic stimulation (often elevated in HF) further enhanced INaL. L-type Ca2+ current was decreased in HF when Ca2+ was buffered, but CaMKII-mediated Ca2+-dependent facilitation upregulated physiological L-type Ca2+ current to the control level. Furthermore, L-type Ca2+ current response to β-adrenergic stimulation was significantly attenuated in HF. Inward NCX current was upregulated at phase 3 of AP in HF when assessed by combining experimental data and computational modeling. CONCLUSIONS Our results suggest that CaMKII-dependent upregulation of INaL in HF significantly contributes to AP prolongation and increased short-term variability of AP repolarization, which may lead to increased arrhythmia propensity, and is further exacerbated by adrenergic stress.
Collapse
Affiliation(s)
- Bence Hegyi
- Department of Pharmacology, University of California Davis, Davis
| | - Stefano Morotti
- Department of Pharmacology, University of California Davis, Davis
| | - Caroline Liu
- Department of Pharmacology, University of California Davis, Davis
| | | | - Julie Bossuyt
- Department of Pharmacology, University of California Davis, Davis
| | | | - Leighton T. Izu
- Department of Pharmacology, University of California Davis, Davis
| | - Ye Chen-Izu
- Department of Pharmacology, University of California Davis, Davis
- Department of Biomedical Engineering, University of California Davis, Davis
- Department of Internal Medicine/Cardiology, University of California Davis, Davis
| | - Tamás Bányász
- Department of Pharmacology, University of California Davis, Davis
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Eleonora Grandi
- Department of Pharmacology, University of California Davis, Davis
| | - Donald M. Bers
- Department of Pharmacology, University of California Davis, Davis
| |
Collapse
|
26
|
Coppini R, Ferrantini C, Mugelli A, Poggesi C, Cerbai E. Altered Ca 2+ and Na + Homeostasis in Human Hypertrophic Cardiomyopathy: Implications for Arrhythmogenesis. Front Physiol 2018; 9:1391. [PMID: 30420810 PMCID: PMC6215954 DOI: 10.3389/fphys.2018.01391] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/13/2018] [Indexed: 12/12/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common mendelian heart disease, with a prevalence of 1/500. HCM is a primary cause of sudden death, due to an heightened risk of ventricular tachyarrhythmias that often occur in young asymptomatic patients. HCM can slowly progress toward heart failure, either with preserved or reduced ejection fraction, due to worsening of diastolic function. Accumulation of intra-myocardial fibrosis and replacement scars underlies heart failure progression and represents a substrate for sustained arrhythmias in end-stage patients. However, arrhythmias and mechanical abnormalities may occur in hearts with little or no fibrosis, prompting toward functional pathomechanisms. By studying viable cardiomyocytes and trabeculae isolated from inter-ventricular septum samples of non-failing HCM patients with symptomatic obstruction who underwent myectomy operations, we identified that specific abnormalities of intracellular Ca2+ handling are associated with increased cellular arrhytmogenesis and diastolic dysfunction. In HCM cardiomyocytes, diastolic Ca2+ concentration is increased both in the cytosol and in the sarcoplasmic reticulum and the rate of Ca2+ transient decay is slower, while the amplitude of Ca2+-release is preserved. Ca2+ overload is the consequence of an increased Ca2+ entry via L-type Ca2+-current [due to prolongation the action potential (AP) plateau], combined with a reduced rate of Ca2+-extrusion through the Na+/Ca2+ exchanger [due to increased cytosolic (Na+)] and a lower expression of SERCA. Increased late Na+ current (INaL) plays a major role, as it causes both AP prolongation and Na+ overload. Intracellular Ca2+ overload determines an higher frequency of Ca2+ waves leading to delayed-afterdepolarizations (DADs) and premature contractions, but is also linked with the increased diastolic tension and slower relaxation of HCM myocardium. Sustained increase of intracellular [Ca2+] goes hand-in-hand with the increased activation of Ca2+/calmodulin-dependent protein-kinase-II (CaMKII) and augmented phosphorylation of its targets, including Ca2+ handling proteins. In transgenic HCM mouse models, we found that Ca2+ overload, CaMKII and increased INaL drive myocardial remodeling since the earliest stages of disease and underlie the development of hypertrophy, diastolic dysfunction and the arrhythmogenic substrate. In conclusion, diastolic dysfunction and arrhythmogenesis in human HCM myocardium are driven by functional alterations at cellular and molecular level that may be targets of innovative therapies.
Collapse
Affiliation(s)
- Raffaele Coppini
- Department of Neuroscience, Psychology, Drug Sciences and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Cecilia Ferrantini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessandro Mugelli
- Department of Neuroscience, Psychology, Drug Sciences and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Corrado Poggesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Elisabetta Cerbai
- Department of Neuroscience, Psychology, Drug Sciences and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| |
Collapse
|
27
|
Johnson DM, Antoons G. Arrhythmogenic Mechanisms in Heart Failure: Linking β-Adrenergic Stimulation, Stretch, and Calcium. Front Physiol 2018; 9:1453. [PMID: 30374311 PMCID: PMC6196916 DOI: 10.3389/fphys.2018.01453] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/25/2018] [Indexed: 12/22/2022] Open
Abstract
Heart failure (HF) is associated with elevated sympathetic tone and mechanical load. Both systems activate signaling transduction pathways that increase cardiac output, but eventually become part of the disease process itself leading to further worsening of cardiac function. These alterations can adversely contribute to electrical instability, at least in part due to the modulation of Ca2+ handling at the level of the single cardiac myocyte. The major aim of this review is to provide a definitive overview of the links and cross talk between β-adrenergic stimulation, mechanical load, and arrhythmogenesis in the setting of HF. We will initially review the role of Ca2+ in the induction of both early and delayed afterdepolarizations, the role that β-adrenergic stimulation plays in the initiation of these and how the propensity for these may be altered in HF. We will then go onto reviewing the current data with regards to the link between mechanical load and afterdepolarizations, the associated mechano-sensitivity of the ryanodine receptor and other stretch activated channels that may be associated with HF-associated arrhythmias. Furthermore, we will discuss how alterations in local Ca2+ microdomains during the remodeling process associated the HF may contribute to the increased disposition for β-adrenergic or stretch induced arrhythmogenic triggers. Finally, the potential mechanisms linking β-adrenergic stimulation and mechanical stretch will be clarified, with the aim of finding common modalities of arrhythmogenesis that could be targeted by novel therapeutic agents in the setting of HF.
Collapse
Affiliation(s)
- Daniel M Johnson
- Department of Cardiothoracic Surgery, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Gudrun Antoons
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
28
|
Ferrantini C, Pioner JM, Mazzoni L, Gentile F, Tosi B, Rossi A, Belardinelli L, Tesi C, Palandri C, Matucci R, Cerbai E, Olivotto I, Poggesi C, Mugelli A, Coppini R. Late sodium current inhibitors to treat exercise-induced obstruction in hypertrophic cardiomyopathy: an in vitro study in human myocardium. Br J Pharmacol 2018; 175:2635-2652. [PMID: 29579779 PMCID: PMC6003658 DOI: 10.1111/bph.14223] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 12/19/2022] Open
Abstract
Background and Purpose In 30–40% of hypertrophic cardiomyopathy (HCM) patients, symptomatic left ventricular (LV) outflow gradients develop only during exercise due to catecholamine‐induced LV hypercontractility (inducible obstruction). Negative inotropic pharmacological options are limited to β‐blockers or disopyramide, with low efficacy and tolerability. We assessed the potential of late sodium current (INaL)‐inhibitors to treat inducible obstruction in HCM. Experimental Approach The electrophysiological and mechanical responses to β‐adrenoceptor stimulation were studied in human myocardium from HCM and control patients. Effects of INaL‐inhibitors (ranolazine and GS‐967) in HCM samples were investigated under conditions simulating rest and exercise. Key Results In cardiomyocytes and trabeculae from 18 surgical septal samples of patients with obstruction, the selective INaL‐inhibitor GS‐967 (0.5 μM) hastened twitch kinetics, decreased diastolic [Ca2+] and shortened action potentials, matching the effects of ranolazine (10μM). Mechanical responses to isoprenaline (inotropic and lusitropic) were comparable in HCM and control myocardium. However, isoprenaline prolonged action potentials in HCM myocardium, while it shortened them in controls. Unlike disopyramide, neither GS‐967 nor ranolazine reduced force at rest. However, in the presence of isoprenaline, they reduced Ca2+‐transient amplitude and twitch tension, while the acceleration of relaxation was maintained. INaL‐inhibitors were more effective than disopyramide in reducing contractility during exercise. Finally, INaL‐inhibitors abolished arrhythmias induced by isoprenaline. Conclusions and Implications Ranolazine and GS‐967 reduced septal myocardium tension during simulated exercise in vitro and therefore have the potential to ameliorate symptoms caused by inducible obstruction in HCM patients, with some advantages over disopyramide and β‐blockers.
Collapse
Affiliation(s)
- Cecilia Ferrantini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | - Josè Manuel Pioner
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Luca Mazzoni
- Department NeuroFarBa, University of Florence, Florence, Italy
| | - Francesca Gentile
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Benedetta Tosi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessandra Rossi
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | | | - Chiara Tesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Chiara Palandri
- Department NeuroFarBa, University of Florence, Florence, Italy
| | - Rosanna Matucci
- Department NeuroFarBa, University of Florence, Florence, Italy
| | | | - Iacopo Olivotto
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | - Corrado Poggesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | | |
Collapse
|
29
|
Schobesberger S, Wright P, Tokar S, Bhargava A, Mansfield C, Glukhov AV, Poulet C, Buzuk A, Monszpart A, Sikkel M, Harding SE, Nikolaev VO, Lyon AR, Gorelik J. T-tubule remodelling disturbs localized β2-adrenergic signalling in rat ventricular myocytes during the progression of heart failure. Cardiovasc Res 2018; 113:770-782. [PMID: 28505272 PMCID: PMC5437368 DOI: 10.1093/cvr/cvx074] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/11/2017] [Indexed: 11/12/2022] Open
Abstract
Aims Cardiomyocyte β2-adrenergic receptor (β2AR) cyclic adenosine monophosphate (cAMP) signalling is regulated by the receptors' subcellular location within transverse tubules (T-tubules), via interaction with structural and regulatory proteins, which form a signalosome. In chronic heart failure (HF), β2ARs redistribute from T-tubules to the cell surface, which disrupts functional signalosomes and leads to diffuse cAMP signalling. However, the functional consequences of structural changes upon β2AR-cAMP signalling during progression from hypertrophy to advanced HF are unknown. Methods and results Rat left ventricular myocytes were isolated at 4-, 8-, and 16-week post-myocardial infarction (MI), β2ARs were stimulated either via whole-cell perfusion or locally through the nanopipette of the scanning ion conductance microscope. cAMP release was measured via a Förster Resonance Energy Transfer-based sensor Epac2-camps. Confocal imaging of di-8-ANNEPS-stained cells and immunoblotting were used to determine structural alterations. At 4-week post-MI, T-tubule regularity, density and junctophilin-2 (JPH2) expression were significantly decreased. The amplitude of local β2AR-mediated cAMP in T-tubules was reduced and cAMP diffused throughout the cytosol instead of being locally confined. This was accompanied by partial caveolin-3 (Cav-3) dissociation from the membrane. At 8-week post-MI, the β2AR-mediated cAMP response was observed at the T-tubules and the sarcolemma (crest). Finally, at 16-week post-MI, the whole cell β2AR-mediated cAMP signal was depressed due to adenylate cyclase dysfunction, while overall Cav-3 levels were significantly increased and a substantial portion of Cav-3 dissociated into the cytosol. Overexpression of JPH2 in failing cells in vitro or AAV9.SERCA2a gene therapy in vivo did not improve β2AR-mediated signal compartmentation or reduce cAMP diffusion. Conclusion Although changes in T-tubule structure and β2AR-mediated cAMP signalling are significant even at 4-week post-MI, progression to the HF phenotype is not linear. At 8-week post-MI the loss of β2AR-mediated cAMP is temporarily reversed. Complete disorganization of β2AR-mediated cAMP signalling due to changes in functional receptor localization and cellular structure occurs at 16-week post-MI.
Collapse
Affiliation(s)
- Sophie Schobesberger
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12 0NN, UK.,Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Martinistraße, Hamburg D-20246, Germany
| | - Peter Wright
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Sergiy Tokar
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Anamika Bhargava
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12 0NN, UK.,Department of Biotechnology, Indian Institute of Technology Hyderabad, Ordnance Factory Estate, Yeddumailaram, 502205 Telangana, India
| | - Catherine Mansfield
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Alexey V Glukhov
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Claire Poulet
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Andrey Buzuk
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Aron Monszpart
- Department of Computer Science, University College London, Gower Street, London WC1E 6BT, UK
| | - Markus Sikkel
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Sian E Harding
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Martinistraße, Hamburg D-20246, Germany
| | - Alexander R Lyon
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12 0NN, UK.,NIHR Cardiovascular Biomedical Research Unit, Department of Cardiology, Royal Brompton Hospital, Sydney Street, London SW3 6NP, UK
| | - Julia Gorelik
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| |
Collapse
|
30
|
Hegyi B, Bossuyt J, Ginsburg KS, Mendoza LM, Talken L, Ferrier WT, Pogwizd SM, Izu LT, Chen-Izu Y, Bers DM. Altered Repolarization Reserve in Failing Rabbit Ventricular Myocytes: Calcium and β-Adrenergic Effects on Delayed- and Inward-Rectifier Potassium Currents. Circ Arrhythm Electrophysiol 2018; 11:e005852. [PMID: 29437761 PMCID: PMC5813707 DOI: 10.1161/circep.117.005852] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 12/11/2017] [Indexed: 12/25/2022]
Abstract
BACKGROUND Electrophysiological remodeling and increased susceptibility for cardiac arrhythmias are hallmarks of heart failure (HF). Ventricular action potential duration (APD) is typically prolonged in HF, with reduced repolarization reserve. However, underlying K+ current changes are often measured in nonphysiological conditions (voltage clamp, low pacing rates, cytosolic Ca2+ buffers). METHODS AND RESULTS We measured the major K+ currents (IKr, IKs, and IK1) and their Ca2+- and β-adrenergic dependence in rabbit ventricular myocytes in chronic pressure/volume overload-induced HF (versus age-matched controls). APD was significantly prolonged only at lower pacing rates (0.2-1 Hz) in HF under physiological ionic conditions and temperature. However, when cytosolic Ca2+ was buffered, APD prolongation in HF was also significant at higher pacing rates. Beat-to-beat variability of APD was also significantly increased in HF. Both IKr and IKs were significantly upregulated in HF under action potential clamp, but only when cytosolic Ca2+ was not buffered. CaMKII (Ca2+/calmodulin-dependent protein kinase II) inhibition abolished IKs upregulation in HF, but it did not affect IKr. IKs response to β-adrenergic stimulation was also significantly diminished in HF. IK1 was also decreased in HF regardless of Ca2+ buffering, CaMKII inhibition, or β-adrenergic stimulation. CONCLUSIONS At baseline Ca2+-dependent upregulation of IKr and IKs in HF counterbalances the reduced IK1, maintaining repolarization reserve (especially at higher heart rates) in physiological conditions, unlike conditions of strong cytosolic Ca2+ buffering. However, under β-adrenergic stimulation, reduced IKs responsiveness severely limits integrated repolarizing K+ current and repolarization reserve in HF. This would increase arrhythmia propensity in HF, especially during adrenergic stress.
Collapse
Affiliation(s)
- Bence Hegyi
- From the Department of Pharmacology (B.H., J.B., K.S.G., L.T.I., Y.C.-I., D.M.B.), School of Medicine, Dean's Office (L.T.), Surgical Research Facility, School of Medicine (W.T.F.), Department of Biomedical Engineering (Y.C.-I.), Department of Internal Medicine/Cardiology (Y.C.-I.), University of California, Davis; Echocardiography Laboratory, University of California, Davis Medical Center, Sacramento (L.M.M.); and Department of Medicine, University of Alabama at Birmingham (S.M.P.)
| | - Julie Bossuyt
- From the Department of Pharmacology (B.H., J.B., K.S.G., L.T.I., Y.C.-I., D.M.B.), School of Medicine, Dean's Office (L.T.), Surgical Research Facility, School of Medicine (W.T.F.), Department of Biomedical Engineering (Y.C.-I.), Department of Internal Medicine/Cardiology (Y.C.-I.), University of California, Davis; Echocardiography Laboratory, University of California, Davis Medical Center, Sacramento (L.M.M.); and Department of Medicine, University of Alabama at Birmingham (S.M.P.)
| | - Kenneth S Ginsburg
- From the Department of Pharmacology (B.H., J.B., K.S.G., L.T.I., Y.C.-I., D.M.B.), School of Medicine, Dean's Office (L.T.), Surgical Research Facility, School of Medicine (W.T.F.), Department of Biomedical Engineering (Y.C.-I.), Department of Internal Medicine/Cardiology (Y.C.-I.), University of California, Davis; Echocardiography Laboratory, University of California, Davis Medical Center, Sacramento (L.M.M.); and Department of Medicine, University of Alabama at Birmingham (S.M.P.)
| | - Lynette M Mendoza
- From the Department of Pharmacology (B.H., J.B., K.S.G., L.T.I., Y.C.-I., D.M.B.), School of Medicine, Dean's Office (L.T.), Surgical Research Facility, School of Medicine (W.T.F.), Department of Biomedical Engineering (Y.C.-I.), Department of Internal Medicine/Cardiology (Y.C.-I.), University of California, Davis; Echocardiography Laboratory, University of California, Davis Medical Center, Sacramento (L.M.M.); and Department of Medicine, University of Alabama at Birmingham (S.M.P.)
| | - Linda Talken
- From the Department of Pharmacology (B.H., J.B., K.S.G., L.T.I., Y.C.-I., D.M.B.), School of Medicine, Dean's Office (L.T.), Surgical Research Facility, School of Medicine (W.T.F.), Department of Biomedical Engineering (Y.C.-I.), Department of Internal Medicine/Cardiology (Y.C.-I.), University of California, Davis; Echocardiography Laboratory, University of California, Davis Medical Center, Sacramento (L.M.M.); and Department of Medicine, University of Alabama at Birmingham (S.M.P.)
| | - William T Ferrier
- From the Department of Pharmacology (B.H., J.B., K.S.G., L.T.I., Y.C.-I., D.M.B.), School of Medicine, Dean's Office (L.T.), Surgical Research Facility, School of Medicine (W.T.F.), Department of Biomedical Engineering (Y.C.-I.), Department of Internal Medicine/Cardiology (Y.C.-I.), University of California, Davis; Echocardiography Laboratory, University of California, Davis Medical Center, Sacramento (L.M.M.); and Department of Medicine, University of Alabama at Birmingham (S.M.P.)
| | - Steven M Pogwizd
- From the Department of Pharmacology (B.H., J.B., K.S.G., L.T.I., Y.C.-I., D.M.B.), School of Medicine, Dean's Office (L.T.), Surgical Research Facility, School of Medicine (W.T.F.), Department of Biomedical Engineering (Y.C.-I.), Department of Internal Medicine/Cardiology (Y.C.-I.), University of California, Davis; Echocardiography Laboratory, University of California, Davis Medical Center, Sacramento (L.M.M.); and Department of Medicine, University of Alabama at Birmingham (S.M.P.)
| | - Leighton T Izu
- From the Department of Pharmacology (B.H., J.B., K.S.G., L.T.I., Y.C.-I., D.M.B.), School of Medicine, Dean's Office (L.T.), Surgical Research Facility, School of Medicine (W.T.F.), Department of Biomedical Engineering (Y.C.-I.), Department of Internal Medicine/Cardiology (Y.C.-I.), University of California, Davis; Echocardiography Laboratory, University of California, Davis Medical Center, Sacramento (L.M.M.); and Department of Medicine, University of Alabama at Birmingham (S.M.P.)
| | - Ye Chen-Izu
- From the Department of Pharmacology (B.H., J.B., K.S.G., L.T.I., Y.C.-I., D.M.B.), School of Medicine, Dean's Office (L.T.), Surgical Research Facility, School of Medicine (W.T.F.), Department of Biomedical Engineering (Y.C.-I.), Department of Internal Medicine/Cardiology (Y.C.-I.), University of California, Davis; Echocardiography Laboratory, University of California, Davis Medical Center, Sacramento (L.M.M.); and Department of Medicine, University of Alabama at Birmingham (S.M.P.)
| | - Donald M Bers
- From the Department of Pharmacology (B.H., J.B., K.S.G., L.T.I., Y.C.-I., D.M.B.), School of Medicine, Dean's Office (L.T.), Surgical Research Facility, School of Medicine (W.T.F.), Department of Biomedical Engineering (Y.C.-I.), Department of Internal Medicine/Cardiology (Y.C.-I.), University of California, Davis; Echocardiography Laboratory, University of California, Davis Medical Center, Sacramento (L.M.M.); and Department of Medicine, University of Alabama at Birmingham (S.M.P.).
| |
Collapse
|
31
|
Abstract
Myocardial injury, mechanical stress, neurohormonal activation, inflammation, and/or aging all lead to cardiac remodeling, which is responsible for cardiac dysfunction and arrhythmogenesis. Of the key histological components of cardiac remodeling, fibrosis either in the form of interstitial, patchy, or dense scars, constitutes a key histological substrate of arrhythmias. Here we discuss current research findings focusing on the role of fibrosis, in arrhythmogenesis. Numerous studies have convincingly shown that patchy or interstitial fibrosis interferes with myocardial electrophysiology by slowing down action potential propagation, initiating reentry, promoting after-depolarizations, and increasing ectopic automaticity. Meanwhile, there has been increasing appreciation of direct involvement of myofibroblasts, the activated form of fibroblasts, in arrhythmogenesis. Myofibroblasts undergo phenotypic changes with expression of gap-junctions and ion channels thereby forming direct electrical coupling with cardiomyocytes, which potentially results in profound disturbances of electrophysiology. There is strong evidence that systemic and regional inflammatory processes contribute to fibrogenesis (i.e., structural remodeling) and dysfunction of ion channels and Ca2+ homeostasis (i.e., electrical remodeling). Recognizing the pivotal role of fibrosis in the arrhythmogenesis has promoted clinical research on characterizing fibrosis by means of cardiac imaging or fibrosis biomarkers for clinical stratification of patients at higher risk of lethal arrhythmia, as well as preclinical research on the development of antifibrotic therapies. At the end of this review, we discuss remaining key questions in this area and propose new research approaches. © 2017 American Physiological Society. Compr Physiol 7:1009-1049, 2017.
Collapse
Affiliation(s)
- My-Nhan Nguyen
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia
| | - Helen Kiriazis
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Xiao-Ming Gao
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia
| | - Xiao-Jun Du
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia
| |
Collapse
|
32
|
Rozier K, Bondarenko VE. Distinct physiological effects of β1- and β2-adrenoceptors in mouse ventricular myocytes: insights from a compartmentalized mathematical model. Am J Physiol Cell Physiol 2017; 312:C595-C623. [DOI: 10.1152/ajpcell.00273.2016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 01/03/2017] [Accepted: 01/18/2017] [Indexed: 01/08/2023]
Abstract
The β1- and β2-adrenergic signaling systems play different roles in the functioning of cardiac cells. Experimental data show that the activation of the β1-adrenergic signaling system produces significant inotropic, lusitropic, and chronotropic effects in the heart, whereas the effects of the β2-adrenergic signaling system is less apparent. In this paper, a comprehensive compartmentalized experimentally based mathematical model of the combined β1- and β2-adrenergic signaling systems in mouse ventricular myocytes is developed to simulate the experimental findings and make testable predictions of the behavior of the cardiac cells under different physiological conditions. Simulations describe the dynamics of major signaling molecules in different subcellular compartments; kinetics and magnitudes of phosphorylation of ion channels, transporters, and Ca2+ handling proteins; modifications of action potential shape and duration; and [Ca2+]i and [Na+]i dynamics upon stimulation of β1- and β2-adrenergic receptors (β1- and β2-ARs). The model reveals physiological conditions when β2-ARs do not produce significant physiological effects and when their effects can be measured experimentally. Simulations demonstrated that stimulation of β2-ARs with isoproterenol caused a marked increase in the magnitude of the L-type Ca2+ current, [Ca2+]i transient, and phosphorylation of phospholamban only upon additional application of pertussis toxin or inhibition of phosphodiesterases of type 3 and 4. The model also made testable predictions of the changes in magnitudes of [Ca2+]i and [Na+]i fluxes, the rate of decay of [Na+]i concentration upon both combined and separate stimulation of β1- and β2-ARs, and the contribution of phosphorylation of PKA targets to the changes in the action potential and [Ca2+]i transient.
Collapse
Affiliation(s)
- Kelvin Rozier
- Department of Mathematics and Statistics, Georgia State University, Atlanta, Georgia; and
| | - Vladimir E. Bondarenko
- Department of Mathematics and Statistics, Georgia State University, Atlanta, Georgia; and
- Neuroscience Institute, Georgia State University, Atlanta, Georgia
| |
Collapse
|
33
|
Masarone D, Limongelli G, Rubino M, Valente F, Vastarella R, Ammendola E, Gravino R, Verrengia M, Salerno G, Pacileo G. Management of Arrhythmias in Heart Failure. J Cardiovasc Dev Dis 2017; 4:E3. [PMID: 29367535 PMCID: PMC5715690 DOI: 10.3390/jcdd4010003] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 02/03/2017] [Accepted: 02/22/2017] [Indexed: 12/14/2022] Open
Abstract
Heart failure patients are predisposed to develop arrhythmias. Supraventricular arrhythmias can exacerbate the heart failure symptoms by decreasing the effective cardiac output and their control require pharmacological, electrical, or catheter-based intervention. In the setting of atrial flutter or atrial fibrillation, anticoagulation becomes paramount to prevent systemic or cerebral embolism. Patients with heart failure are also prone to develop ventricular arrhythmias that can present a challenge to the managing clinician. The management strategy depends on the type of arrhythmia, the underlying structural heart disease, the severity of heart failure, and the range from optimization of heart failure therapy to catheter ablation. Patients with heart failure, irrespective of ejection fraction are at high risk for developing sudden cardiac death, however risk stratification is a clinical challenge and requires a multiparametric evaluation for identification of patients who should undergo implantation of a cardioverter defibrillator. Finally, patients with heart failure can also develop symptomatic bradycardia, caused by sinus node dysfunction or atrio-ventricular block. The treatment of bradycardia in these patients with pacing is usually straightforward but needs some specific issue.
Collapse
Affiliation(s)
- Daniele Masarone
- Cardiologia SUN-Heart Failure Unit, Department of Cardiothoracic Sciences, Second University of Naples, via L. Bianchi, Naples 80100, Italy.
| | - Giuseppe Limongelli
- Cardiologia SUN-Heart Failure Unit, Department of Cardiothoracic Sciences, Second University of Naples, via L. Bianchi, Naples 80100, Italy.
| | - Marta Rubino
- Cardiologia SUN-Heart Failure Unit, Department of Cardiothoracic Sciences, Second University of Naples, via L. Bianchi, Naples 80100, Italy.
| | - Fabio Valente
- Cardiologia SUN-Heart Failure Unit, Department of Cardiothoracic Sciences, Second University of Naples, via L. Bianchi, Naples 80100, Italy.
| | - Rossella Vastarella
- Cardiologia SUN-Heart Failure Unit, Department of Cardiothoracic Sciences, Second University of Naples, via L. Bianchi, Naples 80100, Italy.
| | - Ernesto Ammendola
- Cardiologia SUN-Heart Failure Unit, Department of Cardiothoracic Sciences, Second University of Naples, via L. Bianchi, Naples 80100, Italy.
| | - Rita Gravino
- Cardiologia SUN-Heart Failure Unit, Department of Cardiothoracic Sciences, Second University of Naples, via L. Bianchi, Naples 80100, Italy.
| | - Marina Verrengia
- Cardiologia SUN-Heart Failure Unit, Department of Cardiothoracic Sciences, Second University of Naples, via L. Bianchi, Naples 80100, Italy.
| | - Gemma Salerno
- Cardiologia SUN-Heart Failure Unit, Department of Cardiothoracic Sciences, Second University of Naples, via L. Bianchi, Naples 80100, Italy.
| | - Giuseppe Pacileo
- Cardiologia SUN-Heart Failure Unit, Department of Cardiothoracic Sciences, Second University of Naples, via L. Bianchi, Naples 80100, Italy.
| |
Collapse
|
34
|
Myagmar BE, Flynn JM, Cowley PM, Swigart PM, Montgomery MD, Thai K, Nair D, Gupta R, Deng DX, Hosoda C, Melov S, Baker AJ, Simpson PC. Adrenergic Receptors in Individual Ventricular Myocytes: The Beta-1 and Alpha-1B Are in All Cells, the Alpha-1A Is in a Subpopulation, and the Beta-2 and Beta-3 Are Mostly Absent. Circ Res 2017; 120:1103-1115. [PMID: 28219977 DOI: 10.1161/circresaha.117.310520] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 02/13/2017] [Accepted: 02/17/2017] [Indexed: 12/20/2022]
Abstract
RATIONALE It is unknown whether every ventricular myocyte expresses all 5 of the cardiac adrenergic receptors (ARs), β1, β2, β3, α1A, and α1B. The β1 and β2 are thought to be the dominant myocyte ARs. OBJECTIVE Quantify the 5 cardiac ARs in individual ventricular myocytes. METHODS AND RESULTS We studied ventricular myocytes from wild-type mice, mice with α1A and α1B knockin reporters, and β1 and β2 knockout mice. Using individual isolated cells, we measured knockin reporters, mRNAs, signaling (phosphorylation of extracellular signal-regulated kinase and phospholamban), and contraction. We found that the β1 and α1B were present in all myocytes. The α1A was present in 60%, with high levels in 20%. The β2 and β3 were detected in only ≈5% of myocytes, mostly in different cells. In intact heart, 30% of total β-ARs were β2 and 20% were β3, both mainly in nonmyocytes. CONCLUSION The dominant ventricular myocyte ARs present in all cells are the β1 and α1B. The β2 and β3 are mostly absent in myocytes but are abundant in nonmyocytes. The α1A is in just over half of cells, but only 20% have high levels. Four distinct myocyte AR phenotypes are defined: 30% of cells with β1 and α1B only; 60% that also have the α1A; and 5% each that also have the β2 or β3. The results raise cautions in experimental design, such as receptor overexpression in myocytes that do not express the AR normally. The data suggest new paradigms in cardiac adrenergic signaling mechanisms.
Collapse
Affiliation(s)
- Bat-Erdene Myagmar
- From the Department of Medicine, VA Medical Center, San Francisco, CA (B.-E.M., P.M.C., P.M.S., M.D.M., K.T., D.N., R.G., D.X.D., C.H., A.J.B., P.C.S.); Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco (B.-E.M., P.M.C., M.D.M., D.X.D., C.H., A.J.B., P.C.S.); and Buck Institute for Research on Aging, Novato, CA (J.M.F., S.M.)
| | - James M Flynn
- From the Department of Medicine, VA Medical Center, San Francisco, CA (B.-E.M., P.M.C., P.M.S., M.D.M., K.T., D.N., R.G., D.X.D., C.H., A.J.B., P.C.S.); Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco (B.-E.M., P.M.C., M.D.M., D.X.D., C.H., A.J.B., P.C.S.); and Buck Institute for Research on Aging, Novato, CA (J.M.F., S.M.)
| | - Patrick M Cowley
- From the Department of Medicine, VA Medical Center, San Francisco, CA (B.-E.M., P.M.C., P.M.S., M.D.M., K.T., D.N., R.G., D.X.D., C.H., A.J.B., P.C.S.); Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco (B.-E.M., P.M.C., M.D.M., D.X.D., C.H., A.J.B., P.C.S.); and Buck Institute for Research on Aging, Novato, CA (J.M.F., S.M.)
| | - Philip M Swigart
- From the Department of Medicine, VA Medical Center, San Francisco, CA (B.-E.M., P.M.C., P.M.S., M.D.M., K.T., D.N., R.G., D.X.D., C.H., A.J.B., P.C.S.); Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco (B.-E.M., P.M.C., M.D.M., D.X.D., C.H., A.J.B., P.C.S.); and Buck Institute for Research on Aging, Novato, CA (J.M.F., S.M.)
| | - Megan D Montgomery
- From the Department of Medicine, VA Medical Center, San Francisco, CA (B.-E.M., P.M.C., P.M.S., M.D.M., K.T., D.N., R.G., D.X.D., C.H., A.J.B., P.C.S.); Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco (B.-E.M., P.M.C., M.D.M., D.X.D., C.H., A.J.B., P.C.S.); and Buck Institute for Research on Aging, Novato, CA (J.M.F., S.M.)
| | - Kevin Thai
- From the Department of Medicine, VA Medical Center, San Francisco, CA (B.-E.M., P.M.C., P.M.S., M.D.M., K.T., D.N., R.G., D.X.D., C.H., A.J.B., P.C.S.); Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco (B.-E.M., P.M.C., M.D.M., D.X.D., C.H., A.J.B., P.C.S.); and Buck Institute for Research on Aging, Novato, CA (J.M.F., S.M.)
| | - Divya Nair
- From the Department of Medicine, VA Medical Center, San Francisco, CA (B.-E.M., P.M.C., P.M.S., M.D.M., K.T., D.N., R.G., D.X.D., C.H., A.J.B., P.C.S.); Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco (B.-E.M., P.M.C., M.D.M., D.X.D., C.H., A.J.B., P.C.S.); and Buck Institute for Research on Aging, Novato, CA (J.M.F., S.M.)
| | - Rumita Gupta
- From the Department of Medicine, VA Medical Center, San Francisco, CA (B.-E.M., P.M.C., P.M.S., M.D.M., K.T., D.N., R.G., D.X.D., C.H., A.J.B., P.C.S.); Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco (B.-E.M., P.M.C., M.D.M., D.X.D., C.H., A.J.B., P.C.S.); and Buck Institute for Research on Aging, Novato, CA (J.M.F., S.M.)
| | - David X Deng
- From the Department of Medicine, VA Medical Center, San Francisco, CA (B.-E.M., P.M.C., P.M.S., M.D.M., K.T., D.N., R.G., D.X.D., C.H., A.J.B., P.C.S.); Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco (B.-E.M., P.M.C., M.D.M., D.X.D., C.H., A.J.B., P.C.S.); and Buck Institute for Research on Aging, Novato, CA (J.M.F., S.M.)
| | - Chihiro Hosoda
- From the Department of Medicine, VA Medical Center, San Francisco, CA (B.-E.M., P.M.C., P.M.S., M.D.M., K.T., D.N., R.G., D.X.D., C.H., A.J.B., P.C.S.); Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco (B.-E.M., P.M.C., M.D.M., D.X.D., C.H., A.J.B., P.C.S.); and Buck Institute for Research on Aging, Novato, CA (J.M.F., S.M.)
| | - Simon Melov
- From the Department of Medicine, VA Medical Center, San Francisco, CA (B.-E.M., P.M.C., P.M.S., M.D.M., K.T., D.N., R.G., D.X.D., C.H., A.J.B., P.C.S.); Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco (B.-E.M., P.M.C., M.D.M., D.X.D., C.H., A.J.B., P.C.S.); and Buck Institute for Research on Aging, Novato, CA (J.M.F., S.M.)
| | - Anthony J Baker
- From the Department of Medicine, VA Medical Center, San Francisco, CA (B.-E.M., P.M.C., P.M.S., M.D.M., K.T., D.N., R.G., D.X.D., C.H., A.J.B., P.C.S.); Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco (B.-E.M., P.M.C., M.D.M., D.X.D., C.H., A.J.B., P.C.S.); and Buck Institute for Research on Aging, Novato, CA (J.M.F., S.M.)
| | - Paul C Simpson
- From the Department of Medicine, VA Medical Center, San Francisco, CA (B.-E.M., P.M.C., P.M.S., M.D.M., K.T., D.N., R.G., D.X.D., C.H., A.J.B., P.C.S.); Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco (B.-E.M., P.M.C., M.D.M., D.X.D., C.H., A.J.B., P.C.S.); and Buck Institute for Research on Aging, Novato, CA (J.M.F., S.M.).
| |
Collapse
|
35
|
Kotlo K, Samarel AM, Chen HY, Aldstadt J, Danziger RS. Global comparison of phosphoproteins in human and rodent hearts: implications for translational studies of myosin light chain and troponin phosphorylations. SPRINGERPLUS 2016; 5:808. [PMID: 27390648 PMCID: PMC4916117 DOI: 10.1186/s40064-016-2469-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 05/29/2016] [Indexed: 11/10/2022]
Abstract
Cardiac remodeling and failure are regulated by a myriad of cardiac protein phosphorylations. In the present study, cardiac phosphoprotein patterns were examined in rodent and human hearts Left ventricular tissue samples were obtained from human systolic failing (n = 5) and control (n = 5) hearts and from two rat models of hypertensive heart failure, i.e., spontaneously hypertensive heart failure and Dahl salt-sensitive rats and corresponding controls. Phosphoproteins were separated by 2D-DIGE with Cydye staining, phosphoprotein patterns were analyzed using pixel intensity in rectified images. Specific phosphoproteins which were different in human versus rodent hearts were identified by MALDI-TOF/TOF Mass Spectrometry. Targeted pair-wise analyses showed differences (p < 0.05) in 26 % of the pixels, which included pixels containing phosphorylated troponin T, myosin light chain, peroxiredoxin, and haptoglobin. These results show differences in rodent versus human cardiac remodeling which will influence the translation rodent studies to humans in this area.
Collapse
|
36
|
Effects of zacopride, a moderate I K1 channel agonist, on triggered arrhythmia and contractility in human ventricular myocardium. Pharmacol Res 2016; 115:309-318. [PMID: 27914945 DOI: 10.1016/j.phrs.2016.11.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 10/13/2016] [Accepted: 11/20/2016] [Indexed: 11/23/2022]
Abstract
Ventricular tachycardia is the leading cause of sudden arrhythmic death in the U.S. Recently, the moderate IK1 channel activator, zacopride, was shown to suppress triggered ventricular tachycardia in rats. Nonetheless, concerns were raised about the possibility of pro-arrhythmic activity after IK1 channel stimulation based on the promising anti-arrhythmic strategy of IK1 blockade in other animal models. Therefore, the goal of the current study was to investigate the ex-vivo effects of zacopride on triggered arrhythmia and contractility in ventricular human myocardium in order to validate data that was solely obtained from animal models. Application of 100nmol/L isoproterenol and 0.5mmol/L caffeine led to triggered arrhythmia in isolated cardiac muscles from non-failing and end-stage failing hearts. However, the occurrence of arrhythmia in muscles of non-failing hearts was markedly higher than those of end-stage failing hearts. Interestingly, zacopride eliminated the ex-vivo triggered arrhythmia in these muscles of non-failing and failing hearts in a concentration-dependent manner, with an effective IC50 in the range of 28-40μmol/L. Conversely, in the absence of isoproterenol/caffeine, zacopride led to a negative inotropic effect in a concentration-dependent manner. Reduced cardiac contraction was clearly observed at high zacopride concentration of 200μmol/L, along with the occurrence of contractile alternans in muscles of non-failing and failing hearts. Zacopride shows promising antiarrhythmic effects against triggered arrhythmia in human ventricular myocardium. However, in the absence of Ca2+ overload/arrhythmia, zacopride, albeit at high concentrations, decreases the force of contraction and increases the likelihood of occurrence of contractile alternans, which may predispose the heart to contractile dysfunction and/or arrhythmia. Overall, our results represent a key step in translating this drug from the benchtop to the bedside in the research area.
Collapse
|
37
|
Barbagallo F, Xu B, Reddy GR, West T, Wang Q, Fu Q, Li M, Shi Q, Ginsburg KS, Ferrier W, Isidori AM, Naro F, Patel HH, Bossuyt J, Bers D, Xiang YK. Genetically Encoded Biosensors Reveal PKA Hyperphosphorylation on the Myofilaments in Rabbit Heart Failure. Circ Res 2016; 119:931-43. [PMID: 27576469 DOI: 10.1161/circresaha.116.308964] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 08/29/2016] [Indexed: 01/05/2023]
Abstract
RATIONALE In heart failure, myofilament proteins display abnormal phosphorylation, which contributes to contractile dysfunction. The mechanisms underlying the dysregulation of protein phosphorylation on myofilaments is not clear. OBJECTIVE This study aims to understand the mechanisms underlying altered phosphorylation of myofilament proteins in heart failure. METHODS AND RESULTS We generate a novel genetically encoded protein kinase A (PKA) biosensor anchored onto the myofilaments in rabbit cardiac myocytes to examine PKA activity at the myofilaments in responses to adrenergic stimulation. We show that PKA activity is shifted from the sarcolemma to the myofilaments in hypertrophic failing rabbit myocytes. In particular, the increased PKA activity on the myofilaments is because of an enhanced β2 adrenergic receptor signal selectively directed to the myofilaments together with a reduced phosphodiesterase activity associated with the myofibrils. Mechanistically, the enhanced PKA activity on the myofilaments is associated with downregulation of caveolin-3 in the hypertrophic failing rabbit myocytes. Reintroduction of caveolin-3 in the failing myocytes is able to normalize the distribution of β2 adrenergic receptor signal by preventing PKA signal access to the myofilaments and to restore contractile response to adrenergic stimulation. CONCLUSIONS In hypertrophic rabbit myocytes, selectively enhanced β2 adrenergic receptor signaling toward the myofilaments contributes to elevated PKA activity and PKA phosphorylation of myofilament proteins. Reintroduction of caveolin-3 is able to confine β2 adrenergic receptor signaling and restore myocyte contractility in response to β adrenergic stimulation.
Collapse
Affiliation(s)
- Federica Barbagallo
- From the Department of Pharmacology, University of California at Davis (F.B., B.X., G.R.R., T.W., Q.W., Q.F., M.L., Q.S., K.S.G., J.B., D.B., Y.K.X.); Department of Experimental Medicine (F.B., A.M.I.) and Department of Anatomical, Histological, Forensic, and Orthopedic Sciences (F.N.), Sapienza University of Rome, Italy; Department of Medicine and Epidemiology, School of Veterinary Medicine, and Surgical Research Facility, School of Medicine, University of California, Davis (W.F.); VA San Diego Healthcare System, La Jolla, CA (H.H.P.); Department of Anesthesiology, University of California, San Diego, La Jolla (H.H.P.); and VA Northern California Healthcare System, Mather (Y.K.X.)
| | - Bing Xu
- From the Department of Pharmacology, University of California at Davis (F.B., B.X., G.R.R., T.W., Q.W., Q.F., M.L., Q.S., K.S.G., J.B., D.B., Y.K.X.); Department of Experimental Medicine (F.B., A.M.I.) and Department of Anatomical, Histological, Forensic, and Orthopedic Sciences (F.N.), Sapienza University of Rome, Italy; Department of Medicine and Epidemiology, School of Veterinary Medicine, and Surgical Research Facility, School of Medicine, University of California, Davis (W.F.); VA San Diego Healthcare System, La Jolla, CA (H.H.P.); Department of Anesthesiology, University of California, San Diego, La Jolla (H.H.P.); and VA Northern California Healthcare System, Mather (Y.K.X.)
| | - Gopireddy R Reddy
- From the Department of Pharmacology, University of California at Davis (F.B., B.X., G.R.R., T.W., Q.W., Q.F., M.L., Q.S., K.S.G., J.B., D.B., Y.K.X.); Department of Experimental Medicine (F.B., A.M.I.) and Department of Anatomical, Histological, Forensic, and Orthopedic Sciences (F.N.), Sapienza University of Rome, Italy; Department of Medicine and Epidemiology, School of Veterinary Medicine, and Surgical Research Facility, School of Medicine, University of California, Davis (W.F.); VA San Diego Healthcare System, La Jolla, CA (H.H.P.); Department of Anesthesiology, University of California, San Diego, La Jolla (H.H.P.); and VA Northern California Healthcare System, Mather (Y.K.X.)
| | - Toni West
- From the Department of Pharmacology, University of California at Davis (F.B., B.X., G.R.R., T.W., Q.W., Q.F., M.L., Q.S., K.S.G., J.B., D.B., Y.K.X.); Department of Experimental Medicine (F.B., A.M.I.) and Department of Anatomical, Histological, Forensic, and Orthopedic Sciences (F.N.), Sapienza University of Rome, Italy; Department of Medicine and Epidemiology, School of Veterinary Medicine, and Surgical Research Facility, School of Medicine, University of California, Davis (W.F.); VA San Diego Healthcare System, La Jolla, CA (H.H.P.); Department of Anesthesiology, University of California, San Diego, La Jolla (H.H.P.); and VA Northern California Healthcare System, Mather (Y.K.X.)
| | - Qingtong Wang
- From the Department of Pharmacology, University of California at Davis (F.B., B.X., G.R.R., T.W., Q.W., Q.F., M.L., Q.S., K.S.G., J.B., D.B., Y.K.X.); Department of Experimental Medicine (F.B., A.M.I.) and Department of Anatomical, Histological, Forensic, and Orthopedic Sciences (F.N.), Sapienza University of Rome, Italy; Department of Medicine and Epidemiology, School of Veterinary Medicine, and Surgical Research Facility, School of Medicine, University of California, Davis (W.F.); VA San Diego Healthcare System, La Jolla, CA (H.H.P.); Department of Anesthesiology, University of California, San Diego, La Jolla (H.H.P.); and VA Northern California Healthcare System, Mather (Y.K.X.)
| | - Qin Fu
- From the Department of Pharmacology, University of California at Davis (F.B., B.X., G.R.R., T.W., Q.W., Q.F., M.L., Q.S., K.S.G., J.B., D.B., Y.K.X.); Department of Experimental Medicine (F.B., A.M.I.) and Department of Anatomical, Histological, Forensic, and Orthopedic Sciences (F.N.), Sapienza University of Rome, Italy; Department of Medicine and Epidemiology, School of Veterinary Medicine, and Surgical Research Facility, School of Medicine, University of California, Davis (W.F.); VA San Diego Healthcare System, La Jolla, CA (H.H.P.); Department of Anesthesiology, University of California, San Diego, La Jolla (H.H.P.); and VA Northern California Healthcare System, Mather (Y.K.X.)
| | - Minghui Li
- From the Department of Pharmacology, University of California at Davis (F.B., B.X., G.R.R., T.W., Q.W., Q.F., M.L., Q.S., K.S.G., J.B., D.B., Y.K.X.); Department of Experimental Medicine (F.B., A.M.I.) and Department of Anatomical, Histological, Forensic, and Orthopedic Sciences (F.N.), Sapienza University of Rome, Italy; Department of Medicine and Epidemiology, School of Veterinary Medicine, and Surgical Research Facility, School of Medicine, University of California, Davis (W.F.); VA San Diego Healthcare System, La Jolla, CA (H.H.P.); Department of Anesthesiology, University of California, San Diego, La Jolla (H.H.P.); and VA Northern California Healthcare System, Mather (Y.K.X.)
| | - Qian Shi
- From the Department of Pharmacology, University of California at Davis (F.B., B.X., G.R.R., T.W., Q.W., Q.F., M.L., Q.S., K.S.G., J.B., D.B., Y.K.X.); Department of Experimental Medicine (F.B., A.M.I.) and Department of Anatomical, Histological, Forensic, and Orthopedic Sciences (F.N.), Sapienza University of Rome, Italy; Department of Medicine and Epidemiology, School of Veterinary Medicine, and Surgical Research Facility, School of Medicine, University of California, Davis (W.F.); VA San Diego Healthcare System, La Jolla, CA (H.H.P.); Department of Anesthesiology, University of California, San Diego, La Jolla (H.H.P.); and VA Northern California Healthcare System, Mather (Y.K.X.)
| | - Kenneth S Ginsburg
- From the Department of Pharmacology, University of California at Davis (F.B., B.X., G.R.R., T.W., Q.W., Q.F., M.L., Q.S., K.S.G., J.B., D.B., Y.K.X.); Department of Experimental Medicine (F.B., A.M.I.) and Department of Anatomical, Histological, Forensic, and Orthopedic Sciences (F.N.), Sapienza University of Rome, Italy; Department of Medicine and Epidemiology, School of Veterinary Medicine, and Surgical Research Facility, School of Medicine, University of California, Davis (W.F.); VA San Diego Healthcare System, La Jolla, CA (H.H.P.); Department of Anesthesiology, University of California, San Diego, La Jolla (H.H.P.); and VA Northern California Healthcare System, Mather (Y.K.X.)
| | - William Ferrier
- From the Department of Pharmacology, University of California at Davis (F.B., B.X., G.R.R., T.W., Q.W., Q.F., M.L., Q.S., K.S.G., J.B., D.B., Y.K.X.); Department of Experimental Medicine (F.B., A.M.I.) and Department of Anatomical, Histological, Forensic, and Orthopedic Sciences (F.N.), Sapienza University of Rome, Italy; Department of Medicine and Epidemiology, School of Veterinary Medicine, and Surgical Research Facility, School of Medicine, University of California, Davis (W.F.); VA San Diego Healthcare System, La Jolla, CA (H.H.P.); Department of Anesthesiology, University of California, San Diego, La Jolla (H.H.P.); and VA Northern California Healthcare System, Mather (Y.K.X.)
| | - Andrea M Isidori
- From the Department of Pharmacology, University of California at Davis (F.B., B.X., G.R.R., T.W., Q.W., Q.F., M.L., Q.S., K.S.G., J.B., D.B., Y.K.X.); Department of Experimental Medicine (F.B., A.M.I.) and Department of Anatomical, Histological, Forensic, and Orthopedic Sciences (F.N.), Sapienza University of Rome, Italy; Department of Medicine and Epidemiology, School of Veterinary Medicine, and Surgical Research Facility, School of Medicine, University of California, Davis (W.F.); VA San Diego Healthcare System, La Jolla, CA (H.H.P.); Department of Anesthesiology, University of California, San Diego, La Jolla (H.H.P.); and VA Northern California Healthcare System, Mather (Y.K.X.)
| | - Fabio Naro
- From the Department of Pharmacology, University of California at Davis (F.B., B.X., G.R.R., T.W., Q.W., Q.F., M.L., Q.S., K.S.G., J.B., D.B., Y.K.X.); Department of Experimental Medicine (F.B., A.M.I.) and Department of Anatomical, Histological, Forensic, and Orthopedic Sciences (F.N.), Sapienza University of Rome, Italy; Department of Medicine and Epidemiology, School of Veterinary Medicine, and Surgical Research Facility, School of Medicine, University of California, Davis (W.F.); VA San Diego Healthcare System, La Jolla, CA (H.H.P.); Department of Anesthesiology, University of California, San Diego, La Jolla (H.H.P.); and VA Northern California Healthcare System, Mather (Y.K.X.)
| | - Hemal H Patel
- From the Department of Pharmacology, University of California at Davis (F.B., B.X., G.R.R., T.W., Q.W., Q.F., M.L., Q.S., K.S.G., J.B., D.B., Y.K.X.); Department of Experimental Medicine (F.B., A.M.I.) and Department of Anatomical, Histological, Forensic, and Orthopedic Sciences (F.N.), Sapienza University of Rome, Italy; Department of Medicine and Epidemiology, School of Veterinary Medicine, and Surgical Research Facility, School of Medicine, University of California, Davis (W.F.); VA San Diego Healthcare System, La Jolla, CA (H.H.P.); Department of Anesthesiology, University of California, San Diego, La Jolla (H.H.P.); and VA Northern California Healthcare System, Mather (Y.K.X.)
| | - Julie Bossuyt
- From the Department of Pharmacology, University of California at Davis (F.B., B.X., G.R.R., T.W., Q.W., Q.F., M.L., Q.S., K.S.G., J.B., D.B., Y.K.X.); Department of Experimental Medicine (F.B., A.M.I.) and Department of Anatomical, Histological, Forensic, and Orthopedic Sciences (F.N.), Sapienza University of Rome, Italy; Department of Medicine and Epidemiology, School of Veterinary Medicine, and Surgical Research Facility, School of Medicine, University of California, Davis (W.F.); VA San Diego Healthcare System, La Jolla, CA (H.H.P.); Department of Anesthesiology, University of California, San Diego, La Jolla (H.H.P.); and VA Northern California Healthcare System, Mather (Y.K.X.)
| | - Donald Bers
- From the Department of Pharmacology, University of California at Davis (F.B., B.X., G.R.R., T.W., Q.W., Q.F., M.L., Q.S., K.S.G., J.B., D.B., Y.K.X.); Department of Experimental Medicine (F.B., A.M.I.) and Department of Anatomical, Histological, Forensic, and Orthopedic Sciences (F.N.), Sapienza University of Rome, Italy; Department of Medicine and Epidemiology, School of Veterinary Medicine, and Surgical Research Facility, School of Medicine, University of California, Davis (W.F.); VA San Diego Healthcare System, La Jolla, CA (H.H.P.); Department of Anesthesiology, University of California, San Diego, La Jolla (H.H.P.); and VA Northern California Healthcare System, Mather (Y.K.X.)
| | - Yang K Xiang
- From the Department of Pharmacology, University of California at Davis (F.B., B.X., G.R.R., T.W., Q.W., Q.F., M.L., Q.S., K.S.G., J.B., D.B., Y.K.X.); Department of Experimental Medicine (F.B., A.M.I.) and Department of Anatomical, Histological, Forensic, and Orthopedic Sciences (F.N.), Sapienza University of Rome, Italy; Department of Medicine and Epidemiology, School of Veterinary Medicine, and Surgical Research Facility, School of Medicine, University of California, Davis (W.F.); VA San Diego Healthcare System, La Jolla, CA (H.H.P.); Department of Anesthesiology, University of California, San Diego, La Jolla (H.H.P.); and VA Northern California Healthcare System, Mather (Y.K.X.).
| |
Collapse
|
38
|
Terentyev D, Hamilton S. Regulation of sarcoplasmic reticulum Ca 2+ release by serine-threonine phosphatases in the heart. J Mol Cell Cardiol 2016; 101:156-164. [PMID: 27585747 DOI: 10.1016/j.yjmcc.2016.08.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 08/26/2016] [Accepted: 08/27/2016] [Indexed: 12/17/2022]
Abstract
The amount and timing of Ca2+ release from the sarcoplasmic reticulum (SR) during cardiac cycle are the main determinants of cardiac contractility. Reversible phosphorylation of the SR Ca2+ release channel, ryanodine receptor type 2 (RyR2) is the central mechanism of regulation of Ca2+ release in cardiomyocytes. Three major serine-threonine phosphatases including PP1, PP2A and PP2B (calcineurin) have been implicated in modulation of RyR2 function. Changes in expression levels of these phosphatases, their activity and targeting to the RyR2 macromolecular complex were demonstrated in many animal models of cardiac disease and humans and are implicated in cardiac arrhythmia and heart failure. Here we review evidence in support of regulation of RyR2-mediated SR Ca2+ release by serine-threonine phosphatases and the role and mechanisms of dysregulation of phosphatases in various disease states.
Collapse
Affiliation(s)
- Dmitry Terentyev
- The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Department of Medicine, Cardiovascular Research Center, United States.
| | - Shanna Hamilton
- Cardiff University, School of Medicine, Wales Heart Research Institute, United Kingdom
| |
Collapse
|
39
|
Gloschat CR, Koppel AC, Aras KK, Brennan JA, Holzem KM, Efimov IR. Arrhythmogenic and metabolic remodelling of failing human heart. J Physiol 2016; 594:3963-80. [PMID: 27019074 DOI: 10.1113/jp271992] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 03/21/2016] [Indexed: 12/24/2022] Open
Abstract
Heart failure (HF) is a major cause of morbidity and mortality worldwide. The global burden of HF continues to rise, with prevalence rates estimated at 1-2% and incidence approaching 5-10 per 1000 persons annually. The complex pathophysiology of HF impacts virtually all aspects of normal cardiac function - from structure and mechanics to metabolism and electrophysiology - leading to impaired mechanical contraction and sudden cardiac death. Pharmacotherapy and device therapy are the primary methods of treating HF, but neither is able to stop or reverse disease progression. Thus, there is an acute need to translate basic research into improved HF therapy. Animal model investigations are a critical component of HF research. However, the translation from cellular and animal models to the bedside is hampered by significant differences between species and among physiological scales. Our studies over the last 8 years show that hypotheses generated in animal models need to be validated in human in vitro models. Importantly, however, human heart investigations can establish translational platforms for safety and efficacy studies before embarking on costly and risky clinical trials. This review summarizes recent developments in human HF investigations of electrophysiology remodelling, metabolic remodelling, and β-adrenergic remodelling and discusses promising new technologies for HF research.
Collapse
Affiliation(s)
- C R Gloschat
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
| | - A C Koppel
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
| | - K K Aras
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
| | - J A Brennan
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
| | - K M Holzem
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
| | - I R Efimov
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
| |
Collapse
|
40
|
Abstract
Stress-response kinases, the mitogen-activated protein kinases (MAPKs) are activated in response to the challenge of a myriad of stressors. c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinases (ERKs), and p38 MAPKs are the predominant members of the MAPK family in the heart. Extensive studies have revealed critical roles of activated MAPKs in the processes of cardiac injury and heart failure and many other cardiovascular diseases. Recently, emerging evidence suggests that MAPKs also promote the development of cardiac arrhythmias. Thus, understanding the functional impact of MAPKs in the heart could shed new light on the development of novel therapeutic approaches to improve cardiac function and prevent arrhythmia development in the patients. This review will summarize the recent findings on the role of MAPKs in cardiac remodeling and arrhythmia development and point to the critical need of future studies to further elucidate the fundamental mechanisms of MAPK activation and arrhythmia development in the heart.
Collapse
|
41
|
Gomez JF, Cardona K, Trenor B. Lessons learned from multi-scale modeling of the failing heart. J Mol Cell Cardiol 2015; 89:146-59. [PMID: 26476237 DOI: 10.1016/j.yjmcc.2015.10.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 10/07/2015] [Accepted: 10/14/2015] [Indexed: 12/26/2022]
Abstract
Heart failure constitutes a major public health problem worldwide. Affected patients experience a number of changes in the electrical function of the heart that predispose to potentially lethal cardiac arrhythmias. Due to the multitude of electrophysiological changes that may occur during heart failure, the scientific literature is complex and sometimes ambiguous, perhaps because these findings are highly dependent on the etiology, the stage of heart failure, and the experimental model used to study these changes. Nevertheless, a number of common features of failing hearts have been documented. Prolongation of the action potential (AP) involving ion channel remodeling and alterations in calcium handling have been established as the hallmark characteristics of myocytes isolated from failing hearts. Intercellular uncoupling and fibrosis are identified as major arrhythmogenic factors. Multi-scale computational simulations are a powerful tool that complements experimental and clinical research. The development of biophysically detailed computer models of single myocytes and cardiac tissues has contributed greatly to our understanding of processes underlying excitation and repolarization in the heart. The electrical, structural, and metabolic remodeling that arises in cardiac tissues during heart failure has been addressed from different computational perspectives to further understand the arrhythmogenic substrate. This review summarizes the contributions from computational modeling and simulation to predict the underlying mechanisms of heart failure phenotypes and their implications for arrhythmogenesis, ranging from the cellular level to whole-heart simulations. The main aspects of heart failure are presented in several related sections. An overview of the main electrophysiological and structural changes that have been observed experimentally in failing hearts is followed by the description and discussion of the simulation work in this field at the cellular level, and then in 2D and 3D cardiac structures. The implications for arrhythmogenesis in heart failure are also discussed including therapeutic measures, such as drug effects and cardiac resynchronization therapy. Finally, the future challenges in heart failure modeling and simulation will be discussed.
Collapse
Affiliation(s)
- Juan F Gomez
- Instituto de Investigación Interuniversitario en Bioingeniería y Tecnología Orientada, al Ser Humano (I3BH), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain.
| | - Karen Cardona
- Instituto de Investigación Interuniversitario en Bioingeniería y Tecnología Orientada, al Ser Humano (I3BH), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain.
| | - Beatriz Trenor
- Instituto de Investigación Interuniversitario en Bioingeniería y Tecnología Orientada, al Ser Humano (I3BH), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain.
| |
Collapse
|
42
|
Nguyen MN, Kiriazis H, Ruggiero D, Gao XM, Su Y, Jian A, Han LP, McMullen JR, Du XJ. Spontaneous ventricular tachyarrhythmias in β2-adrenoceptor transgenic mice in relation to cardiac interstitial fibrosis. Am J Physiol Heart Circ Physiol 2015; 309:H946-57. [PMID: 26116714 DOI: 10.1152/ajpheart.00405.2015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 06/26/2015] [Indexed: 12/21/2022]
Abstract
Myocardial fibrosis is regarded as a pivotal proarrhythmic substrate, but there have been no comprehensive studies showing a correlation between the severity of fibrosis and ventricular tachyarrhythmias (VTAs). Our purpose was to document this relationship in a transgenic (TG) strain of mice with fibrotic cardiomyopathy. TG mice with cardiac overexpression of β2-adrenoceptors (β2-AR mice) and non-TG (NTG) littermates were studied at 4-12 mo of age. VTA was quantified by ECG telemetry. The effect of pharmacological blockade of β2-ARs on VTA was examined. Myocardial collagen content was determined by hydroxyproline assay. NTG and TG mice displayed circadian variation in heart rate, which was higher in TG mice than in NTG mice (P <0.05). Frequent spontaneous ventricular ectopic beats (VEBs) and ventricular tachycardia (VT) were prominent in TG mice but not present in NTG mice. The frequency of VEB and VT episodes in TG mice increased with age (P < 0.01). Ventricular collagen content was greater in TG mice than in NTG mice (P <0.001) and correlated with age (r = 0.71, P < 0.01). The number of VEBs or VT episodes correlated with age (r = 0.83 and r = 0.73) and the content of total or cross-linked collagen (r = 0.62∼0.66, all P <0.01). While having no effect in younger β2-TG mice, β2-AR blockade reduced the frequency of VTA in old β2-TG mice with more severe fibrosis. In conclusion, β2-TG mice exhibit interstitial fibrosis and spontaneous onset of VTA, becoming more severe with aging. The extent of cardiac fibrosis is a major determinant for both the frequency of VTA and proarrhythmic action of β2-AR activation.
Collapse
Affiliation(s)
- My-Nhan Nguyen
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Helen Kiriazis
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Diego Ruggiero
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; University of Milan, Milan, Italy
| | - Xiao-Ming Gao
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Yidan Su
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Anne Jian
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Li-Ping Han
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; WenZhou Medical University, WenZhou, China; and
| | - Julie R McMullen
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Physiology, Monash University, Melbourne, Australia
| | - Xiao-Jun Du
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; Central Clinical School, Monash University, Melbourne, Victoria, Australia;
| |
Collapse
|
43
|
|
44
|
Xie L, Pi X, Townley-Tilson WHD, Li N, Wehrens XHT, Entman ML, Taffet GE, Mishra A, Peng J, Schisler JC, Meissner G, Patterson C. PHD2/3-dependent hydroxylation tunes cardiac response to β-adrenergic stress via phospholamban. J Clin Invest 2015; 125:2759-71. [PMID: 26075818 DOI: 10.1172/jci80369] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 05/06/2015] [Indexed: 01/08/2023] Open
Abstract
Ischemic heart disease is the leading cause of heart failure. Both clinical trials and experimental animal studies demonstrate that chronic hypoxia can induce contractile dysfunction even before substantial ventricular damage, implicating a direct role of oxygen in the regulation of cardiac contractile function. Prolyl hydroxylase domain (PHD) proteins are well recognized as oxygen sensors and mediate a wide variety of cellular events by hydroxylating a growing list of protein substrates. Both PHD2 and PHD3 are highly expressed in the heart, yet their functional roles in modulating contractile function remain incompletely understood. Here, we report that combined deletion of Phd2 and Phd3 dramatically decreased expression of phospholamban (PLN), resulted in sustained activation of calcium/calmodulin-activated kinase II (CaMKII), and sensitized mice to chronic β-adrenergic stress-induced myocardial injury. We have provided evidence that thyroid hormone receptor-α (TR-α), a transcriptional regulator of PLN, interacts with PHD2 and PHD3 and is hydroxylated at 2 proline residues. Inhibition of PHDs increased the interaction between TR-α and nuclear receptor corepressor 2 (NCOR2) and suppressed Pln transcription. Together, these observations provide mechanistic insight into how oxygen directly modulates cardiac contractility and suggest that cardiac function could be modulated therapeutically by tuning PHD enzymatic activity.
Collapse
|
45
|
Lang D, Holzem K, Kang C, Xiao M, Hwang HJ, Ewald GA, Yamada KA, Efimov IR. Arrhythmogenic remodeling of β2 versus β1 adrenergic signaling in the human failing heart. Circ Arrhythm Electrophysiol 2015; 8:409-19. [PMID: 25673629 DOI: 10.1161/circep.114.002065] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 01/27/2015] [Indexed: 01/10/2023]
Abstract
BACKGROUND Arrhythmia is the major cause of death in patients with heart failure, for which β-adrenergic receptor blockers are a mainstay therapy. But the role of β-adrenergic signaling in electrophysiology and arrhythmias has never been studied in human ventricles. METHODS AND RESULTS We used optical imaging of action potentials and [Ca(2+)]i transients to compare the β1- and β2-adrenergic responses in left ventricular wedge preparations of human donor and failing hearts. β1-Stimulation significantly increased conduction velocity, shortened action potential duration, and [Ca(2+)]i transients duration (CaD) in donor but not in failing hearts, because of desensitization of β1-adrenergic receptor in heart failure. In contrast, β2-stimulation increased conduction velocity in both donor and failing hearts but shortened action potential duration only in failing hearts. β2-Stimulation also affected transmural heterogeneity in action potential duration but not in [Ca(2+)]i transients duration. Both β1- and β2-stimulation augmented the vulnerability and frequency of ectopic activity and enhanced substrates for ventricular tachycardia in failing, but not in donor, hearts. Both β1- and β2-stimulation enhanced Purkinje fiber automaticity, whereas only β2-stimulation promoted Ca-mediated premature ventricular contractions in heart failure. CONCLUSIONS During end-stage heart failure, β2-stimulation creates arrhythmogenic substrates via conduction velocity regulation and transmurally heterogeneous repolarization. β2-Stimulation is, therefore, more arrhythmogenic than β1-stimulation. In particular, β2-stimulation increases the transmural difference between [Ca(2+)]i transients duration and action potential duration, which facilitates the formation of delayed afterdepolarizations.
Collapse
Affiliation(s)
- Di Lang
- From the Department of Biomedical Engineering (D.L., K.H., C.K., M.X., H.J.H., I.R.E.) and Department of Medicine (G.A.E., K.A.Y., I.R.E.), Washington University School of Medicine, St. Louis, MO; L'Institut de Rythmologie et Modélisation Cardiaque LIRYC, Université de Bordeaux, Bordeaux, France (I.R.E.); and Moscow Institute of Physics and Technology, Moscow, Russia (I.R.E.)
| | - Katherine Holzem
- From the Department of Biomedical Engineering (D.L., K.H., C.K., M.X., H.J.H., I.R.E.) and Department of Medicine (G.A.E., K.A.Y., I.R.E.), Washington University School of Medicine, St. Louis, MO; L'Institut de Rythmologie et Modélisation Cardiaque LIRYC, Université de Bordeaux, Bordeaux, France (I.R.E.); and Moscow Institute of Physics and Technology, Moscow, Russia (I.R.E.)
| | - Chaoyi Kang
- From the Department of Biomedical Engineering (D.L., K.H., C.K., M.X., H.J.H., I.R.E.) and Department of Medicine (G.A.E., K.A.Y., I.R.E.), Washington University School of Medicine, St. Louis, MO; L'Institut de Rythmologie et Modélisation Cardiaque LIRYC, Université de Bordeaux, Bordeaux, France (I.R.E.); and Moscow Institute of Physics and Technology, Moscow, Russia (I.R.E.)
| | - Mengqian Xiao
- From the Department of Biomedical Engineering (D.L., K.H., C.K., M.X., H.J.H., I.R.E.) and Department of Medicine (G.A.E., K.A.Y., I.R.E.), Washington University School of Medicine, St. Louis, MO; L'Institut de Rythmologie et Modélisation Cardiaque LIRYC, Université de Bordeaux, Bordeaux, France (I.R.E.); and Moscow Institute of Physics and Technology, Moscow, Russia (I.R.E.)
| | - Hye Jin Hwang
- From the Department of Biomedical Engineering (D.L., K.H., C.K., M.X., H.J.H., I.R.E.) and Department of Medicine (G.A.E., K.A.Y., I.R.E.), Washington University School of Medicine, St. Louis, MO; L'Institut de Rythmologie et Modélisation Cardiaque LIRYC, Université de Bordeaux, Bordeaux, France (I.R.E.); and Moscow Institute of Physics and Technology, Moscow, Russia (I.R.E.)
| | - Gregory A Ewald
- From the Department of Biomedical Engineering (D.L., K.H., C.K., M.X., H.J.H., I.R.E.) and Department of Medicine (G.A.E., K.A.Y., I.R.E.), Washington University School of Medicine, St. Louis, MO; L'Institut de Rythmologie et Modélisation Cardiaque LIRYC, Université de Bordeaux, Bordeaux, France (I.R.E.); and Moscow Institute of Physics and Technology, Moscow, Russia (I.R.E.)
| | - Kathryn A Yamada
- From the Department of Biomedical Engineering (D.L., K.H., C.K., M.X., H.J.H., I.R.E.) and Department of Medicine (G.A.E., K.A.Y., I.R.E.), Washington University School of Medicine, St. Louis, MO; L'Institut de Rythmologie et Modélisation Cardiaque LIRYC, Université de Bordeaux, Bordeaux, France (I.R.E.); and Moscow Institute of Physics and Technology, Moscow, Russia (I.R.E.)
| | - Igor R Efimov
- From the Department of Biomedical Engineering (D.L., K.H., C.K., M.X., H.J.H., I.R.E.) and Department of Medicine (G.A.E., K.A.Y., I.R.E.), Washington University School of Medicine, St. Louis, MO; L'Institut de Rythmologie et Modélisation Cardiaque LIRYC, Université de Bordeaux, Bordeaux, France (I.R.E.); and Moscow Institute of Physics and Technology, Moscow, Russia (I.R.E.).
| |
Collapse
|
46
|
Bao L, Lu F, Chen H, Min Q, Chen X, Song Y, Zhao B, Bu H, Sun H. High concentration of epigallocatechin-3-gallate increased the incidences of arrhythmia and diastolic dysfunction via β2-adrenoceptor. J Food Sci 2015; 80:T659-63. [PMID: 25676191 DOI: 10.1111/1750-3841.12803] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 12/18/2014] [Indexed: 11/28/2022]
Abstract
Epigallocatechin-3-gallate (EGCG) is the major and most potent representative in green tea, which has been proved to modulate myocardial contractility. Whether EGCG has some negative effects on cardiac function is not known. In the present study, we investigated the effects of EGCG at different doses on cardiac contraction and explored whether β2 -adrenoceptor (β2 AR) was involved in EGCG-induced cardiac effects. Isolated rat hearts were mounted on the Langendorff system and perfused with different concentrations of EGCG in low or normal calcium Krebs-Henseleit (KH) buffer. The contraction of hearts was measured. Ventricular myocytes were cultured with EGCG and isoprenaline (ISO, 10(-7) M) for 12 h. ICI118,551 (55 nM) was used to inhibit β2 AR. Cardiomyocyte shortening, viability, and responsiveness to ISO (10(-9) M) were measured. EGCG dose dependently enhanced contractility of perfused heart in low calcium KH buffer. In the normal calcium KH buffer, EGCG at low dose (20 μM) increased heart contraction, while at high dose (50 μM), it increased the incidences of arrhythmia and diastolic dysfunction. In isolated ventricular myocytes, EGCG at the concentration of 0.001 to 1.0 μΜ did not affect their contraction. However, the responsiveness to ISO and the survival of myocytes were increased by EGCG (0.01 μM). The increased responsiveness was partially abolished by ICI118,551. The data obtained in this study demonstrated that EGCG at low dose conferred cardioprotection, yet at high dose increased the incidences of arrhythmia and diastolic dysfunction. β2 AR was involved in EGCG-induced cardiac effects.
Collapse
Affiliation(s)
- Lei Bao
- Dept. of Clinical Medicine, Xuzhou Medical College, Xuzhou, 221004, Jiangsu, China
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Woo AYH, Song Y, Xiao RP, Zhu W. Biased β2-adrenoceptor signalling in heart failure: pathophysiology and drug discovery. Br J Pharmacol 2014; 172:5444-56. [PMID: 25298054 DOI: 10.1111/bph.12965] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 08/27/2014] [Accepted: 09/28/2014] [Indexed: 12/27/2022] Open
Abstract
The body is constantly faced with a dynamic requirement for blood flow. The heart is able to respond to these changing needs by adjusting cardiac output based on cues emitted by circulating catecholamine levels. Cardiac β-adrenoceptors transduce the signal produced by catecholamine stimulation via Gs proteins to their downstream effectors to increase heart contractility. During heart failure, cardiac output is insufficient to meet the needs of the body; catecholamine levels are high and β-adrenoceptors become hyperstimulated. The hyperstimulated β1-adrenoceptors induce a cardiotoxic effect, which could be counteracted by the cardioprotective effect of β2-adrenoceptor-mediated Gi signalling. However, β2-adrenoceptor-Gi signalling negates the stimulatory effect of the Gs signalling on cardiomyocyte contraction and further exacerbates cardiodepression. Here, further to the localization of β1- and β2-adrenoceptors and β2-adrenoceptor-mediated β-arrestin signalling in cardiomyocytes, we discuss features of the dysregulation of β-adrenoceptor subtype signalling in the failing heart, and conclude that Gi-biased β2-adrenoceptor signalling is a pathogenic pathway in heart failure that plays a crucial role in cardiac remodelling. In contrast, β2-adrenoceptor-Gs signalling increases cardiomyocyte contractility without causing cardiotoxicity. Finally, we discuss a novel therapeutic approach for heart failure using a Gs-biased β2-adrenoceptor agonist and a β1-adrenoceptor antagonist in combination. This combination treatment normalizes the β-adrenoceptor subtype signalling in the failing heart and produces therapeutic effects that outperform traditional heart failure therapies in animal models. The present review illustrates how the concept of biased signalling can be applied to increase our understanding of the pathophysiology of diseases and in the development of novel therapies.
Collapse
Affiliation(s)
- Anthony Yiu-Ho Woo
- Institute of Molecular Medicine, Centre for Life Sciences, Peking University, Beijing, China.,Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Ying Song
- Institute of Molecular Medicine, Centre for Life Sciences, Peking University, Beijing, China
| | - Rui-Ping Xiao
- Institute of Molecular Medicine, Centre for Life Sciences, Peking University, Beijing, China.,Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Weizhong Zhu
- Department of Pharmacology, Nantong University School of Pharmacy, Nantong, China
| |
Collapse
|
48
|
Wengrowski AM, Wang X, Tapa S, Posnack NG, Mendelowitz D, Kay MW. Optogenetic release of norepinephrine from cardiac sympathetic neurons alters mechanical and electrical function. Cardiovasc Res 2014; 105:143-50. [PMID: 25514932 DOI: 10.1093/cvr/cvu258] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIMS Release of norepinephrine (NE) from sympathetic neurons enhances heart rate (HR) and developed force through activation of β-adrenergic receptors, and this sympathoexcitation is a key risk for the generation of cardiac arrhythmias. Studies of β-adrenergic modulation of cardiac function typically involve the administration of exogenous β-adrenergic receptor agonists to directly elicit global β-adrenergic receptor activation by bypassing the involvement of sympathetic nerve terminals. In this work, we use a novel method to activate sympathetic fibres within the myocardium of Langendorff-perfused hearts while measuring changes in electrical and mechanical function. METHODS AND RESULTS The light-activated optogenetic protein channelrhodopsin-2 (ChR2) was expressed in murine catecholaminergic sympathetic neurons. Sympathetic fibres were then photoactivated to examine changes in contractile force, HR, and cardiac electrical activity. Incidence of arrhythmia was measured with and without exposure to photoactivation of sympathetic fibres, and hearts were optically mapped to detect changes in action potential durations and conduction velocities. Results demonstrate facilitation of both developed force and HR after photostimulated release of NE, with increases in contractile force and HR of 34.5 ± 5.5 and 25.0 ± 9.3%, respectively. Photostimulation of sympathetic fibres also made hearts more susceptible to arrhythmia, with greater incidence and severity. In addition, optically mapped action potentials displayed a small but significant shortening of the plateau phase (-5.5 ± 1.0 ms) after photostimulation. CONCLUSION This study characterizes a powerful and clinically relevant new model for studies of cardiac arrhythmias generated by increasing the activity of sympathetic nerve terminals and the resulting activation of myocyte β-adrenergic receptors.
Collapse
Affiliation(s)
- Anastasia M Wengrowski
- Department of Biomedical Engineering, The George Washington University, Phillips Hall, Room 607, 801 22nd Street NW, Washington, DC 20052, USA
| | - Xin Wang
- Department of Pharmacology and Physiology, The George Washington University, Washington, DC, USA
| | - Srinivas Tapa
- Department of Biomedical Engineering, The George Washington University, Phillips Hall, Room 607, 801 22nd Street NW, Washington, DC 20052, USA
| | - Nikki Gillum Posnack
- Department of Pharmacology and Physiology, The George Washington University, Washington, DC, USA
| | - David Mendelowitz
- Department of Pharmacology and Physiology, The George Washington University, Washington, DC, USA
| | - Matthew W Kay
- Department of Biomedical Engineering, The George Washington University, Phillips Hall, Room 607, 801 22nd Street NW, Washington, DC 20052, USA Department of Pharmacology and Physiology, The George Washington University, Washington, DC, USA
| |
Collapse
|
49
|
Bodzenta-Łukaszyk A, Kokot M. Pharmacological consequences of inhaled drug delivery to small airways in the treatment of asthma. Adv Ther 2014; 31:803-16. [PMID: 25116888 DOI: 10.1007/s12325-014-0143-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Indexed: 11/29/2022]
Abstract
Small peripheral airways are an important target for the anti-inflammatory treatment of asthma. To make anti-inflammatory drugs (inhaled corticosteroids [ICS]) effectively reach small airways, they should be delivered using inhalation techniques containing high proportions of fine or super-fine particles. Higher proportions of fine particles are associated with higher systemic absorption of ICS leading to an increased risk of endogenous cortisol suppression. Ciclesonide, despite the highest proportion of fine and super-fine particle fractions, is the only ICS not associated with an increased risk of systemic adverse effects, including cortisol suppression. In contrary to ICS, bronchodilators should not be administered to peripheral airways. This does not improve their efficacy and may increase their risk of cardiotoxicity. Thus, from a pharmacological point of view and the theory of aerosols' deposition, fixed combinations of ICS and long-acting beta agonists are always suboptimal. In many cases, the best solution may be to use fine-particle ciclesonide and a non-fine particle beta agonist administered from separate inhalers.
Collapse
Affiliation(s)
- Anna Bodzenta-Łukaszyk
- Clinical Department of Allergic and Internal Diseases, Medical University of Białystok, Bialystok, Poland,
| | | |
Collapse
|
50
|
Liu T, Takimoto E, Dimaano VL, DeMazumder D, Kettlewell S, Smith G, Sidor A, Abraham TP, O'Rourke B. Inhibiting mitochondrial Na+/Ca2+ exchange prevents sudden death in a Guinea pig model of heart failure. Circ Res 2014; 115:44-54. [PMID: 24780171 DOI: 10.1161/circresaha.115.303062] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
RATIONALE In cardiomyocytes from failing hearts, insufficient mitochondrial Ca(2+) accumulation secondary to cytoplasmic Na(+) overload decreases NAD(P)H/NAD(P)(+) redox potential and increases oxidative stress when workload increases. These effects are abolished by enhancing mitochondrial Ca(2+) with acute treatment with CGP-37157 (CGP), an inhibitor of the mitochondrial Na(+)/Ca(2+) exchanger. OBJECTIVE Our aim was to determine whether chronic CGP treatment mitigates contractile dysfunction and arrhythmias in an animal model of heart failure (HF) and sudden cardiac death (SCD). METHODS AND RESULTS Here, we describe a novel guinea pig HF/SCD model using aortic constriction combined with daily β-adrenergic receptor stimulation (ACi) and show that chronic CGP treatment (ACi plus CGP) attenuates cardiac hypertrophic remodeling, pulmonary edema, and interstitial fibrosis and prevents cardiac dysfunction and SCD. In the ACi group 4 weeks after pressure overload, fractional shortening and the rate of left ventricular pressure development decreased by 36% and 32%, respectively, compared with sham-operated controls; in contrast, cardiac function was completely preserved in the ACi plus CGP group. CGP treatment also significantly reduced the incidence of premature ventricular beats and prevented fatal episodes of ventricular fibrillation, but did not prevent QT prolongation. Without CGP treatment, mortality was 61% in the ACi group <4 weeks of aortic constriction, whereas the death rate in the ACi plus CGP group was not different from sham-operated animals. CONCLUSIONS The findings demonstrate the critical role played by altered mitochondrial Ca(2+) dynamics in the development of HF and HF-associated SCD; moreover, they reveal a novel strategy for treating SCD and cardiac decompensation in HF.
Collapse
Affiliation(s)
- Ting Liu
- From the Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (T.L., E.T., V.L.D., D.D., A.S., T.P.A., B.O.'R.); and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (S.K., G.S.)
| | - Eiki Takimoto
- From the Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (T.L., E.T., V.L.D., D.D., A.S., T.P.A., B.O.'R.); and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (S.K., G.S.)
| | - Veronica L Dimaano
- From the Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (T.L., E.T., V.L.D., D.D., A.S., T.P.A., B.O.'R.); and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (S.K., G.S.)
| | - Deeptankar DeMazumder
- From the Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (T.L., E.T., V.L.D., D.D., A.S., T.P.A., B.O.'R.); and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (S.K., G.S.)
| | - Sarah Kettlewell
- From the Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (T.L., E.T., V.L.D., D.D., A.S., T.P.A., B.O.'R.); and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (S.K., G.S.)
| | - Godfrey Smith
- From the Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (T.L., E.T., V.L.D., D.D., A.S., T.P.A., B.O.'R.); and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (S.K., G.S.)
| | - Agnieszka Sidor
- From the Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (T.L., E.T., V.L.D., D.D., A.S., T.P.A., B.O.'R.); and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (S.K., G.S.)
| | - Theodore P Abraham
- From the Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (T.L., E.T., V.L.D., D.D., A.S., T.P.A., B.O.'R.); and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (S.K., G.S.)
| | - Brian O'Rourke
- From the Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (T.L., E.T., V.L.D., D.D., A.S., T.P.A., B.O.'R.); and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (S.K., G.S.).
| |
Collapse
|