1
|
Quan Y, Ding S, Wang Y, Chen X, Zhou B, Zhou Y. Real-time cardiomyocyte contraction sensing via a neo-flexible magnetic sensor. Biosens Bioelectron 2025; 277:117294. [PMID: 40014947 DOI: 10.1016/j.bios.2025.117294] [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: 12/17/2024] [Revised: 02/10/2025] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
Assessing heart disease and evaluating drug-induced cardiotoxicity require a deep understanding of the contractile properties of cardiac tissue, particularly at the level of individual cardiomyocytes. Traditional methods for measuring cardiomyocyte contractility present several challenges, including limitations in real-time detection, complex and costly sensing platforms, and biocompatibility issues. To address these challenges, we introduce an innovative magnetic sensor that utilizes a flexible coil cantilever, pioneering the application of electromagnetic induction for detecting cardiomyocyte contractility. This marks the first time such technology has been deployed in typical laboratory settings with straightforward configurations. When cardiomyocytes are cultured on these coils and subjected to a static magnetic field, their contractions induce oscillations in the coils, generating an electromotive force that converts mechanical pulsations into electrical signals. This advanced platform enables long-term, real-time monitoring of cardiac functional characteristics, including contractility, beating rate, and rhythm. It also enables the quantitative assessment of cardiovascular dynamics, such as in response to drugs like isoproterenol and verapamil. Offering a uniquely simple, stable, and efficient method for evaluating drug-induced cardiotoxicity, this novel detection platform underscores the transformative potential of flexible magnetic sensors in real-time cellular detection applications.
Collapse
Affiliation(s)
- Yue Quan
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau
| | - Sen Ding
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau
| | - Yuxin Wang
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau
| | - Bingpu Zhou
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau.
| | - Yinning Zhou
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau.
| |
Collapse
|
2
|
Wright PT, Gorelik J, Harding SE. Electrophysiological Remodeling: Cardiac T-Tubules and ß-Adrenoceptors. Cells 2021; 10:cells10092456. [PMID: 34572106 PMCID: PMC8468945 DOI: 10.3390/cells10092456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 01/09/2023] Open
Abstract
Beta-adrenoceptors (βAR) are often viewed as archetypal G-protein coupled receptors. Over the past fifteen years, investigations in cardiovascular biology have provided remarkable insights into this receptor family. These studies have shifted pharmacological dogma, from one which centralized the receptor to a new focus on structural micro-domains such as caveolae and t-tubules. Important studies have examined, separately, the structural compartmentation of ion channels and βAR. Despite links being assumed, relatively few studies have specifically examined the direct link between structural remodeling and electrical remodeling with a focus on βAR. In this review, we will examine the nature of receptor and ion channel dysfunction on a substrate of cardiomyocyte microdomain remodeling, as well as the likely ramifications for cardiac electrophysiology. We will then discuss the advances in methodologies in this area with a specific focus on super-resolution microscopy, fluorescent imaging, and new approaches involving microdomain specific, polymer-based agonists. The advent of powerful computational modelling approaches has allowed the science to shift from purely empirical work, and may allow future investigations based on prediction. Issues such as the cross-reactivity of receptors and cellular heterogeneity will also be discussed. Finally, we will speculate as to the potential developments within this field over the next ten years.
Collapse
Affiliation(s)
- Peter T. Wright
- School of Life & Health Sciences, University of Roehampton, Holybourne Avenue, London SW15 4JD, UK;
- Cardiac Section, National Heart and Lung Institute (NHLI), Faculty of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK;
| | - Julia Gorelik
- Cardiac Section, National Heart and Lung Institute (NHLI), Faculty of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK;
| | - Sian E. Harding
- Cardiac Section, National Heart and Lung Institute (NHLI), Faculty of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK;
- Correspondence:
| |
Collapse
|
3
|
Perez DM. Targeting Adrenergic Receptors in Metabolic Therapies for Heart Failure. Int J Mol Sci 2021; 22:5783. [PMID: 34071350 PMCID: PMC8198887 DOI: 10.3390/ijms22115783] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/20/2021] [Accepted: 05/22/2021] [Indexed: 12/14/2022] Open
Abstract
The heart has a reduced capacity to generate sufficient energy when failing, resulting in an energy-starved condition with diminished functions. Studies have identified numerous changes in metabolic pathways in the failing heart that result in reduced oxidation of both glucose and fatty acid substrates, defects in mitochondrial functions and oxidative phosphorylation, and inefficient substrate utilization for the ATP that is produced. Recent early-phase clinical studies indicate that inhibitors of fatty acid oxidation and antioxidants that target the mitochondria may improve heart function during failure by increasing compensatory glucose oxidation. Adrenergic receptors (α1 and β) are a key sympathetic nervous system regulator that controls cardiac function. β-AR blockers are an established treatment for heart failure and α1A-AR agonists have potential therapeutic benefit. Besides regulating inotropy and chronotropy, α1- and β-adrenergic receptors also regulate metabolic functions in the heart that underlie many cardiac benefits. This review will highlight recent studies that describe how adrenergic receptor-mediated metabolic pathways may be able to restore cardiac energetics to non-failing levels that may offer promising therapeutic strategies.
Collapse
Affiliation(s)
- Dianne M Perez
- The Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH 44195, USA
| |
Collapse
|
4
|
Hu D, Huang X, Zheng C, Zhu Y, Chen L, Lin H, Liao Y. [Contribution of sympathetic activation to antihypertrophic memory after regression of exercise-induced physiological myocardial hypertrophy in mice]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:495-503. [PMID: 33963707 DOI: 10.12122/j.issn.1673-4254.2021.04.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate whether anti-hypertrophic memory exists after regression of exercise-induced physiological myocardial hypertrophy (PMH) and explore the contribution of sympathetic activation to hypertrophic memory formation. OBJECTIVE Seventy-two mice were randomized equally into 6 groups, including sedentary sham-operated group, exercise hypertrophic preconditioning (EHP) + sham operation group, bisoprolol (an adrenergic β1 receptor blocker) + EHP + sham operation group (biso+Exe+Sham group), sedentary group with transverse aortic constriction (TAC) (Sed+TAC group), EHP+ TAC group (Exe+TAC group), and bisoprolol+EHP+TAC group (biso+Exe+TAC group). The mice in the EHP groups were subjected to 3 weeks of swimming training, and in the bisoprolol groups, bisoprolol was administered by gavage once daily from two days before till the end of the training. One week after the training, TAC or sham surgery was performed. Echocardiography and hemodynamic measurements were performed to evaluate cardiac function of the mice, and the myocardial tissues were examined histologically to detect cardiac remodeling. OBJECTIVE Compared with the sedentary group, the mice receiving 3 weeks of swimming training had significantly increased heart weight to body weight ratio (HW/BW), HW to tibia length ratio (HW/TL), and the cross-sectional area of the cardiomyocytes (P < 0.05). One week after the training, exercise-induced PMH rapidly diminished and both HW/BW and HW/TL recovered the baseline levels. Treatment with bisoprolol obviously prevented the occurrence of PMH. Four weeks after TAC, the left ventricular posterior wall thickness, HW/BW, HW/TL, left ventricular end diastolic pressure and cross-sectional area of cardiomyocytes were all significantly lower (P < 0.05) while the left ejection fraction and maximal change rate of left ventricular pressure were significantly higher (P < 0.05) in Exe + TAC group than in Sed + TAC group. No significant difference was found in these parameters between biso + Exe + TAC group and Sed + TAC group. OBJECTIVE Anti-hypertrophic memory exists even after the regression of exercise-induced PMH, which may be attributed to the activation of sympathetic nervous system during exercise.
Collapse
Affiliation(s)
- D Hu
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - X Huang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - C Zheng
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Y Zhu
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - L Chen
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - H Lin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Y Liao
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
5
|
Zhao W, Chen Y, Yang W, Han Y, Wang Z, Huang F, Qiu Z, Yang K, Jin W. Effects of Cardiomyocyte-Specific Deletion of STAT3-A Murine Model of Heart Failure With Preserved Ejection Fraction. Front Cardiovasc Med 2020; 7:613123. [PMID: 33365331 PMCID: PMC7750364 DOI: 10.3389/fcvm.2020.613123] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/17/2020] [Indexed: 11/13/2022] Open
Abstract
Aims: There is a high incidence of heart failure with preserved ejection fraction (HFpEF), but the options of treatment are limited. A new animal model of HFpEF is urgently needed for in-depth research on HFpEF. Signal transducer and activator of transcription 3 (STAT3) may affect the passive stiffness of myocardium, which determines cardiac diastolic function. We hypothesized that cardiomyocyte-specific deletion of STAT3 increases cardiac passive stiffness, which results the murine features of HFpEF. Methods and Results: Cardiomyocyte-specific deletion of STAT3 (STAT3cKO) mice was generated by the Cre/FLOXp method. The STAT3cKO mice showed heavier cardiac fibrosis and cardiac hypertrophy comparing with wild-type (WT) mice. Furthermore, STAT3cKO mice showed increased serum brain natriuretic peptide (BNP) level, and growth stimulation expressed gene 2 (ST2) level. Other indicators reflecting cardiac passive stiffness and diastolic function, including end diastolic pressure volume relation, MV A value, MV E value, E/A and E/E' had different fold changes. All these changes were accompanied by decreasing levels of protein kinase G (PKG). Bioinformatic analysis of STAT3cKO mice hearts suggested cGMP-PKG signaling pathway might participate in the pathogenesis of HFpEF by means of adjusting different biological functions. Conclusions: Cardiomyocyte-specific deletion of STAT3 results in a murine HFpEF model which imitates the clinical characteristics partly by affecting cardiac PKG levels. Better understanding of the factors influencing HFpEF may finally provided innovative therapies.
Collapse
Affiliation(s)
- Weilin Zhao
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanjia Chen
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenbo Yang
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanxin Han
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyan Wang
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fanyi Huang
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zeping Qiu
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ke Yang
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Jin
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Mayourian J, Ceholski DK, Gonzalez DM, Cashman TJ, Sahoo S, Hajjar RJ, Costa KD. Physiologic, Pathologic, and Therapeutic Paracrine Modulation of Cardiac Excitation-Contraction Coupling. Circ Res 2019; 122:167-183. [PMID: 29301848 DOI: 10.1161/circresaha.117.311589] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cardiac excitation-contraction coupling (ECC) is the orchestrated process of initial myocyte electrical excitation, which leads to calcium entry, intracellular trafficking, and subsequent sarcomere shortening and myofibrillar contraction. Neurohumoral β-adrenergic signaling is a well-established mediator of ECC; other signaling mechanisms, such as paracrine signaling, have also demonstrated significant impact on ECC but are less well understood. For example, resident heart endothelial cells are well-known physiological paracrine modulators of cardiac myocyte ECC mainly via NO and endothelin-1. Moreover, recent studies have demonstrated other resident noncardiomyocyte heart cells (eg, physiological fibroblasts and pathological myofibroblasts), and even experimental cardiotherapeutic cells (eg, mesenchymal stem cells) are also capable of altering cardiomyocyte ECC through paracrine mechanisms. In this review, we first focus on the paracrine-mediated effects of resident and therapeutic noncardiomyocytes on cardiomyocyte hypertrophy, electrophysiology, and calcium handling, each of which can modulate ECC, and then discuss the current knowledge about key paracrine factors and their underlying mechanisms of action. Next, we provide a case example demonstrating the promise of tissue-engineering approaches to study paracrine effects on tissue-level contractility. More specifically, we present new functional and molecular data on the effects of human adult cardiac fibroblast conditioned media on human engineered cardiac tissue contractility and ion channel gene expression that generally agrees with previous murine studies but also suggests possible species-specific differences. By contrast, paracrine secretions by human dermal fibroblasts had no discernible effect on human engineered cardiac tissue contractile function and gene expression. Finally, we discuss systems biology approaches to help identify key stem cell paracrine mediators of ECC and their associated mechanistic pathways. Such integration of tissue-engineering and systems biology methods shows promise to reveal novel insights into paracrine mediators of ECC and their underlying mechanisms of action, ultimately leading to improved cell-based therapies for patients with heart disease.
Collapse
Affiliation(s)
- Joshua Mayourian
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Delaine K Ceholski
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - David M Gonzalez
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Timothy J Cashman
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Susmita Sahoo
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Roger J Hajjar
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Kevin D Costa
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
7
|
Mangmool S, Parichatikanond W, Kurose H. Therapeutic Targets for Treatment of Heart Failure: Focus on GRKs and β-Arrestins Affecting βAR Signaling. Front Pharmacol 2018; 9:1336. [PMID: 30538631 PMCID: PMC6277550 DOI: 10.3389/fphar.2018.01336] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 10/30/2018] [Indexed: 12/19/2022] Open
Abstract
Heart failure (HF) is a heart disease that is classified into two main types: HF with reduced ejection fraction (HFrEF) and HF with preserved ejection fraction (HFpEF). Both types of HF lead to significant risk of mortality and morbidity. Pharmacological treatment with β-adrenergic receptor (βAR) antagonists (also called β-blockers) has been shown to reduce the overall hospitalization and mortality rates and improve the clinical outcomes in HF patients with HFrEF but not HFpEF. Although, the survival rate of patients suffering from HF continues to drop, the management of HF still faces several limitations and discrepancies highlighting the need to develop new treatment strategies. Overstimulation of the sympathetic nervous system is an adaptive neurohormonal response to acute myocardial injury and heart damage, whereas prolonged exposure to catecholamines causes defects in βAR regulation, including a reduction in the amount of βARs and an increase in βAR desensitization due to the upregulation of G protein-coupled receptor kinases (GRKs) in the heart, contributing in turn to the progression of HF. Several studies show that myocardial GRK2 activity and expression are raised in the failing heart. Furthermore, β-arrestins play a pivotal role in βAR desensitization and, interestingly, can mediate their own signal transduction without any G protein-dependent pathway involved. In this review, we provide new insight into the role of GRKs and β-arrestins on how they affect βAR signaling regarding the molecular and cellular pathophysiology of HF. Additionally, we discuss the therapeutic potential of targeting GRKs and β-arrestins for the treatment of HF.
Collapse
Affiliation(s)
- Supachoke Mangmool
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | | | - Hitoshi Kurose
- Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
8
|
Crocini C, Coppini R, Ferrantini C, Yan P, Loew LM, Poggesi C, Cerbai E, Pavone FS, Sacconi L. T-Tubular Electrical Defects Contribute to Blunted β-Adrenergic Response in Heart Failure. Int J Mol Sci 2016; 17:ijms17091471. [PMID: 27598150 PMCID: PMC5037749 DOI: 10.3390/ijms17091471] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 08/17/2016] [Accepted: 08/30/2016] [Indexed: 11/18/2022] Open
Abstract
Alterations of the β-adrenergic signalling, structural remodelling, and electrical failure of T-tubules are hallmarks of heart failure (HF). Here, we assess the effect of β-adrenoceptor activation on local Ca2+ release in electrically coupled and uncoupled T-tubules in ventricular myocytes from HF rats. We employ an ultrafast random access multi-photon (RAMP) microscope to simultaneously record action potentials and Ca2+ transients from multiple T-tubules in ventricular cardiomyocytes from a HF rat model of coronary ligation compared to sham-operated rats as a control. We confirmed that β-adrenergic stimulation increases the frequency of Ca2+ sparks, reduces Ca2+ transient variability, and hastens the decay of Ca2+ transients: all these effects are similarly exerted by β-adrenergic stimulation in control and HF cardiomyocytes. Conversely, β-adrenergic stimulation in HF cells accelerates a Ca2+ rise exclusively in the proximity of T-tubules that regularly conduct the action potential. The delayed Ca2+ rise found at T-tubules that fail to conduct the action potential is instead not affected by β-adrenergic signalling. Taken together, these findings indicate that HF cells globally respond to β-adrenergic stimulation, except at T-tubules that fail to conduct action potentials, where the blunted effect of the β-adrenergic signalling may be directly caused by the lack of electrical activity.
Collapse
Affiliation(s)
- Claudia Crocini
- European Laboratory for Non-Linear Spectroscopy, Florence 50019, Italy.
- National Institute of Optics, National Research Council, Florence 50125, Italy.
| | - Raffaele Coppini
- Division of Pharmacology, Department "NeuroFarBa", University of Florence, Florence 50139, Italy.
| | - Cecilia Ferrantini
- Division of Physiology, Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy.
| | - Ping Yan
- R. D. Berlin Center for Cell Analysis and Modeling, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | - Leslie M Loew
- R. D. Berlin Center for Cell Analysis and Modeling, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | - Corrado Poggesi
- Division of Physiology, Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy.
| | - Elisabetta Cerbai
- Division of Pharmacology, Department "NeuroFarBa", University of Florence, Florence 50139, Italy.
| | - Francesco S Pavone
- European Laboratory for Non-Linear Spectroscopy, Florence 50019, Italy.
- Department of Physics and Astronomy, University of Florence, Sesto Fiorentino 50019, Italy.
| | - Leonardo Sacconi
- European Laboratory for Non-Linear Spectroscopy, Florence 50019, Italy.
- National Institute of Optics, National Research Council, Florence 50125, Italy.
| |
Collapse
|
9
|
Helmes M, Najafi A, Palmer BM, Breel E, Rijnveld N, Iannuzzi D, van der Velden J. Mimicking the cardiac cycle in intact cardiomyocytes using diastolic and systolic force clamps; measuring power output. Cardiovasc Res 2016; 111:66-73. [PMID: 27037258 PMCID: PMC5853507 DOI: 10.1093/cvr/cvw072] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 03/26/2016] [Indexed: 01/08/2023] Open
Abstract
Aims A single isolated cardiomyocyte is the smallest functional unit of the heart. Yet, all single isolated cardiomyocyte experiments have been limited by the lack of proper methods that could reproduce a physiological cardiac cycle. We aimed to investigate the contractile properties of a single cardiomyocyte that correctly mimic the cardiac cycle. Methods and results By adjusting the parameters of the feedback loop, using a suitably engineered feedback system and recording the developed force and the length of a single rat cardiomyocyte during contraction and relaxation, we were able to construct force–length (FL) relations analogous to the pressure–volume (PV) relations at the whole heart level. From the cardiac loop graphs, we obtained, for the first time, the power generated by one single cardiomyocyte. Conclusion Here, we introduce a new approach that by combining mechanics, electronics, and a new type optical force transducer can measure the FL relationship of a single isolated cardiomyocyte undergoing a mechanical loop that mimics the PV cycle of a beating heart.
Collapse
Affiliation(s)
- Michiel Helmes
- Department of Physiology, VU University Medical Center, Institute for Cardiovascular Research (ICaR-VU), van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands IonOptix Llc., Milton, MA, USA
| | - Aref Najafi
- Department of Physiology, VU University Medical Center, Institute for Cardiovascular Research (ICaR-VU), van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands ICIN-Netherlands Heart Institute, Utrecht, The Netherlands
| | | | - Ernst Breel
- Biophotonics and Medical Imaging and Laserlab, VU University Amsterdam, Amsterdam, The Netherlands Optics11 BV, Amsterdam, The Netherlands
| | | | - Davide Iannuzzi
- Biophotonics and Medical Imaging and Laserlab, VU University Amsterdam, Amsterdam, The Netherlands
| | - Jolanda van der Velden
- Department of Physiology, VU University Medical Center, Institute for Cardiovascular Research (ICaR-VU), van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands ICIN-Netherlands Heart Institute, Utrecht, The Netherlands
| |
Collapse
|
10
|
Zhang W, Qu X, Chen B, Snyder M, Wang M, Li B, Tang Y, Chen H, Zhu W, Zhan L, Yin N, Li D, Xie L, Liu Y, Zhang JJ, Fu XY, Rubart M, Song LS, Huang XY, Shou W. Critical Roles of STAT3 in β-Adrenergic Functions in the Heart. Circulation 2016; 133:48-61. [PMID: 26628621 PMCID: PMC4698100 DOI: 10.1161/circulationaha.115.017472] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 10/02/2015] [Indexed: 01/08/2023]
Abstract
BACKGROUND β-Adrenergic receptors (βARs) play paradoxical roles in the heart. On one hand, βARs augment cardiac performance to fulfill the physiological demands, but on the other hand, prolonged activations of βARs exert deleterious effects that result in heart failure. The signal transducer and activator of transcription 3 (STAT3) plays a dynamic role in integrating multiple cytokine signaling pathways in a number of tissues. Altered activation of STAT3 has been observed in failing hearts in both human patients and animal models. Our objective is to determine the potential regulatory roles of STAT3 in cardiac βAR-mediated signaling and function. METHODS AND RESULTS We observed that STAT3 can be directly activated in cardiomyocytes by β-adrenergic agonists. To follow up this finding, we analyzed βAR function in cardiomyocyte-restricted STAT3 knockouts and discovered that the conditional loss of STAT3 in cardiomyocytes markedly reduced the cardiac contractile response to acute βAR stimulation, and caused disengagement of calcium coupling and muscle contraction. Under chronic β-adrenergic stimulation, Stat3cKO hearts exhibited pronounced cardiomyocyte hypertrophy, cell death, and subsequent cardiac fibrosis. Biochemical and genetic data supported that Gαs and Src kinases are required for βAR-mediated activation of STAT3. Finally, we demonstrated that STAT3 transcriptionally regulates several key components of βAR pathway, including β1AR, protein kinase A, and T-type Ca(2+) channels. CONCLUSIONS Our data demonstrate for the first time that STAT3 has a fundamental role in βAR signaling and functions in the heart. STAT3 serves as a critical transcriptional regulator for βAR-mediated cardiac stress adaption, pathological remodeling, and heart failure.
Collapse
Affiliation(s)
- Wenjun Zhang
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.).
| | - Xiuxia Qu
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Biyi Chen
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Marylynn Snyder
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Meijing Wang
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Baiyan Li
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Yue Tang
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Hanying Chen
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Wuqiang Zhu
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Li Zhan
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Ni Yin
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Deqiang Li
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Li Xie
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Ying Liu
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - J Jillian Zhang
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Xin-Yuan Fu
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Michael Rubart
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Long-Sheng Song
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Xin-Yun Huang
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Weinian Shou
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.).
| |
Collapse
|
11
|
Calcium sensitizers: What have we learned over the last 25years? Int J Cardiol 2016; 203:543-8. [DOI: 10.1016/j.ijcard.2015.10.240] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 10/30/2015] [Accepted: 10/31/2015] [Indexed: 01/10/2023]
|
12
|
Ferron AJT, Jacobsen BB, Sant’Ana PG, de Campos DHS, de Tomasi LC, Luvizotto RDAM, Cicogna AC, Leopoldo AS, Lima-Leopoldo AP. Cardiac Dysfunction Induced by Obesity Is Not Related to β-Adrenergic System Impairment at the Receptor-Signalling Pathway. PLoS One 2015; 10:e0138605. [PMID: 26390297 PMCID: PMC4577087 DOI: 10.1371/journal.pone.0138605] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 09/01/2015] [Indexed: 01/29/2023] Open
Abstract
Obesity has been shown to impair myocardial performance. Some factors have been suggested as responsible for possible cardiac abnormalities in models of obesity, among them beta-adrenergic (βA) system, an important mechanism of regulation of myocardial contraction and relaxation. The objective of present study was to evaluate the involvement of βA system components in myocardial dysfunction induced by obesity. Thirty-day-old male Wistar rats were distributed in control (C, n = 25) and obese (Ob, n = 25) groups. The C group was fed a standard diet and Ob group was fed four unsaturated high-fat diets for 15 weeks. Cardiac function was evaluated by isolated papillary muscle preparation and βA system evaluated by using cumulative concentrations of isoproterenol and Western blot. After 15 weeks, the Ob rats developed higher adiposity index than C rats and several comorbidities; however, were not associated with changes in systolic blood pressure. Obesity caused structural changes and the myocardial responsiveness to post-rest contraction stimulus and increased extracellular calcium (Ca2+) was compromised. There were no changes in cardiac function between groups after βA stimulation. The obesity was not accompanied by changes in protein expression of G protein subunit alpha (Gsα) and βA receptors (β1AR and β2AR). In conclusion, the myocardial dysfunction caused by unsaturated high-fat diet-induced obesity, after 15 weeks, is not related to βAR system impairment at the receptor-signalling pathway.
Collapse
Affiliation(s)
- Artur Junio Togneri Ferron
- Center of Physical Education and Sports, Department of Sports, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Bruno Barcellos Jacobsen
- Center of Physical Education and Sports, Department of Sports, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Paula Grippa Sant’Ana
- Department of Clinical and Cardiology, School of Medicine, UNESP- Univ. Estadual Paulista, Botucatu, São Paulo, Brazil
| | | | - Loreta Casquel de Tomasi
- Department of Clinical and Cardiology, School of Medicine, UNESP- Univ. Estadual Paulista, Botucatu, São Paulo, Brazil
| | | | - Antonio Carlos Cicogna
- Department of Clinical and Cardiology, School of Medicine, UNESP- Univ. Estadual Paulista, Botucatu, São Paulo, Brazil
| | - André Soares Leopoldo
- Center of Physical Education and Sports, Department of Sports, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Ana Paula Lima-Leopoldo
- Center of Physical Education and Sports, Department of Sports, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
- * E-mail:
| |
Collapse
|
13
|
Singh A, Mallick BN. Targeting modulation of noradrenalin release in the brain for amelioration of REMS loss-associated effects. J Transl Int Med 2015; 3:8-16. [PMID: 27847879 PMCID: PMC4936468 DOI: 10.4103/2224-4018.154288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Rapid eye movement sleep (REMS) loss affects most of the physiological processes, and it has been proposed that REMS maintains normal physiological processes. Changes in cultural, social, personal traits and life-style severely affect the amount and pattern of sleep, including REMS, which then manifests symptoms in animals, including humans. The effects may vary from simple fatigue and irritability to severe patho-physiological and behavioral deficits such as cognitive and behavioral dysfunctions. It has been a challenge to identify a molecule(s) that may have a potential for treating REMS loss-associated symptoms, which are very diverse. For decades, the critical role of locus coeruleus neurons in regulating REMS has been known, which has further been supported by the fact that the noradrenalin (NA) level is elevated in the brain after REMS loss. In this review, we have collected evidence from the published literature, including those from this laboratory, and argue that factors that affect REMS and vice versa modulate the level of a common molecule, the NA. Further, NA is known to affect the physiological processes affected by REMS loss. Therefore, we propose that modulation of the level of NA in the brain may be targeted for treating REMS loss-related symptoms. Further, we also argue that among the various ways to affect the release of NA-level, targeting α2 adrenoceptor autoreceptor on the pre-synaptic terminal may be the better option for ameliorating REMS loss-associated symptoms.
Collapse
Affiliation(s)
- Abhishek Singh
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | | |
Collapse
|
14
|
Bondarenko VE. A compartmentalized mathematical model of the β1-adrenergic signaling system in mouse ventricular myocytes. PLoS One 2014; 9:e89113. [PMID: 24586529 PMCID: PMC3931689 DOI: 10.1371/journal.pone.0089113] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 01/14/2014] [Indexed: 01/08/2023] Open
Abstract
The β1-adrenergic signaling system plays an important role in the functioning of cardiac cells. Experimental data shows that the activation of this system produces inotropy, lusitropy, and chronotropy in the heart, such as increased magnitude and relaxation rates of [Ca2+]i transients and contraction force, and increased heart rhythm. However, excessive stimulation of β1-adrenergic receptors leads to heart dysfunction and heart failure. In this paper, a comprehensive, experimentally based mathematical model of the β1-adrenergic signaling system for mouse ventricular myocytes is developed, which includes major subcellular functional compartments (caveolae, extracaveolae, and cytosol). The model describes biochemical reactions that occur during stimulation of β1-adrenoceptors, changes in ionic currents, and modifications of Ca2+ handling system. Simulations describe the dynamics of major signaling molecules, such as cyclic AMP and protein kinase A, in different subcellular compartments; the effects of inhibition of phosphodiesterases on cAMP production; kinetics and magnitudes of phosphorylation of ion channels, transporters, and Ca2+ handling proteins; modifications of action potential shape and duration; magnitudes and relaxation rates of [Ca2+]i transients; changes in intracellular and transmembrane Ca2+ fluxes; and [Na+]i fluxes and dynamics. The model elucidates complex interactions of ionic currents upon activation of β1-adrenoceptors at different stimulation frequencies, which ultimately lead to a relatively modest increase in action potential duration and significant increase in [Ca2+]i transients. In particular, the model includes two subpopulations of the L-type Ca2+ channels, in caveolae and extracaveolae compartments, and their effects on the action potential and [Ca2+]i transients are investigated. The presented model can be used by researchers for the interpretation of experimental data and for the developments of mathematical models for other species or for pathological conditions.
Collapse
Affiliation(s)
- Vladimir E. Bondarenko
- Department of Mathematics and Statistics and Neuroscience Institute, Georgia State University, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
15
|
Li S, Chen G, Li RA. Calcium signalling of human pluripotent stem cell-derived cardiomyocytes. J Physiol 2013; 591:5279-90. [PMID: 24018947 DOI: 10.1113/jphysiol.2013.256495] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Loss of cardiomyocytes (CMs), which lack the innate ability to regenerate, due to ageing or pathophysiological conditions (e.g. myocardial infarction or MI) is generally considered irreversible, and can lead to conditions from cardiac arrhythmias to heart failure. Human (h) pluripotent stem cells (PSCs), including embryonic stem cells (ESC) and induced pluripotent stem cells (iPSCs), can self-renew while maintaining their pluripotency to differentiate into all cell types, including CMs. Therefore, hPSCs provide a potential unlimited ex vivo source of human CMs for disease modelling, drug discovery, cardiotoxicity screening and cell-based heart therapies. As a fundamental property of working CMs, Ca(2+) signalling and its role in excitation-contraction coupling are well described. However, the biology of these processes in hPSC-CMs is just becoming understood. Here we review what is known about the immature Ca(2+)-handling properties of hPSC-CMs, at the levels of global transients and sparks, and the underlying molecular basis in relation to the development of various in vitro approaches to drive their maturation.
Collapse
Affiliation(s)
- Sen Li
- R. A. Li: 5/F Hong Kong Jockey Club Building for Interdisciplinary Research, 5 Sassoon Road, Pokfulam, Hong Kong.
| | | | | |
Collapse
|
16
|
Lymperopoulos A, Rengo G, Koch WJ. Adrenergic nervous system in heart failure: pathophysiology and therapy. Circ Res 2013; 113:739-753. [PMID: 23989716 PMCID: PMC3843360 DOI: 10.1161/circresaha.113.300308] [Citation(s) in RCA: 433] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 03/28/2013] [Indexed: 12/17/2022]
Abstract
Heart failure (HF), the leading cause of death in the western world, develops when a cardiac injury or insult impairs the ability of the heart to pump blood and maintain tissue perfusion. It is characterized by a complex interplay of several neurohormonal mechanisms that become activated in the syndrome to try and sustain cardiac output in the face of decompensating function. Perhaps the most prominent among these neurohormonal mechanisms is the adrenergic (or sympathetic) nervous system (ANS), whose activity and outflow are enormously elevated in HF. Acutely, and if the heart works properly, this activation of the ANS will promptly restore cardiac function. However, if the cardiac insult persists over time, chances are the ANS will not be able to maintain cardiac function, the heart will progress into a state of chronic decompensated HF, and the hyperactive ANS will continue to push the heart to work at a level much higher than the cardiac muscle can handle. From that point on, ANS hyperactivity becomes a major problem in HF, conferring significant toxicity to the failing heart and markedly increasing its morbidity and mortality. The present review discusses the role of the ANS in cardiac physiology and in HF pathophysiology, the mechanisms of regulation of ANS activity and how they go awry in chronic HF, methods of measuring ANS activity in HF, the molecular alterations in heart physiology that occur in HF, along with their pharmacological and therapeutic implications, and, finally, drugs and other therapeutic modalities used in HF treatment that target or affect the ANS and its effects on the failing heart.
Collapse
Affiliation(s)
- Anastasios Lymperopoulos
- Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Ft. Lauderdale, FL, USA
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, and Division of Cardiology, Fondazione Salvatore Maugeri, Telese Terme, Italy
| | - Walter J. Koch
- Center for Translational Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
17
|
Vatner SF, Park M, Yan L, Lee GJ, Lai L, Iwatsubo K, Ishikawa Y, Pessin J, Vatner DE. Adenylyl cyclase type 5 in cardiac disease, metabolism, and aging. Am J Physiol Heart Circ Physiol 2013; 305:H1-8. [PMID: 23624627 DOI: 10.1152/ajpheart.00080.2013] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
G protein-coupled receptor/adenylyl cyclase (AC)/cAMP signaling is crucial for all cellular responses to physiological and pathophysiological stimuli. There are nine isoforms of membrane-bound AC, with type 5 being one of the two major isoforms in the heart. Since the role of AC in the heart in regulating cAMP and acute changes in inotropic and chronotropic state are well known, this review will address our current understanding of the distinct regulatory role of the AC5 isoform in response to chronic stress. Transgenic overexpression of AC5 in cardiomyocytes of the heart (AC5-Tg) improves baseline cardiac function but impairs the ability of the heart to withstand stress. For example, chronic catecholamine stimulation induces cardiomyopathy, which is more severe in AC5-Tg mice, mediated through the AC5/sirtuin 1/forkhead box O3a pathway. Conversely, disrupting AC5, i.e., AC5 knockout, protects the heart from chronic catecholamine cardiomyopathy as well as the cardiomyopathies resulting from chronic pressure overload or aging. Moreover, AC5 knockout results in a 30% increase in a healthy life span, resembling the most widely studied model of longevity, i.e., calorie restriction. These two models of longevity share similar gene regulation in the heart, muscle, liver, and brain in that they are both protected against diabetes, obesity, and diabetic and aging cardiomyopathy. A pharmacological inhibitor of AC5 also provides protection against cardiac stress, diabetes, and obesity. Thus AC5 inhibition has novel, potential therapeutic applicability to several diseases not only in the heart but also in aging, diabetes, and obesity.
Collapse
Affiliation(s)
- Stephen F Vatner
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, Newark, NJ 07103, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Goonasekera SA, Hammer K, Auger-Messier M, Bodi I, Chen X, Zhang H, Reiken S, Elrod JW, Correll RN, York AJ, Sargent MA, Hofmann F, Moosmang S, Marks AR, Houser SR, Bers DM, Molkentin JD. Decreased cardiac L-type Ca²⁺ channel activity induces hypertrophy and heart failure in mice. J Clin Invest 2011; 122:280-90. [PMID: 22133878 DOI: 10.1172/jci58227] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Accepted: 10/12/2011] [Indexed: 02/06/2023] Open
Abstract
Antagonists of L-type Ca²⁺ channels (LTCCs) have been used to treat human cardiovascular diseases for decades. However, these inhibitors can have untoward effects in patients with heart failure, and their overall therapeutic profile remains nebulous given differential effects in the vasculature when compared with those in cardiomyocytes. To investigate this issue, we examined mice heterozygous for the gene encoding the pore-forming subunit of LTCC (calcium channel, voltage-dependent, L type, α1C subunit [Cacna1c mice; referred to herein as α1C⁻/⁺ mice]) and mice in which this gene was loxP targeted to achieve graded heart-specific gene deletion (termed herein α1C-loxP mice). Adult cardiomyocytes from the hearts of α1C⁻/⁺ mice at 10 weeks of age showed a decrease in LTCC current and a modest decrease in cardiac function, which we initially hypothesized would be cardioprotective. However, α1C⁻/⁺ mice subjected to pressure overload stimulation, isoproterenol infusion, and swimming showed greater cardiac hypertrophy, greater reductions in ventricular performance, and greater ventricular dilation than α1C⁺/⁺ controls. The same detrimental effects were observed in α1C-loxP animals with a cardiomyocyte-specific deletion of one allele. More severe reductions in α1C protein levels with combinatorial deleted alleles produced spontaneous cardiac hypertrophy before 3 months of age, with early adulthood lethality. Mechanistically, our data suggest that a reduction in LTCC current leads to neuroendocrine stress, with sensitized and leaky sarcoplasmic reticulum Ca²⁺ release as a compensatory mechanism to preserve contractility. This state results in calcineurin/nuclear factor of activated T cells signaling that promotes hypertrophy and disease.
Collapse
Affiliation(s)
- Sanjeewa A Goonasekera
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Howard Hughes Medical Institute, Cincinnati, Ohio 45229, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Sharma V, McNeill JH. Parallel effects of β-adrenoceptor blockade on cardiac function and fatty acid oxidation in the diabetic heart: Confronting the maze. World J Cardiol 2011; 3:281-302. [PMID: 21949571 PMCID: PMC3176897 DOI: 10.4330/wjc.v3.i9.281] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 07/18/2011] [Accepted: 07/25/2011] [Indexed: 02/06/2023] Open
Abstract
Diabetic cardiomyopathy is a disease process in which diabetes produces a direct and continuous myocardial insult even in the absence of ischemic, hypertensive or valvular disease. The β-blocking agents bisoprolol, carvedilol and metoprolol have been shown in large-scale randomized controlled trials to reduce heart failure mortality. In this review, we summarize the results of our studies investigating the effects of β-blocking agents on cardiac function and metabolism in diabetic heart failure, and the complex inter-related mechanisms involved. Metoprolol inhibits fatty acid oxidation at the mitochondrial level but does not prevent lipotoxicity; its beneficial effects are more likely to be due to pro-survival effects of chronic treatment. These studies have expanded our understanding of the range of effects produced by β-adrenergic blockade and show how interconnected the signaling pathways of function and metabolism are in the heart. Although our initial hypothesis that inhibition of fatty acid oxidation would be a key mechanism of action was disproved, unexpected results led us to some intriguing regulatory mechanisms of cardiac metabolism. The first was upstream stimulatory factor-2-mediated repression of transcriptional master regulator PGC-1α, most likely occurring as a consequence of the improved function; it is unclear whether this effect is unique to β-blockers, although repression of carnitine palmitoyltransferase (CPT)-1 has not been reported with other drugs which improve function. The second was the identification of a range of covalent modifications which can regulate CPT-1 directly, mediated by a signalome at the level of the mitochondria. We also identified an important interaction between β-adrenergic signaling and caveolins, which may be a key mechanism of action of β-adrenergic blockade. Our experience with this labyrinthine signaling web illustrates that initial hypotheses and anticipated directions do not have to be right in order to open up meaningful directions or reveal new information.
Collapse
Affiliation(s)
- Vijay Sharma
- Vijay Sharma, John H McNeill, Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, V6T 1Z3.F, Canada
| | | |
Collapse
|
20
|
Roe ND, Ren J. Akt2 knockout mitigates chronic iNOS inhibition-induced cardiomyocyte atrophy and contractile dysfunction despite persistent insulin resistance. Toxicol Lett 2011; 207:222-31. [PMID: 21964073 DOI: 10.1016/j.toxlet.2011.09.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2011] [Revised: 09/11/2011] [Accepted: 09/12/2011] [Indexed: 11/18/2022]
Abstract
Increased levels of inducible nitric oxide synthase (iNOS) during cardiac stress such as ischemia-reperfusion, sepsis and hypertension may display both beneficial and detrimental roles in cardiac contractile performance. However, the precise role of iNOS in the maintenance of cardiac contractile function remains elusive. This study was designed to determine the impact of chronic iNOS inhibition on cardiac contractile function and the underlying mechanism involved with a special focus on the NO downstream signaling molecule Akt. Male C57 or Akt2 knockout [Akt2(-/-)] mice were injected with the specific iNOS inhibitor 1400W (2 mg/kg/d) or saline for 7 days. Both 1400W and Akt2 knockout dampened glucose and insulin tolerance without additive effects. Treatment of 1400W decreased heart and liver weights as well as cardiomyocyte cross-sectional area in C57 but not Akt2 knockout mice. 1400W but not Akt2 knockout compromised cardiomyocyte mechanical properties including decreased peak shortening and maximal velocity of shortening/relengthening, prolonged relengthening duration, reduced intracellular Ca(2+) release and decay rate, the effects of which were ablated or attenuated by Akt2 knockout. Akt2 knockout but not 1400W increased the levels of intracellular Ca(2+) regulatory proteins including SERCA2a and phospholamban phosphorylation. 1400W reduced the level of anti-apoptotic protein Bcl-2, the effect of which was unaffected by Akt2 knockout. Neither 1400W nor Akt2 knockout significantly affected ER stress, autophagy, the post-insulin receptor signaling Akt, GSK3β and AMPK, as well as the stress signaling IκB, JNK, ERK and p38 with the exception of elevated IκB phosphorylation with jointed effect of 1400W and Akt2 knockout. Taken together, these data indicated that an essential role of iNOS in the maintenance of cardiac morphology and function possibly through an Akt2-dependent mechanism.
Collapse
Affiliation(s)
- Nathan D Roe
- Division of Pharmaceutical Sciences & Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | | |
Collapse
|
21
|
Harvey RD, Calaghan SC. Caveolae create local signalling domains through their distinct protein content, lipid profile and morphology. J Mol Cell Cardiol 2011; 52:366-75. [PMID: 21782827 DOI: 10.1016/j.yjmcc.2011.07.007] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Revised: 06/21/2011] [Accepted: 07/07/2011] [Indexed: 01/02/2023]
Abstract
Compartmentation of signalling allows multiple stimuli to achieve diverse cellular responses with only a limited pool of second messengers. This spatial control of signalling is achieved, in part, by cellular structures which bring together elements of a particular cascade. One such structure is the caveola, a flask-shaped lipid raft. Caveolae are well-recognised as signalosomes, platforms for assembly of signalling complexes of receptors, effectors and their targets, which can facilitate efficient and specific cellular responses. Here we extend this simple model and present evidence to show how the protein and lipid profiles of caveolae, as well as their characteristic morphology, define their roles in creating local signalling domains in the cardiac myocyte. This article is part of a Special Issue entitled "Local Signaling in Myocytes."
Collapse
Affiliation(s)
- Robert D Harvey
- Department of Pharmacology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | | |
Collapse
|
22
|
MacDougall DA, Agarwal SR, Stopford EA, Chu H, Collins JA, Longster AL, Colyer J, Harvey RD, Calaghan S. Caveolae compartmentalise β2-adrenoceptor signals by curtailing cAMP production and maintaining phosphatase activity in the sarcoplasmic reticulum of the adult ventricular myocyte. J Mol Cell Cardiol 2011; 52:388-400. [PMID: 21740911 PMCID: PMC3270222 DOI: 10.1016/j.yjmcc.2011.06.014] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 06/02/2011] [Accepted: 06/20/2011] [Indexed: 01/24/2023]
Abstract
Inotropy and lusitropy in the ventricular myocyte can be efficiently induced by activation of β1-, but not β2-, adrenoceptors (ARs). Compartmentation of β2-AR-derived cAMP-dependent signalling underlies this functional discrepancy. Here we investigate the mechanism by which caveolae (specialised sarcolemmal invaginations rich in cholesterol and caveolin-3) contribute to compartmentation in the adult rat ventricular myocyte. Selective activation of β2-ARs (with zinterol/CGP20712A) produced little contractile response in control cells but pronounced inotropic and lusitropic responses in cells treated with the cholesterol-depleting agent methyl-β-cyclodextrin (MBCD). This was not linked to modulation of L-type Ca2+ current, but instead to a discrete PKA-mediated phosphorylation of phospholamban at Ser16. Application of a cell-permeable inhibitor of caveolin-3 scaffolding interactions mimicked the effect of MBCD on phosphorylated phospholamban (pPLB) during β2-AR stimulation, consistent with MBCD acting via caveolae. Biosensor experiments revealed β2-AR mobilisation of cAMP in PKA II signalling domains of intact cells only after MBCD treatment, providing a real-time demonstration of cAMP freed from caveolar constraint. Other proteins have roles in compartmentation, so the effects of phosphodiesterase (PDE), protein phosphatase (PP) and phosphoinositide-3-kinase (PI3K) inhibitors on pPLB and contraction were compared in control and MBCD treated cells. PP inhibition alone was conspicuous in showing robust de-compartmentation of β2-AR-derived signalling in control cells and a comparatively diminutive effect after cholesterol depletion. Collating all evidence, we promote the novel concept that caveolae limit β2-AR-cAMP signalling by providing a platform that not only attenuates production of cAMP but also prevents inhibitory modulation of PPs at the sarcoplasmic reticulum. This article is part of a Special Issue entitled “Local Signaling in Myocytes”.
Collapse
Affiliation(s)
- David A. MacDougall
- Institute of Membrane and Systems Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Shailesh R. Agarwal
- Department of Pharmacology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | | | - Hongjin Chu
- Institute of Membrane and Systems Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Jennifer A. Collins
- Institute of Membrane and Systems Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Anna L. Longster
- Institute of Membrane and Systems Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - John Colyer
- Institute of Membrane and Systems Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Robert D. Harvey
- Department of Pharmacology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Sarah Calaghan
- Institute of Membrane and Systems Biology, University of Leeds, Leeds, LS2 9JT, UK
- Corresponding author at: Institute of Membrane and Systems Biology, Garstang 7.52d, University of Leeds, Leeds LS2 9JT, UK. Tel.: + 44 113 343 4309; fax: + 44 113 343 4228.
| |
Collapse
|
23
|
Adameova A, Abdellatif Y, Dhalla NS. Role of the excessive amounts of circulating catecholamines and glucocorticoids in stress-induced heart disease. Can J Physiol Pharmacol 2010; 87:493-514. [PMID: 19767873 DOI: 10.1139/y09-042] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Various stressful stimuli are known to activate the sympathetic nervous system to release catecholamines and the hypothalamic-pituitary-adrenal axis to release glucocorticoids in the circulation. Although initial actions of both catecholamines and glucocorticoids are beneficial for the function of the cardiovascular system, their delayed effects on the heart are deleterious. Glucocorticoids not only increase plasma levels of catecholamines by inhibiting their extraneuronal uptake, but they have also been shown to induce supersensitivity to catecholamines in the heart by upregulating different components of the betta-adrenoceptor signal transduction system. Low concentrations of catecholamines stimulate the heart by promoting Ca2+ movements, whereas excessive amounts of catecholamines produce cardiac dysfunction by inducing intracellular Ca2+ overload in cardiomyocytes. Several studies have shown, however, that under stressful conditions high concentrations of catecholamines become oxidized to form aminolutins and generate oxyradicals. These oxidation products of catecholamines have been demonstrated to produce coronary spasm, arrhythmias, and cardiac dysfunction by inducing Ca2+-handling abnormalities in both sarcolemmal and sarcoplasmic reticulum, defects in energy production by mitochondria, and myocardial cell damage. In this article we have focused the discussion to highlight the interrelationship between catecholamines and glucocorticoids and to emphasize the role of oxidation products of catecholamines in the development of stress-induced heart disease.
Collapse
Affiliation(s)
- Adriana Adameova
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, and Department of Physiology, Faculty of Medicine, University of Manitoba, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada
| | | | | |
Collapse
|
24
|
Triposkiadis F, Karayannis G, Giamouzis G, Skoularigis J, Louridas G, Butler J. The sympathetic nervous system in heart failure physiology, pathophysiology, and clinical implications. J Am Coll Cardiol 2009; 54:1747-62. [PMID: 19874988 DOI: 10.1016/j.jacc.2009.05.015] [Citation(s) in RCA: 668] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2009] [Revised: 05/11/2009] [Accepted: 05/14/2009] [Indexed: 01/12/2023]
Abstract
Heart failure is a syndrome characterized initially by left ventricular dysfunction that triggers countermeasures aimed to restore cardiac output. These responses are compensatory at first but eventually become part of the disease process itself leading to further worsening cardiac function. Among these responses is the activation of the sympathetic nervous system (SNS) that provides inotropic support to the failing heart increasing stroke volume, and peripheral vasoconstriction to maintain mean arterial perfusion pressure, but eventually accelerates disease progression affecting survival. Activation of SNS has been attributed to withdrawal of normal restraining influences and enhancement of excitatory inputs including changes in: 1) peripheral baroreceptor and chemoreceptor reflexes; 2) chemical mediators that control sympathetic outflow; and 3) central integratory sites. The interface between the sympathetic fibers and the cardiovascular system is formed by the adrenergic receptors (ARs). Dysregulation of cardiac beta(1)-AR signaling and transduction are key features of heart failure progression. In contrast, cardiac beta(2)-ARs and alpha(1)-ARs may function in a compensatory fashion to maintain cardiac inotropy. Adrenergic receptor polymorphisms may have an impact on the adaptive mechanisms, susceptibilities, and pharmacological responses of SNS. The beta-AR blockers and the inhibitors of the renin-angiotensin-aldosterone axis form the mainstay of current medical management of chronic heart failure. Conversely, central sympatholytics have proved harmful, whereas sympathomimetic inotropes are still used in selected patients with hemodynamic instability. This review summarizes the changes in SNS in heart failure and examines how modulation of SNS activity may affect morbidity and mortality from this syndrome.
Collapse
|
25
|
Elucidating the role of reversible protein phosphorylation in sepsis-induced myocardial dysfunction. Shock 2009; 32:49-54. [PMID: 19533850 DOI: 10.1097/shk.0b013e3181991926] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mortality in children with sepsis is most often related to diminished cardiac output with cardiovascular collapse, resulting in impaired oxygen delivery and, ultimately, end-organ failure. Although cardiovascular "collapse" is commonly observed in individuals with septic shock, the hemodynamic causes of this differ greatly. In children, intrinsic myocardial dysfunction is most commonly present, whereas the systemic vascular resistance is typically high. This pattern is distinct from adults with sepsis where the principal hemodynamic profile shows elevated cardiac output, but substantially reduced systemic vascular resistance. Various studies support the concept that myocardial dysfunction, as occurs in pediatric septic patients, is due to intrinsic abnormalities in cardiomyocyte function and is not related to hypoperfusion as a result of low systemic vascular resistance. Importantly, when examined more closely, data from adults with septic shock also reveal that intrinsic myocardial dysfunction may play a larger role than previously appreciated. As a result, cardiovascular support, especially in pediatric sepsis, requires a treatment strategy directed at the underlying mechanism(s) responsible for this dysfunction. Thus, it is imperative to gain a better understanding of the myocardial derangements that occur during sepsis to identify targets that will ultimately influence the management of children with septic shock and favorably alter the associated mortality. We hypothesize that key signaling pathways that control myocardial calcium flux, regulated to key kinases and phosphatases, influence myocyte contractility in sepsis. Thus, we review the data relevant to the sepsis-induced intracellular alterations in calcium flux in the cardiomyocyte, with an emphasis on changes in the phosphorylation state of the contractile proteins regulated by the balance between kinases and phosphatases. We believe that therapies modulating the activity of these key proteins may provide an improvement in calcium handling and myocardial contractility and alter the clinical outcomes in sepsis.
Collapse
|
26
|
Abstract
AbstractThe aim of this study is to investigate the mechanism of positive inotropic effect of obestatin on in vitro heart preparations of Rana ridibunda frog. The application of increasing amounts of obestatin in the concentration range from 1 μmol/l to 1 μmol/l significantly enhances the force of contraction of excised and cannulated frog hearts. This effect was partially reduced in the presence of prazosin (3 μmol/l). Propranolol (30 μmol/l), pertussis toxin (2 ng/ml) and the specific inhibitor of cAMP-dependent protein kinase (PKA) Rp-adenosine 3′,5′-cyclic monophosphothioate triethylamine (30 μmol/l) completely blocked the obestatin-induced increase of the force of frog heart contractions. It is concluded that, via its receptor molecule, obestatin activates neuronal pertussis toxin sensitive G-protein(s) that further enhance the secretion of epinephrine from sympathetic neurons. This epinephrine activates mainly the myocardial β-adrenoreceptors and PKA downstream targets, and is responsible for the observed positive inotropic effect of obestatin. An alternative explanation of our data is that obestatin directly enhances the effect of myocardial β-adrenergic signaling.
Collapse
|
27
|
Muthumala A, Drenos F, Elliott PM, Humphries SE. Role of beta adrenergic receptor polymorphisms in heart failure: systematic review and meta-analysis. Eur J Heart Fail 2007; 10:3-13. [PMID: 18158268 DOI: 10.1016/j.ejheart.2007.11.008] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2007] [Revised: 09/18/2007] [Accepted: 11/19/2007] [Indexed: 01/08/2023] Open
Abstract
Heart Failure (HF) is a common disorder associated with substantial morbidity and mortality. beta adrenergic receptors (betaAR) are the primary pathway through which cardiac function is influenced. Chronic beta(1)AR activation is implicated in the pathogenesis of HF and betaAR blockade improves survival in left ventricular systolic dysfunction. Common functional polymorphisms in beta adrenergic receptor genes (ADRB) have been associated with HF phenotypes, and with pharmacogenetic interaction with beta adrenergic receptor blockers (beta blockers). However, these associations have not been consistently replicated. The evidence for ADRB variant involvement in pathogenesis, progression and response to beta blockers in HF is reviewed. In addition, a meta-analysis of three studies analysing the effect of ADRB1 Arg389Gly polymorphism on left ventricular remodelling with the use of beta blockers, demonstrating a 5% improvement in left ventricular ejection fraction in Arg389 homozygotes, is presented. There is now accumulating molecular evidence for a different functional response to beta blockers associated with this polymorphism. In the future, confirmed genotypic associations may enable patients to be identified who are either at greater risk of developing HF, whose HF may rapidly progress, or who are unlikely to benefit from beta blockers, and such patients may benefit from targeted aggressive therapy.
Collapse
Affiliation(s)
- Amal Muthumala
- Centre for Cardiovascular Genetics, Rayne Institute, Royal Free and University College Medical School, London WC1E 6JF, UK.
| | | | | | | |
Collapse
|
28
|
Birkeland JAK, Swift F, Tovsrud N, Enger U, Lunde PK, Qvigstad E, Levy FO, Sejersted OM, Sjaastad I. Serotonin increases L-type Ca2+ current and SR Ca2+ content through 5-HT4 receptors in failing rat ventricular cardiomyocytes. Am J Physiol Heart Circ Physiol 2007; 293:H2367-76. [PMID: 17660386 DOI: 10.1152/ajpheart.01375.2006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Rats with congestive heart failure (CHF) develop ventricular inotropic responsiveness to serotonin (5-HT), mediated through 5-HT2A and 5-HT4 receptors. Human ventricle is similarly responsive to 5-HT through 5-HT4 receptors. We studied isolated ventricular cardiomyocytes to clarify the effects of 5-HT on intracellular Ca2+ handling. Left-ventricular cardiomyocytes were isolated from male Wistar rats 6 wk after induction of postinfarction CHF. Contractile function and Ca2+ transients were measured in field-stimulated cardiomyocytes, and L-type Ca2+ current ( ICa,L) and sarcoplasmic reticulum (SR) Ca2+ content were measured in voltage-clamped cells. Protein phosphorylation was measured by Western blotting or phosphoprotein gel staining. 5-HT4- and 5-HT2A-receptor stimulation induced a positive inotropic response of 33 and 18% (both P < 0.05) and also increased the Ca2+ transient (44 and 6%, respectively; both P < 0.05). ICa,L and SR Ca2+ content increased only after 5-HT4-receptor stimulation (57 and 65%; both P < 0.05). Phospholamban serine16 (PLB-Ser16) and troponin I phosphorylation increased by 26 and 13% after 5-HT4-receptor stimulation ( P < 0.05). 5-HT2A-receptor stimulation increased the action potential duration and did not significantly change the phosphorylation of PLB-Ser16 or troponin I, but it increased myosin light chain 2 (MLC2) phosphorylation. In conclusion, the positive inotropic response to 5-HT4 stimulation results from increased ICa,L and increased phosphorylation of PLB-Ser16, which increases the SR Ca2+ content. 5-HT4 stimulation is thus, like β-adrenoceptor stimulation, possibly energetically unfavorable in CHF. 5-HT2A-receptor stimulation, previously studied in acute CHF, induces a positive inotropic response also in chronic CHF, probably mediated by MLC2 phosphorylation.
Collapse
MESH Headings
- Action Potentials
- Adrenergic beta-Agonists/pharmacology
- Animals
- Calcium Channels, L-Type/drug effects
- Calcium Channels, L-Type/metabolism
- Calcium Signaling/drug effects
- Calcium-Binding Proteins/metabolism
- Cardiac Myosins
- Cardiotonic Agents/metabolism
- Coronary Vessels/surgery
- Disease Models, Animal
- Heart Failure/etiology
- Heart Failure/metabolism
- Heart Failure/physiopathology
- Indoles/pharmacology
- Isoproterenol/pharmacology
- Ketanserin/pharmacology
- Ligation
- Male
- Myocardial Contraction
- Myocardial Infarction/complications
- Myocardial Infarction/metabolism
- Myocardial Infarction/physiopathology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myosin Light Chains
- Phosphorylation
- Rats
- Rats, Wistar
- Receptor, Serotonin, 5-HT2A/metabolism
- Receptors, Serotonin, 5-HT4/metabolism
- Sarcoplasmic Reticulum/drug effects
- Sarcoplasmic Reticulum/metabolism
- Serotonin/metabolism
- Serotonin 5-HT2 Receptor Antagonists
- Serotonin 5-HT4 Receptor Antagonists
- Serotonin Antagonists/pharmacology
- Sulfonamides/pharmacology
- Time Factors
- Troponin I/metabolism
- Ventricular Function/drug effects
Collapse
Affiliation(s)
- Jon Arne Kro Birkeland
- Institute for Experimental Medical Research, Ullevaal Univ. Hospital, Kirkeveien 166, 0407 Oslo, Norway.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Altan VM, Arioglu E, Guner S, Ozcelikay AT. The influence of diabetes on cardiac β-adrenoceptor subtypes. Heart Fail Rev 2007; 12:58-65. [PMID: 17364227 DOI: 10.1007/s10741-007-9005-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2006] [Accepted: 02/13/2007] [Indexed: 11/30/2022]
Abstract
Despite the significant developments in the treatment of diabetes mellitus, diabetic patients still continue to suffer from cardiac complications. The increase of cardiac adrenergic drive may ultimately contribute to the development and progression of diabetic cardiomyopathy. beta-Adrenoceptors play an important role in the regulation of heart function. However, responsiveness of diabetic heart to beta-adrenoceptor agonist stimulation is diminished. The chronotropic responses mediated by beta(1)-subtype, which is mainly responsible for cardiac effects of catecholamines are decreased in the atria of diabetic rats. The expression of cardiac beta(1)-subtype is significantly decreased in diabetic rats as well. beta(2)-Adrenoceptors also increase cardiac function. Although the expression of this subtype is slightly decreased in diabetic rat hearts, beta(2)-mediated chronotropic responses are preserved. On the other hand, functional beta(3)-adrenoceptor subtype was characterized in human heart. Interestingly, stimulation of cardiac beta(3)-adrenoceptors, on the contrary of beta(1)- and beta(2)-subtypes, mediates negative inotropic effect in human ventricular muscle. Cardiac beta(3)-adrenoceptors are upregulated in experimental diabetes as well as in human heart failure. These findings suggest that each beta-adrenoceptor subtype may play an important role in the pathophysiology of diabetes-induced heart disease. However, it is still not known whether the changes in the expression and/or responsiveness of beta-adrenoceptors are adaptive or maladaptive. Therefore, this review outlines the potential roles of these receptor subtypes in cardiac pathologies of diabetes.
Collapse
Affiliation(s)
- V Melih Altan
- Department of Pharmacology, Faculty of Pharmacy, University of Ankara, Tandogan, Ankara, 06100, Turkey.
| | | | | | | |
Collapse
|
30
|
Brum PC, Rolim NPL, Bacurau AVN, Medeiros A. Neurohumoral activation in heart failure: the role of adrenergic receptors. AN ACAD BRAS CIENC 2007; 78:485-503. [PMID: 16936938 DOI: 10.1590/s0001-37652006000300009] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2005] [Accepted: 11/04/2005] [Indexed: 01/08/2023] Open
Abstract
Heart failure (HF) is a common endpoint for many forms of cardiovascular disease and a significant cause of morbidity and mortality. The development of end-stage HF often involves an initial insult to the myocardium that reduces cardiac output and leads to a compensatory increase in sympathetic nervous system activity. Acutely, the sympathetic hyperactivity through the activation of beta-adrenergic receptors increases heart rate and cardiac contractility, which compensate for decreased cardiac output. However, chronic exposure of the heart to elevated levels of catecholamines released from sympathetic nerve terminals and the adrenal gland may lead to further pathologic changes in the heart, resulting in continued elevation of sympathetic tone and a progressive deterioration in cardiac function. On a molecular level, altered beta-adrenergic receptor signaling plays a pivotal role in the genesis and progression of HF. beta-adrenergic receptor number and function are decreased, and downstream mechanisms are altered. In this review we will present an overview of the normal beta-adrenergic receptor pathway in the heart and the consequences of sustained adrenergic activation in HF. The myopathic potential of individual components of the adrenergic signaling will be discussed through the results of research performed in genetic modified animals. Finally, we will discuss the potential clinical impact of beta-adrenergic receptor gene polymorphisms for better understanding the progression of HF.
Collapse
MESH Headings
- Animals
- Cardiac Output, Low/physiopathology
- Disease Models, Animal
- Disease Progression
- Humans
- Mice
- Polymorphism, Genetic
- Receptors, Adrenergic, beta-1/genetics
- Receptors, Adrenergic, beta-1/physiology
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/physiology
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Patricia C Brum
- Laboratório de Fisiologia Cel. e Mol. do Exercício, Departamento de Biodinâmica do Movimento Humano, Escola de Educação Física e Esporte, Universidade de São Paulo, São Paulo, SP, Brasil.
| | | | | | | |
Collapse
|
31
|
Vandecasteele G, Rochais F, Abi-Gerges A, Fischmeister R. Functional localization of cAMP signalling in cardiac myocytes. Biochem Soc Trans 2006; 34:484-8. [PMID: 16856839 DOI: 10.1042/bst0340484] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The cAMP pathway is of cardinal importance for heart physiology and pathology. The spatial organization of the various components of the cAMP pathway is thought to allow the segregation of functional responses triggered by the different neuromediators and hormones that use this pathway. PDEs (phosphodiesterases) hydrolyse cAMP (and cGMP) and play a major role in this process by preventing cAMP diffusion to the whole cytosol and inadequate target activation. The development of olfactory cyclic nucleotide-gated channels to directly monitor cAMP beneath the plasma membrane in real time allows us to gain new insights into the molecular mechanisms responsible for cAMP homoeostasis and hormonal specificity in cardiac cells. The present review summarizes the recent results we obtained using this approach in adult rat ventricular myocytes. In particular, the role of PDEs in the maintenance of specific cAMP signals generated by beta-adrenergic receptors and other G(s)-coupled receptors will be discussed.
Collapse
Affiliation(s)
- G Vandecasteele
- INSERM U769, Faculté de Pharmacie, Université de Paris-Sud 11, Châtenay-Malabry, France.
| | | | | | | |
Collapse
|
32
|
Toh R, Shinohara M, Takaya T, Yamashita T, Masuda S, Kawashima S, Yokoyama M, Yagi N. An X-Ray diffraction study on mouse cardiac cross-bridge function in vivo: effects of adrenergic {beta}-stimulation. Biophys J 2006; 90:1723-8. [PMID: 16339874 PMCID: PMC1367321 DOI: 10.1529/biophysj.105.074062] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2005] [Accepted: 11/18/2005] [Indexed: 11/18/2022] Open
Abstract
To investigate how beta-stimulation affects the contractility of cardiac muscle, x-ray diffraction from cardiac muscle in the left ventricular free wall of a mouse heart was recorded in vivo. To our knowledge, this is the first x-ray diffraction study on a heart in a living body. After the R wave in electrocardiograms, the ratio of the intensities of the equatorial (1,0) and (1,1) reflections decreased for approximately 50 ms from a diastolic value of 2.1 to a minimum of 0.8, and then recovered. The spacing of the (1,0) lattice planes increased for approximately 90 ms from a diastolic value of 37.2 nm to a maximum of 39.1 nm, and then returned to the diastolic level, corresponding to approximately 10% stretch of sarcomere. Stimulation of beta-adrenergic receptor by dobutamine (20 microg/kg/min) accelerated both the decrease in the intensity ratio, which reached a smaller systolic value, and the increase in the lattice spacing. However, the intensity ratio and spacing at the end-diastole were unchanged. The recovery of the lattice spacing during relaxation was also accelerated. The mass transfer to the thin filaments at systole in a beta-stimulated heart was close to the peak value in twitch of frog skeletal muscle at 4 degrees C, showing that the majority of cross-bridges have been recruited with few in reserve.
Collapse
Affiliation(s)
- Ryuji Toh
- Division of Cardiovascular and Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | | | | | | | | | | | | | | |
Collapse
|
33
|
|
34
|
|
35
|
Pavoine C, Defer N. The cardiac beta2-adrenergic signalling a new role for the cPLA2. Cell Signal 2005; 17:141-52. [PMID: 15494206 DOI: 10.1016/j.cellsig.2004.09.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2004] [Revised: 09/01/2004] [Accepted: 09/01/2004] [Indexed: 01/08/2023]
Abstract
The cardiac actions of catecholamines have long been attributed to the predominant beta(1)-AR subtype that couples to the classical Gs/AC/cAMP pathway. Recent research clearly indicates that cardiac beta(2)-ARs play a functional role in healthy heart and assume increasing importance in failing and aged heart. beta(2)-ARs are primarily coupled to an atypical compartmentalized cAMP pathway, regulated by phosphorylation and/or oligomerization of beta(2)-ARs, and under the control of additional beta(2)-AR/Gi-coupled lipidic pathways, the impact of which seems to vary depending on the animal species, the developmental and pathophysiological state. This review focuses, more especially, on one of the last identified beta(2)-AR/Gi pathway, namely the cPLA(2).
Collapse
MESH Headings
- Animals
- Arachidonic Acid/metabolism
- Cardiotonic Agents/pharmacology
- Caveolae/metabolism
- Caveolae/physiology
- Cyclic AMP/metabolism
- Dimerization
- GTP-Binding Protein alpha Subunits, Gi-Go/physiology
- GTP-Binding Protein alpha Subunits, Gs/physiology
- Group IV Phospholipases A2
- Heart/drug effects
- Heart/physiology
- Humans
- Isoenzymes/chemistry
- Isoenzymes/physiology
- Models, Cardiovascular
- Myocardium/enzymology
- Myocardium/metabolism
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/physiology
- Phosphatidylinositol 3-Kinases/physiology
- Phospholipases A/chemistry
- Phospholipases A/physiology
- Receptors, Adrenergic, beta-1/physiology
- Receptors, Adrenergic, beta-2/metabolism
- Receptors, Adrenergic, beta-2/physiology
- Signal Transduction/physiology
- Species Specificity
- Ventricular Dysfunction/metabolism
- Ventricular Dysfunction/physiopathology
Collapse
|
36
|
Bukharaeva EA, Samigullin D, Nikolsky E, Vyskocil F. Protein kinase A cascade regulates quantal release dispersion at frog muscle endplate. J Physiol 2002; 538:837-48. [PMID: 11826168 PMCID: PMC2290098 DOI: 10.1113/jphysiol.2001.012752] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2001] [Accepted: 10/15/2001] [Indexed: 11/08/2022] Open
Abstract
Uniquantal endplate currents (EPCs) were recorded simultaneously at the proximal, central and distal parts of the frog neuromuscular synapse, and their minimal synaptic latencies, latency dispersions and sensitivity to noradrenaline, cAMP and protein kinase A inhibition were measured. The latency dispersion was highest in the proximal part (P90 = 1.25 ms); it decreased to P90 = 0.95 ms in the central part and to P90 = 0.75 ms (60 % of the proximal part) in the distal part. In the proximal parts of the long neuromuscular synapse, stimulation-evoked EPCs with long release latencies were eliminated when the intracellular cAMP was increased by beta1 activation by noradrenaline, by the permeable analogue db-cAMP, by activation of adenylyl cyclase or by inhibition of cAMP hydrolysis. This makes the evoked release more compact, and the amplitude of the reconstructed multiquantal currents increases. Protein kinase A is a target of this regulation, since a specific inhibitor, Rp-cAMP, prevents the action of cAMP in the proximal parts and increases the occurrence of long-latency events in the distal parts of the synapse. Our results show that protein kinase A is involved in the timing of quantal release and can be regulated by presynaptic adrenergic receptors.
Collapse
Affiliation(s)
- Ella A Bukharaeva
- State Medical University, Butlerov st. 49, Kazan, Russian Federation and Institute of Biochemistry and Biophysics, Russian Academy of Sciences, PO Box 30, Kazan, Russian Federation
| | | | | | | |
Collapse
|
37
|
Bukharaeva EA, Samigullin DV, Nikol'skii EE, Vyskochil F. The role of intracellular cAMP in mediating the synchronizing action of noradrenaline on the evoked release of quanta of mediator in the frog synapse. NEUROSCIENCE AND BEHAVIORAL PHYSIOLOGY 2001; 31:473-80. [PMID: 11693470 DOI: 10.1023/a:1010414612669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Experiments on frog neuromuscular junction preparations with extracellular recording of action currents in nerve endings and single-quantum currents from endplates were used to assess the time course of evoked quantum mediator secretion by analyzing histograms showing the distribution of true synaptic delays. Studies using the cyclic AMP analog dibutyryl-cAMP (db-cAMP), the adenylate cyclase activator forskolin, and the nucleotide-dependent phosphodiesterase inhibitor isobutylmethylxanthine, showed that these agents, like noradrenaline, altered the kinetics of secretion of quanta, leading to synchronization of the release of mediator. After preliminary treatment of the neuromuscular preparation with db-cAMP, forskolin, or isobutylmethylxanthine, noradrenaline did not induce the synchronization of mediator release in quanta. It was concluded that the action of noradrenaline on the time course of secretion is mediated by activation of presynaptic beta receptors, increased adenylate cyclase activity, and increases in intracellular cAMP levels.
Collapse
Affiliation(s)
- E A Bukharaeva
- Kazan' State Medical University, and Kazan' Institute of Biochemistry and Biophysics, Russia
| | | | | | | |
Collapse
|
38
|
Fink MA, Zakhary DR, Mackey JA, Desnoyer RW, Apperson-Hansen C, Damron DS, Bond M. AKAP-mediated targeting of protein kinase a regulates contractility in cardiac myocytes. Circ Res 2001; 88:291-7. [PMID: 11179196 DOI: 10.1161/01.res.88.3.291] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Compartmentalization of cAMP-dependent protein kinase A (PKA) by A-kinase anchoring proteins (AKAPs) targets PKA to distinct subcellular locations in many cell types. However, the question of whether AKAP-mediated PKA anchoring in the heart regulates cardiac contractile function has not been addressed. We disrupted AKAP-mediated PKA anchoring in cardiac myocytes by introducing, via adenovirus-mediated gene transfer, Ht31, a peptide that binds the PKA regulatory subunit type II (RII) with high affinity. This peptide competes with endogenous AKAPs for RII binding. Ht31P (a proline-substituted derivative), which does not bind RII, was used as a negative control. We then investigated the effects of Ht31 expression on RII distribution, Ca(2+) cycling, cell shortening, and PKA-dependent substrate phosphorylation. By confocal microscopy, we showed redistribution of RII from the perinuclear region and from periodic transverse striations in Ht31P-expressing cells to a diffuse cytosolic localization in Ht31-expressing cells. In the presence of 10 nmol/L isoproterenol, Ht31-expressing myocytes displayed an increased rate and amplitude of cell shortening and relaxation compared with control cells (uninfected and Ht31P-expressing myocytes); with isoproterenol stimulation we observed decreased time to 90% decline in Ca(2+) but no significant difference between Ht31-expressing and control cells in the rate of Ca(2+) cycling or amplitude of the Ca(2+) transient. The increase in PKA-dependent phosphorylation of troponin I and myosin binding protein C on isoproterenol stimulation was significantly reduced in Ht31-expressing cells compared with controls. Our results demonstrate that, in response to beta-adrenergic stimulation, cardiomyocyte function and substrate phosphorylation by PKA is regulated by targeting of PKA by AKAPs.
Collapse
Affiliation(s)
- M A Fink
- Department of Molecular Cardiology, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Brewis N, Ohst K, Fields K, Rapacciuolo A, Chou D, Bloor C, Dillmann W, Rockman H, Walter G. Dilated cardiomyopathy in transgenic mice expressing a mutant A subunit of protein phosphatase 2A. Am J Physiol Heart Circ Physiol 2000; 279:H1307-18. [PMID: 10993798 DOI: 10.1152/ajpheart.2000.279.3.h1307] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The protein phosphatase 2A (PP2A) holoenzyme consists of a catalytic subunit, C, and two regulatory subunits, A and B. The PP2A core enzyme is composed of subunits A and C. Both the holoenzyme and the core enzyme are similarly abundant in heart tissue. Transgenic mice were generated expressing high levels of a dominant negative mutant of the A subunit (A delta 5) in the heart, skeletal muscle, and smooth muscle that competes with the endogenous A subunit for binding the C subunit but does not bind B subunits. We found that the ratio of core enzyme to holoenzyme was increased in A delta 5-expressing hearts. Importantly, already at day 1 after birth, A delta 5-transgenic mice had an increased heart weight-to-body weight ratio that persisted throughout life. Echocardiographic analysis of A delta 5-transgenic hearts revealed increased end-diastolic and end-systolic dimensions and decreased fractional shortening. In addition, the thickness of the septum and of the left ventricular posterior wall was significantly reduced. On the basis of these findings, we consider the heart phenotype of A delta 5-transgenic mice to be a form of dilated cardiomyopathy that frequently leads to premature death.
Collapse
MESH Headings
- Animals
- Body Weight
- Cardiomyopathy, Dilated/enzymology
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/pathology
- Echocardiography
- Exons/genetics
- Gene Expression/genetics
- Genes, Dominant/genetics
- Holoenzymes/genetics
- Holoenzymes/metabolism
- Mice
- Mice, Transgenic/genetics
- Muscle, Skeletal/enzymology
- Muscle, Skeletal/pathology
- Muscle, Smooth/enzymology
- Muscle, Smooth/pathology
- Mutagenesis, Site-Directed
- Myocardium/enzymology
- Myocardium/pathology
- Organ Size
- Organ Specificity/genetics
- Phosphoprotein Phosphatases/genetics
- Phosphoprotein Phosphatases/metabolism
- Protein Binding/genetics
- Protein Phosphatase 2
- Sequence Deletion/genetics
- Transgenes/genetics
Collapse
Affiliation(s)
- N Brewis
- Department of Pathology, University of California San Diego, La Jolla, California 92093, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Cyclic nucleotides in smooth muscle. ACTA ACUST UNITED AC 2000. [DOI: 10.1016/s1569-2590(00)08008-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
41
|
Abstract
Recent studies have added complexities to the conceptual framework of cardiac beta-adrenergic receptor (beta-AR) signal transduction. Whereas the classical linear G(s)-adenylyl cyclase-cAMP-protein kinase A (PKA) signaling cascade has been corroborated for beta(1)-AR stimulation, the beta(2)-AR signaling pathway bifurcates at the very first postreceptor step, the G protein level. In addition to G(s), beta(2)-AR couples to pertussis toxin-sensitive G(i) proteins, G(i2) and G(i3). The coupling of beta(2)-AR to G(i) proteins mediates, to a large extent, the differential actions of the beta-AR subtypes on cardiac Ca(2+) handling, contractility, cAMP accumulation, and PKA-mediated protein phosphorylation. The extent of G(i) coupling in ventricular myocytes appears to be the basis of the substantial species-to-species diversity in beta(2)-AR-mediated cardiac responses. There is an apparent dissociation of beta(2)-AR-induced augmentations of the intracellular Ca(2+) (Ca(i)) transient and contractility from cAMP production and PKA-dependent cytoplasmic protein phosphorylation. This can be largely explained by G(i)-dependent functional compartmentalization of the beta(2)-AR-directed cAMP/PKA signaling to the sarcolemmal microdomain. This compartmentalization allows the common second messenger, cAMP, to perform selective functions during beta-AR subtype stimulation. Emerging evidence also points to distinctly different roles of these beta-AR subtypes in modulating noncontractile cellular processes. These recent findings not only reveal the diversity and specificity of beta-AR and G protein interactions but also provide new insights for understanding the differential regulation and functionality of beta-AR subtypes in healthy and diseased hearts.
Collapse
MESH Headings
- Adrenergic beta-Agonists/pharmacology
- Adrenergic beta-Antagonists/pharmacology
- Animals
- Calcium/physiology
- Cyclic AMP/physiology
- Cyclic AMP-Dependent Protein Kinases/physiology
- Dogs
- GTP-Binding Proteins/physiology
- Heart/drug effects
- Heart/physiology
- Heart Failure/metabolism
- Humans
- Hydrogen-Ion Concentration
- Mice
- Mice, Transgenic
- Muscle Proteins/drug effects
- Muscle Proteins/physiology
- Myocardial Contraction/drug effects
- Myocardial Contraction/physiology
- Myocardium/metabolism
- Phosphorylation/drug effects
- Protein Processing, Post-Translational/drug effects
- Rats
- Receptors, Adrenergic, beta-1/drug effects
- Receptors, Adrenergic, beta-1/physiology
- Receptors, Adrenergic, beta-2/classification
- Receptors, Adrenergic, beta-2/drug effects
- Receptors, Adrenergic, beta-2/physiology
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Species Specificity
Collapse
Affiliation(s)
- R P Xiao
- Laboratory of Cardiovascular Science, Gerontology Research Center, National Institute on Aging, Baltimore, MD 21224, USA.
| | | | | | | | | |
Collapse
|
42
|
Muth JN, Yamaguchi H, Mikala G, Grupp IL, Lewis W, Cheng H, Song LS, Lakatta EG, Varadi G, Schwartz A. Cardiac-specific overexpression of the alpha(1) subunit of the L-type voltage-dependent Ca(2+) channel in transgenic mice. Loss of isoproterenol-induced contraction. J Biol Chem 1999; 274:21503-6. [PMID: 10419451 DOI: 10.1074/jbc.274.31.21503] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The L-type voltage-dependent calcium channel (L-VDCC) regulates calcium influx in cardiac myocytes. Activation of the beta-adrenergic receptor (betaAR) pathway causes phosphorylation of the L-VDCC and that in turn increases Ca(2+) influx. Targeted expression of the L-VDCC alpha(1) subunit in transgenic (Tg) mouse ventricles resulted in marked blunting of the betaAR pathway. Inotropic and lusitropic responses to isoproterenol and forskolin in Tg hearts were significantly reduced. Likewise, Ca(2+) current augmentation induced by iso- proterenol and forskolin was markedly depressed in Tg cardiomyocytes. Despite no change in betaAR number, isoproterenol-stimulated adenylyl cyclase activity was absent in Tg membranes and NaF and forskolin responses were reduced. We postulate an important pathway for regulation of the betaAR by Ca(2+) channels.
Collapse
Affiliation(s)
- J N Muth
- Institute of Molecular Pharmacology and Biophysics, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Weiss HR, Gong GX, Straznicka M, Yan L, Tse J, Scholz PM. Cyclic GMP and cyclic AMP induced changes in control and hypertrophic cardiac myocyte function interact through cyclic GMP affected cyclic-AMP phosphodiesterases. Can J Physiol Pharmacol 1999. [DOI: 10.1139/y99-039] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We tested the hypothesis that the negative functional effects of cyclic GMP (cGMP) would be greater after increasing cyclic AMP (cAMP), because of the action of cGMP-affected cAMP phosphodiesterases in cardiac myocytes and that this effect would be altered in left ventricular hypertrophy (LVH) produced by aortic valve plication. Myocyte shortening data were collected using a video edge detector, and O2 consumption was measured by O2 electrodes during stimulation (5 ms, 1 Hz, in 2 mM Ca2+) from control (n = 7) and LVH (n = 7) dog ventricular myocytes. cAMP and cGMP were determined by a competitive binding assay. cAMP was increased by forskolin and milrinone (10-6 M). cGMP was increased with zaprinast and decreased by 1H-[1,2,4]oxadiazolo[4,3-a]quinoxilin-1-one (ODQ) both at 10-6 and 10-4 M, with and without forskolin or forskolin + milrinone. Zaprinast significantly decreased percent shortening in control (9 ± 1 to 7 ± 1%) and LVH (10 ± 1 to 7 ± 1%) myocytes. It increased cGMP in control (36 ± 5 to 52 ± 7 fmol/105 myocytes) and from the significantly higher baseline value in LVH (71 ± 12 to 104 ± 18 fmol/105 myocytes). ODQ increased myocyte function and decreased cGMP levels in control and LVH myocytes. Forskolin + milrinone increased cAMP levels in control (6 ± 1 to 15 ± 2 pmol/105 myocytes) and LVH (8 ± 1 to 18 ± 2 pmol/105 myocytes) myocytes, as did forskolin alone. They also significantly increased percent shortening. There were significant negative functional effects of zaprinast after forskolin + milrinone in control (15 ± 2 to 9 ± 1%), which were greater than zaprinast alone, and LVH (12 ± 1 to 9 ± 1%). This was associated with an increase in cGMP and a reduction in the increased cAMP induced by forskolin or milrinone. ODQ did not further increase function after forskolin or milrinone in control myocytes, despite lowering cGMP. However, it prevented the forskolin and milrinone induced increase in cAMP. In hypertrophy, ODQ lowered cGMP and increased function after forskolin. ODQ did not affect cAMP after forskolin and milrinone in LVH. Thus, the level of cGMP was inversely correlated with myocyte function. When cAMP levels were elevated, cGMP was still inversely correlated with myocyte function. This was, in part, related to alterations in cAMP. The interaction between cGMP and cAMP was altered in LVH myocytes.Key words: second messengers, cyclic AMP, cyclic GMP, cardiac myocyte function, cyclic GMP dependent cyclic-AMP phosphodiesterases, left ventricular hypertrophy, dog.
Collapse
|
44
|
Kuschel M, Karczewski P, Hempel P, Schlegel WP, Krause EG, Bartel S. Ser16 prevails over Thr17 phospholamban phosphorylation in the beta-adrenergic regulation of cardiac relaxation. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 276:H1625-33. [PMID: 10330247 DOI: 10.1152/ajpheart.1999.276.5.h1625] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Phospholamban is a critical regulator of sarcoplasmic reticulum Ca2+-ATPase and myocardial contractility. To determine the extent of cross signaling between Ca2+ and cAMP pathways, we have investigated the beta-adrenergic-induced phosphorylation of Ser16 and Thr17 of phospholamban in perfused rat hearts using antibodies recognizing phospholamban phosphorylated at either position. Isoproterenol caused the dose-dependent phosphorylation of Ser16 and Thr17 with strikingly different half-maximal values (EC50 = 4.5 +/- 1.6 and 28. 2 +/- 1.4 nmol/l, respectively). The phosphorylation of Ser16 induced by isoproterenol, forskolin, or 3-isobutyl-1-methylxanthine correlated to increased cardiac relaxation (r = 0.96), whereas phosphorylation of Thr17 did not. Elevation of extracellular Ca2+ did not induce phosphorylation at Thr17; only in the presence of a submaximal dose of isoproterenol, phosphorylation at Thr17 increased eightfold without additional effects on relaxation rate. Thr17 phosphorylation was partially affected by ryanodine and was completely abolished in the presence of 1 micromol/l verapamil or nifedipine. The data indicate that 1) phosphorylation of phospholamban at Ser16 by cAMP-dependent protein kinase is the main regulator of beta-adrenergic-induced cardiac relaxation definitely preceding Thr17 phosphorylation and 2) the beta-adrenergic-mediated phosphorylation of Thr17 by Ca2+-calmodulin-dependent protein kinase required influx of Ca2+ through the L-type Ca2+ channel.
Collapse
Affiliation(s)
- M Kuschel
- Max Delbrück Center for Molecular Medicine, 13125 Berlin-Buch, Germany
| | | | | | | | | | | |
Collapse
|
45
|
Kaye DM, Wiviott SD, Kelly RA. Activation of nitric oxide synthase (NOS3) by mechanical activity alters contractile activity in a Ca2+-independent manner in cardiac myocytes: role of troponin I phosphorylation. Biochem Biophys Res Commun 1999; 256:398-403. [PMID: 10079196 DOI: 10.1006/bbrc.1999.0346] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cardiac myocytes express the calcium-responsive nitric oxide synthase (eNOS or NOS3). Activation of NOS3 by increased intracellular Ca2+ concentration, [Ca2+]i, has been demonstrated to decrease myocyte contractile responsiveness, although this appears to occur in a Ca2+-independent manner. Therefore, the aim of this study was to examine the possibility that contractile activity could be modulated by an NO-mediated alteration in the phosphorylation status of troponin I, which is known to alter myofilament sensitivity to Ca2+. During pacing at 3 Hz, 32P-labeled myocytes exhibited a 59 +/- 9% increase in TnI phosphorylation compared to quiescent cells (p < 0.05), an effect that was significantly attenuated by either methylene blue or l-nitroarginine (l-NA). While exposure to methylene blue significantly increased the contractile amplitude of paced myocytes, this was not accompanied by an alteration in intracellular Ca2+. These data indicate that the NO-mediated effects on myocyte contraction may be elicited through an alteration in myofilament Ca2+ sensitivity that results from an alteration in the phosphorylation status of troponin I.
Collapse
Affiliation(s)
- D M Kaye
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | | | |
Collapse
|
46
|
Borthne K, Langslet A, Lindberg H, Osnes JB, Skomedal T. Endogenous muscarinic activity attenuates adrenergic inotropic effects in field stimulated atrial myocardium from children with congenital heart defects. ACTA PHYSIOLOGICA SCANDINAVICA 1999; 165:9-13. [PMID: 10072091 DOI: 10.1046/j.1365-201x.1999.00459.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
To analyse the possible influence of endogenous muscarinic activity on the inotropic effects of endogenously released noradrenaline in field stimulated myocardial preparations from atria of children with congenital heart defects, we studied the maximal effect of the muscarinic antagonist atropine (1.5 micromol L(-1)). Maximal force of contraction increased by 12.8 +/- 2.0% (SEM), while the maximal rate of development of the force increased by 16.7 +/- 2.7% (SEM). Time to half maximal developed force was 57 +/- 5 s (SEM). Time to peak force, time to relax to the 20% level and relaxation time all decreased significantly after atropine. Compared with endogenous adrenoceptor stimulation alone, the combined effects of partial muscarinic and adrenergic receptor stimulation thus were moderate reductions of the maximal force of contraction and maximal rate of development of the force and increased time to peak force, time to relax to the 20% level and relaxation time. The main effect of the endogenous muscarinic activity probably was to attenuate the effect of the beta-adrenoceptor stimulation. The endogenous muscarinic activity in field stimulated atrial preparations from children is significant, and has to be taken into account in experimental set-ups.
Collapse
Affiliation(s)
- K Borthne
- Institute for Surgical Research, Rikshospitalet, Oslo, Norway
| | | | | | | | | |
Collapse
|
47
|
Simmerman HK, Jones LR. Phospholamban: protein structure, mechanism of action, and role in cardiac function. Physiol Rev 1998; 78:921-47. [PMID: 9790566 DOI: 10.1152/physrev.1998.78.4.921] [Citation(s) in RCA: 422] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A comprehensive discussion is presented of advances in understanding the structure and function of phospholamban (PLB), the principal regulator of the Ca2+-ATPase of cardiac sarcoplasmic reticulum. Extensive historical studies are reviewed to provide perspective on recent developments. Phospholamban gene structure, expression, and regulation are presented in addition to in vitro and in vivo studies of PLB protein structure and activity. Applications of breakthrough experimental technologies in identifying PLB structure-function relationships and in defining its interaction with the Ca2+-ATPase are also highlighted. The current leading viewpoint of PLB's mechanism of action emerges from a critical examination of alternative hypotheses and the most recent experimental evidence. The potential physiological relevance of PLB function in human heart failure is also covered. The interest in PLB across diverse biochemical disciplines portends its continued intense scrutiny and its potential exploitation as a therapeutic target.
Collapse
|
48
|
Gupta RC, Neumann J, Watanabe AM, Sabbah HN. Muscarinic-cholinoceptor mediated attenuation of phospholamban phosphorylation induced by inhibition of phosphodiesterase in ventricular cardiomyocytes: evidence against a cAMP-dependent effect. Mol Cell Biochem 1998; 187:155-61. [PMID: 9788753 DOI: 10.1023/a:1006899931151] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
In intact guinea pig ventricles, acetylcholine (ACH) has been shown to attenuate the positive inotropic effects of isobutylmethylxanthine (IBMX), a phosphodiesterase inhibitor, by reducing protein phosphorylation without altering cAMP levels. In the present study, we tested the hypothesis that the cAMP-independent inhibitory action of ACH is also evident in isolated cardiomyocytes. cAMP-dependent protein kinase (PKA) activity ratio (-cAMP/+cAMP) and phosphorylation of phospholamban (PLB) were determined in unlabeled and 32P-labeled guinea pig ventricular cardiomyocytes, respectively. IBMX increased PKA activity ratio and phosphorylation of PLB in a dose-dependent manner. When cardiomyocytes were incubated simultaneously with IBMX (0-1 mM) and ACH (2 microM), ACH attenuated PLB phosphorylation stimulated by low concentration (1O-100 microM) but not by high concentrations (> 200 microM) of IBMX. EC50 value for IBMX-induced phosphorylation of PLB was 32 +/- 6 microM and increased nearly 3-fold after addition of ACH while PKA activity ratio remained unchanged. The rank order of cyclic nucleotide derivatives to phosphorylate PLB was 8 bromo-cAMP > dibutyryl cAMP > 8 bromo-cGMP > dibutyryl cGMP. ACH reduced phosphorylation of PLB stimulated by 8 bromo-cAMP. We conclude that in isolated cardiomyocytes (1) ACH inhibits phosphorylation of PLB stimulated by either IBMX or 8 bromo-cAMP and (2) ACH does not lower IBMX-stimulated PKA activity ratio. These effects of ACH on PLB phosphorylation cannot be explained by a reduction in IBMX-stimulated cAMP levels but may involve the activation of protein phosphatases.
Collapse
Affiliation(s)
- R C Gupta
- Department of Medicine, Henry Ford Heart and Vascular Institute, Detroit, Michigan 48202, USA
| | | | | | | |
Collapse
|
49
|
Abstract
Protein phosphorylation acts a pivotal mechanism in regulating the contractile state of the heart by modulating particular levels of autonomic control on cardiac force/length relationships. Early studies of changes in cardiac protein phosphorylation focused on key components of the excitation-coupling process, namely phospholamban of the sarcoplasmic reticulum and myofibrillar troponin I. In more recent years the emphasis has shifted towards the identification of other phosphoproteins, and more importantly, the delineation of the mechanistic and signaling pathways regulating the various known phosphoproteins. In addition to cAMP- and Ca(2+)-calmodulin-dependent kinase processes, these have included regulation by protein kinase C and the ever-emerging family of growth factor-related kinases such as the tyrosine-, mitogen- and stress-activated protein kinases. Similarly, the role of protein dephosphorylation by protein phosphatases has been recognized as integral in modulating normal cardiac cellular function. Recent studies involving a variety of cardiovascular pathologies have demonstrated that changes in the phosphorylation states of key cardiac regulatory proteins may underlie cardiac dysfunction in disease states. The emphasis of this comprehensive review will be on discussing the role of cardiac phosphoproteins in regulating myocardial function and pathophysiology based not only on in vitro data, but more importantly, from ex vivo experiments with corroborative physiological and biochemical evidence.
Collapse
Affiliation(s)
- S T Rapundalo
- Department of Biochemistry, Parke-Davis Pharmaceutical Research, Division of Warner-Lambert, Ann Arbor, MI 48105, USA.
| |
Collapse
|
50
|
Sulakhe PV, Vo XT, Morris TE, Pato MD, Khandelwal RL. Protein phosphorylation in rat cardiac microsomes: effects of inhibitors of protein kinase A and of phosphatases. Mol Cell Biochem 1997; 175:109-15. [PMID: 9350040 DOI: 10.1023/a:1006879427457] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The phosphorylation of rat cardiac microsomal proteins was investigated with special attention to the effects of okadaic acid (an inhibitor of protein phosphatases), inhibitor 2 of protein phosphatase 1 and inhibitor of cyclic AMP-dependent protein kinase (protein kinase A). The results showed that okadaic acid (5 microM) modestly but reproducibly augmented the protein kinase A-catalyzed phospholamban (PLN) phosphorylation, although exerted little effect on the calcium/calmodulin kinase-catalyzed PLN phosphorylation. Microsomes contained three other substrates (M(r) 23, 19 and 17 kDa) that were phosphorylated by protein kinase A but not by calcium/calmodulin kinase. The protein kinase A-catalyzed phosphorylation of these three substrates was markedly (2-3 fold) increased by 5 microM okadaic acid. Calmodulin was found to antagonize the action of okadaic acid on such phosphorylation. Protein kinase A inhibitor was found to decrease the protein kinase A-catalyzed phosphorylation of microsomal polypeptides. Unexpectedly, inhibitor 2 was also found to markedly decrease protein kinase A-catalyzed phosphorylation of phospholamban as well these other microsomal substrates. These results are consistent with the views that protein phosphatase 1 is capable of dephosphorylating membrane-associated phospholamban when it is phosphorylated by protein kinase A, but not by calcium/calmodulin kinase, and that under certain conditions, calcium/calmodulin-stimulated protein phosphatase (protein phosphatase 2B) is also able to dephosphorylate PLN phosphorylated by protein kinase A. Additionally, the observations show that protein phosphatase 1 is extremely active against the three protein kinase A substrates (M(r) 23, 19 and 17 kDa) that were present in the isolated microsomes and whose state of phosphorylation was particularly affected in the presence of dimethylsulfoxide. Protein phosphatase 2B is also capable of dephosphorylating these three substrates.
Collapse
Affiliation(s)
- P V Sulakhe
- Department of Physiology, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | | | | | | | | |
Collapse
|