51
|
Feric NT, Pallotta I, Singh R, Bogdanowicz DR, Gustilo M, Chaudhary K, Willette RN, Chendrimada T, Xu X, Graziano MP, Aschar-Sobbi R. Engineered Cardiac Tissues Generated in the Biowire™ II: A Platform for Human-Based Drug Discovery. Toxicol Sci 2019; 172:89-97. [PMID: 31385592 PMCID: PMC6813749 DOI: 10.1093/toxsci/kfz168] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/01/2019] [Accepted: 07/14/2019] [Indexed: 01/14/2023] Open
Abstract
Recent advances in techniques to differentiate human induced pluripotent stem cells (hiPSCs) hold the promise of an unlimited supply of human derived cardiac cells from both healthy and disease populations. That promise has been tempered by the observation that hiPSC-derived cardiomyocytes (hiPSC-CMs) typically retain a fetal-like phenotype, raising concern about the translatability of the in vitro data obtained to drug safety, discovery and development studies. The Biowire™ II platform was used to generate 3D engineered cardiac tissues (ECTs) from hiPSC-CMs and cardiac fibroblasts. Long term electrical stimulation was employed to obtain ECTs that possess a phenotype like that of adult human myocardium including a lack of spontaneous beating, the presence of a positive force-frequency response from 1-4Hz and prominent post-rest potentiation. Pharmacology studies were performed in the ECTs to confirm the presence and functionality of pathways that modulate cardiac contractility in humans. Canonical responses were observed for compounds that act via the β-adrenergic/cAMP-mediated pathway, e.g. isoproterenol and milrinone; the L-type calcium channel, e.g. FPL64176 and nifedipine; and indirectly effect intracellular Ca2+ concentrations, e.g. digoxin. Expected positive inotropic responses were observed for compounds that modulate proteins of the cardiac sarcomere, e.g. omecamtiv mecarbil and levosimendan. ECTs generated in the BiowireTM II platform display adult-like properties and have canonical responses to cardiotherapeutic and cardiotoxic agents that affect contractility in humans via a variety of mechanisms. These data demonstrate that this human-based model can be used to assess the effects of novel compounds on contractility early in the drug discovery and development process.
Collapse
|
52
|
Local membrane charge regulates β 2 adrenergic receptor coupling to G i3. Nat Commun 2019; 10:2234. [PMID: 31110175 PMCID: PMC6527575 DOI: 10.1038/s41467-019-10108-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 04/18/2019] [Indexed: 11/13/2022] Open
Abstract
The β2 adrenergic receptor (β2AR) signals through both Gs and Gi in cardiac myocytes, and the Gi pathway counteracts the Gs pathway. However, Gi coupling is much less efficient than Gs coupling in most cell-based and biochemical assays, making it difficult to study β2AR−Gi interactions. Here we investigate the role of phospholipid composition on Gs and Gi coupling. While negatively charged phospholipids are known to enhance agonist affinity and stabilize an active state of the β2AR, we find that they impair coupling to Gi3 and facilitate coupling to Gs. Positively charged Ca2+ and Mg2+, known to interact with the negative charge on phospholipids, facilitates Gi3 coupling. Mutational analysis suggests that Ca2+ coordinates an interaction between phospholipid and the negatively charged EDGE motif on the amino terminal helix of Gi3. Taken together, our observations suggest that local membrane charge modulates the interaction between β2AR and competing G protein subtypes. In the healthy heart, the β2 adrenergic receptor (β2AR) signals through Gs and Gi proteins but the mechanism underlying G protein selectivity is not fully understood. Here, the authors show that membrane charge and intracellular cations modulate the β2AR−Gi3 interaction.
Collapse
|
53
|
Hamilton S, Terentyev D. Altered Intracellular Calcium Homeostasis and Arrhythmogenesis in the Aged Heart. Int J Mol Sci 2019; 20:ijms20102386. [PMID: 31091723 PMCID: PMC6566636 DOI: 10.3390/ijms20102386] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 04/29/2019] [Accepted: 05/08/2019] [Indexed: 12/17/2022] Open
Abstract
Aging of the heart is associated with a blunted response to sympathetic stimulation, reduced contractility, and increased propensity for arrhythmias, with the risk of sudden cardiac death significantly increased in the elderly population. The altered cardiac structural and functional phenotype, as well as age-associated prevalent comorbidities including hypertension and atherosclerosis, predispose the heart to atrial fibrillation, heart failure, and ventricular tachyarrhythmias. At the cellular level, perturbations in mitochondrial function, excitation-contraction coupling, and calcium homeostasis contribute to this electrical and contractile dysfunction. Major determinants of cardiac contractility are the intracellular release of Ca2+ from the sarcoplasmic reticulum by the ryanodine receptors (RyR2), and the following sequestration of Ca2+ by the sarco/endoplasmic Ca2+-ATPase (SERCa2a). Activity of RyR2 and SERCa2a in myocytes is not only dependent on expression levels and interacting accessory proteins, but on fine-tuned regulation via post-translational modifications. In this paper, we review how aberrant changes in intracellular Ca2+ cycling via these proteins contributes to arrhythmogenesis in the aged heart.
Collapse
Affiliation(s)
- Shanna Hamilton
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Dmitry Terentyev
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
54
|
Bacmeister L, Segin S, Medert R, Lindner D, Freichel M, Camacho Londoño JE. Assessment of PEEP-Ventilation and the Time Point of Parallel-Conductance Determination for Pressure-Volume Analysis Under β-Adrenergic Stimulation in Mice. Front Cardiovasc Med 2019; 6:36. [PMID: 31111037 PMCID: PMC6499229 DOI: 10.3389/fcvm.2019.00036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 03/18/2019] [Indexed: 01/08/2023] Open
Abstract
Aim: Cardiac pressure-volume (PV loop) analysis under β-adrenergic stimulation is a powerful method to simultaneously determine intrinsic cardiac function and β-adrenergic reserve in mouse models. Despite its wide use, several key approaches of this method, which can affect murine cardiac function tremendously, have not been experimentally investigated until now. In this study, we investigate the impact of three lines of action during the complex procedure of PV loop analysis: (i) the ventilation with positive end-expiratory pressure, (ii) the time point of injecting hypertonic saline to estimate parallel-conductance, and (iii) the implications of end-systolic pressure-spikes that may arise under β-adrenergic stimulation. Methods and Results: We performed pressure-volume analysis during β-adrenergic stimulation in an open-chest protocol under Isoflurane/Buprenorphine anesthesia. Our analysis showed that (i) ventilation with 2 cmH2O positive end-expiratory pressure prevented exacerbation of peak inspiratory pressures subsequently protecting mice from macroscopic pulmonary bleedings. (ii) Estimations of parallel-conductance by injecting hypertonic saline prior to pressure-volume recordings induced dilated chamber dimensions as depicted by elevation of end-systolic volume (+113%), end-diastolic volume (+40%), and end-diastolic pressure (+46%). Further, using this experimental approach, the preload-independent contractility (PRSW) was significantly impaired under basal conditions (−17%) and under catecholaminergic stimulation (−14% at 8.25 ng/min Isoprenaline), the β-adrenergic reserve was alleviated, and the incidence of ectopic beats was increased >5-fold. (iii) End-systolic pressure-spikes were observed in 26% of pressure-volume recordings under stimulation with 2.475 and 8.25 ng/min Isoprenaline, which affected the analysis of maximum pressure (+11.5%), end-diastolic volume (−8%), stroke volume (−10%), and cardiac output (−11%). Conclusions: Our results (i) demonstrate the advantages of positive end-expiratory pressure ventilation in open-chest instrumented mice, (ii) underline the perils of injecting hypertonic saline prior to pressure-volume recordings to calibrate for parallel-conductance and (iii) emphasize the necessity to be aware of the consequences of end-systolic pressure-spikes during β-adrenergic stimulation.
Collapse
Affiliation(s)
- Lucas Bacmeister
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, Heidelberg, Germany.,Partner Site Heidelberg/Mannheim, DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany
| | - Sebastian Segin
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, Heidelberg, Germany.,Partner Site Heidelberg/Mannheim, DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany
| | - Rebekka Medert
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, Heidelberg, Germany.,Partner Site Heidelberg/Mannheim, DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany
| | - Diana Lindner
- Allgemeine und Interventionelle Kardiologie, Universitäres Herzzentrum Hamburg, Hamburg, Germany.,Partner Site Hamburg/Kiel/Lübeck, DZHK (German Centre for Cardiovascular Research), Hamburg, Germany
| | - Marc Freichel
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, Heidelberg, Germany.,Partner Site Heidelberg/Mannheim, DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany
| | - Juan E Camacho Londoño
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, Heidelberg, Germany.,Partner Site Heidelberg/Mannheim, DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany
| |
Collapse
|
55
|
Lariccia V, Macrì ML, Matteucci A, Maiolino M, Amoroso S, Magi S. Effects of ticagrelor on the sodium/calcium exchanger 1 (NCX1) in cardiac derived H9c2 cells. Eur J Pharmacol 2019; 850:158-166. [PMID: 30721704 DOI: 10.1016/j.ejphar.2019.01.067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 01/08/2019] [Accepted: 01/17/2019] [Indexed: 12/15/2022]
Abstract
Ticagrelor is a direct acting and reversibly binding P2Y12 antagonist approved for the prevention of thromboembolic events. Clinical effects of ticagrelor cannot be simply accounted for by pure platelet inhibition, and off-target mechanisms can potentially play a role. In particular, recent evidence suggests that ticagrelor may also influence heart function and improve the evolution of myocardial ischemic injury by more direct effects on myocytes. The cardiac sodium/calcium exchanger 1 (NCX1) is a critical player in the generation and control of calcium (Ca2+) signals, which orchestrate multiple myocyte activities in health and disease. Altered expression and/or activity of NCX1 can have profound consequences for the function and fate of myocytes. Whether ticagrelor affects cardiac NCX1 has not been investigated yet. To explore this hypothesis, we analyzed the expression, localization and activity of NCX1 in the heart derived H9c2-NCX1 cells following ticagrelor exposure. We found that ticagrelor concentration- and time-dependently reduced the activity of the cardiac NCX1 in H9c2 cells. In particular, the inhibitory effect of ticagrelor on the Ca2+-influx mode of NCX1 was evident within 1 h and further developed after 24 h, when NCX1 activity was suppressed by about 55% in cells treated with 1 μM ticagrelor. Ticagrelor-induced inhibition of exchanger activity was reached at clinically relevant concentrations, without affecting the expression levels and subcellular distribution of NCX1. Collectively, these findings suggest that cardiac NCX1 is a new downstream target of ticagrelor, which may contribute to the therapeutic profile of ticagrelor in clinical practice.
Collapse
Affiliation(s)
- Vincenzo Lariccia
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy.
| | - Maria Loredana Macrì
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| | - Alessandra Matteucci
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| | - Marta Maiolino
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| | - Salvatore Amoroso
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| | - Simona Magi
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| |
Collapse
|
56
|
Hamilton S, Terentyev D. Proarrhythmic Remodeling of Calcium Homeostasis in Cardiac Disease; Implications for Diabetes and Obesity. Front Physiol 2018. [PMID: 30425651 DOI: 10.3389/fphys.2018.01517, 10.3389/fpls.2018.01517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A rapid growth in the incidence of diabetes and obesity has transpired to a major heath issue and economic burden in the postindustrial world, with more than 29 million patients affected in the United States alone. Cardiovascular defects have been established as the leading cause of mortality and morbidity of diabetic patients. Over the last decade, significant progress has been made in delineating mechanisms responsible for the diminished cardiac contractile function and enhanced propensity for malignant cardiac arrhythmias characteristic of diabetic disease. Rhythmic cardiac contractility relies upon the precise interplay between several cellular Ca2+ transport protein complexes including plasmalemmal L-type Ca2+ channels (LTCC), Na+-Ca2+ exchanger (NCX1), Sarco/endoplasmic Reticulum (SR) Ca2+-ATPase (SERCa2a) and ryanodine receptors (RyR2s), the SR Ca2+ release channels. Here we provide an overview of changes in Ca2+ homeostasis in diabetic ventricular myocytes and discuss the therapeutic potential of targeting Ca2+ handling proteins in the prevention of diabetes-associated cardiomyopathy and arrhythmogenesis.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States.,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States
| | - Dmitry Terentyev
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States.,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States
| |
Collapse
|
57
|
Hamilton S, Terentyev D. Proarrhythmic Remodeling of Calcium Homeostasis in Cardiac Disease; Implications for Diabetes and Obesity. Front Physiol 2018; 9:1517. [PMID: 30425651 PMCID: PMC6218530 DOI: 10.3389/fphys.2018.01517] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/09/2018] [Indexed: 12/28/2022] Open
Abstract
A rapid growth in the incidence of diabetes and obesity has transpired to a major heath issue and economic burden in the postindustrial world, with more than 29 million patients affected in the United States alone. Cardiovascular defects have been established as the leading cause of mortality and morbidity of diabetic patients. Over the last decade, significant progress has been made in delineating mechanisms responsible for the diminished cardiac contractile function and enhanced propensity for malignant cardiac arrhythmias characteristic of diabetic disease. Rhythmic cardiac contractility relies upon the precise interplay between several cellular Ca2+ transport protein complexes including plasmalemmal L-type Ca2+ channels (LTCC), Na+-Ca2+ exchanger (NCX1), Sarco/endoplasmic Reticulum (SR) Ca2+-ATPase (SERCa2a) and ryanodine receptors (RyR2s), the SR Ca2+ release channels. Here we provide an overview of changes in Ca2+ homeostasis in diabetic ventricular myocytes and discuss the therapeutic potential of targeting Ca2+ handling proteins in the prevention of diabetes-associated cardiomyopathy and arrhythmogenesis.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States.,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States
| | - Dmitry Terentyev
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States.,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States
| |
Collapse
|
58
|
Hamilton S, Terentyev D. Proarrhythmic Remodeling of Calcium Homeostasis in Cardiac Disease; Implications for Diabetes and Obesity. Front Physiol 2018; 9:1517. [PMID: 30425651 PMCID: PMC6218530 DOI: 10.3389/fphys.2018.01517,+10.3389/fpls.2018.01517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022] Open
Abstract
A rapid growth in the incidence of diabetes and obesity has transpired to a major heath issue and economic burden in the postindustrial world, with more than 29 million patients affected in the United States alone. Cardiovascular defects have been established as the leading cause of mortality and morbidity of diabetic patients. Over the last decade, significant progress has been made in delineating mechanisms responsible for the diminished cardiac contractile function and enhanced propensity for malignant cardiac arrhythmias characteristic of diabetic disease. Rhythmic cardiac contractility relies upon the precise interplay between several cellular Ca2+ transport protein complexes including plasmalemmal L-type Ca2+ channels (LTCC), Na+-Ca2+ exchanger (NCX1), Sarco/endoplasmic Reticulum (SR) Ca2+-ATPase (SERCa2a) and ryanodine receptors (RyR2s), the SR Ca2+ release channels. Here we provide an overview of changes in Ca2+ homeostasis in diabetic ventricular myocytes and discuss the therapeutic potential of targeting Ca2+ handling proteins in the prevention of diabetes-associated cardiomyopathy and arrhythmogenesis.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States
| | - Dmitry Terentyev
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States,*Correspondence: Dmitry Terentyev,
| |
Collapse
|
59
|
Doliba NM, Babsky AM, Osbakken MD. The Role of Sodium in Diabetic Cardiomyopathy. Front Physiol 2018; 9:1473. [PMID: 30405433 PMCID: PMC6207851 DOI: 10.3389/fphys.2018.01473] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 09/28/2018] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular complications are the major cause of mortality and morbidity in diabetic patients. The changes in myocardial structure and function associated with diabetes are collectively called diabetic cardiomyopathy. Numerous molecular mechanisms have been proposed that could contribute to the development of diabetic cardiomyopathy and have been studied in various animal models of type 1 or type 2 diabetes. The current review focuses on the role of sodium (Na+) in diabetic cardiomyopathy and provides unique data on the linkage between Na+ flux and energy metabolism, studied with non-invasive 23Na, and 31P-NMR spectroscopy, polarography, and mass spectroscopy. 23Na NMR studies allow determination of the intracellular and extracellular Na+ pools by splitting the total Na+ peak into two resonances after the addition of a shift reagent to the perfusate. Using this technology, we found that intracellular Na+ is approximately two times higher in diabetic cardiomyocytes than in control possibly due to combined changes in the activity of Na+–K+ pump, Na+/H+ exchanger 1 (NHE1) and Na+-glucose cotransporter. We hypothesized that the increase in Na+ activates the mitochondrial membrane Na+/Ca2+ exchanger, which leads to a loss of intramitochondrial Ca2+, with a subsequent alteration in mitochondrial bioenergetics and function. Using isolated mitochondria, we showed that the addition of Na+ (1–10 mM) led to a dose-dependent decrease in oxidative phosphorylation and that this effect was reversed by providing extramitochondrial Ca2+ or by inhibiting the mitochondrial Na+/Ca2+ exchanger with diltiazem. Similar experiments with 31P-NMR in isolated superfused mitochondria embedded in agarose beads showed that Na+ (3–30 mM) led to significantly decreased ATP levels and that this effect was stronger in diabetic rats. These data suggest that in diabetic cardiomyocytes, increased Na+ leads to abnormalities in oxidative phosphorylation and a subsequent decrease in ATP levels. In support of these data, using 31P-NMR, we showed that the baseline β-ATP and phosphocreatine (PCr) were lower in diabetic cardiomyocytes than in control, suggesting that diabetic cardiomyocytes have depressed bioenergetic function. Thus, both altered intracellular Na+ levels and bioenergetics and their interactions may significantly contribute to the pathology of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Nicolai M Doliba
- Department of Biochemistry and Biophysics, Institute for Diabetes, Obesity and Metabolism, School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Andriy M Babsky
- Department of Biophysics and Bioinformatics, Ivan Franko National University of Lviv, Lviv, Ukraine
| | - Mary D Osbakken
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, United States
| |
Collapse
|
60
|
Monasky MM, Pappone C, Piccoli M, Ghiroldi A, Micaglio E, Anastasia L. Calcium in Brugada Syndrome: Questions for Future Research. Front Physiol 2018; 9:1088. [PMID: 30147658 PMCID: PMC6095984 DOI: 10.3389/fphys.2018.01088] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/23/2018] [Indexed: 12/11/2022] Open
Abstract
The Brugada syndrome (BrS) is characterized by coved-type ST-segment elevation in the right precordial leads on the electrocardiogram (ECG) and increased risk of sudden cardiac death (SCD). While it is an inheritable disease, determining the true prevalence is a challenge, since patients may report no known family history of the syndrome, present with a normal spontaneous ECG pattern at the time of examination, and test negative for all known BrS-causative genes. In fact, SCD is often the first indication that a person is affected by the syndrome. Men are more likely to be symptomatic than women. Abnormal, low-voltage, fractionated electrograms have been found in the epicardium of the right ventricular outflow tract (RVOT). Ablation of this area abolishes the abnormal electrograms and helps to prevent arrhythmic recurrences. BrS patients are more likely to experience ventricular tachycardia/fibrillation (VT/VF) during fever or during an increase in vagal tone. Isoproterenol helps to reverse the ECG BrS phenotype. In this review, we discuss roles of calcium in various conditions that are relevant to BrS, such as changes in temperature, heart rate, and vagal tone, and the effects of gender and isoproterenol on calcium handling. Studies are warranted to further investigate these mechanisms in models of BrS.
Collapse
Affiliation(s)
| | - Carlo Pappone
- Arrhythmology Department, IRCCS Policlinico San Donato, Milan, Italy
| | - Marco Piccoli
- Stem Cells for Tissue Engineering Lab, IRCCS Policlinico San Donato, Milan, Italy
| | - Andrea Ghiroldi
- Stem Cells for Tissue Engineering Lab, IRCCS Policlinico San Donato, Milan, Italy
| | - Emanuele Micaglio
- Arrhythmology Department, IRCCS Policlinico San Donato, Milan, Italy
| | - Luigi Anastasia
- Stem Cells for Tissue Engineering Lab, IRCCS Policlinico San Donato, Milan, Italy.,Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| |
Collapse
|
61
|
Bögeholz N, Pauls P, Bauer BK, Schulte JS, Frommeyer G, Dechering DG, Boknik P, Kirchhefer U, Müller FU, Pott C, Eckardt L. Overexpression of the Na + /Ca 2+ exchanger influences ouabain-mediated spontaneous Ca 2+ activity but not positive inotropy. Fundam Clin Pharmacol 2018; 33:43-51. [PMID: 30092622 DOI: 10.1111/fcp.12404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 07/16/2018] [Accepted: 08/01/2018] [Indexed: 12/01/2022]
Abstract
Administration of digitalis in heart failure (HF) increases quality of life but does not carry a prognostic benefit. Digitalis is an indirect inhibitor of the Na+ /Ca2+ exchanger (NCX), which is overexpressed in HF. We therefore used the cardiac glycoside ouabain in Ca2+ imaging experiments and patch-clamp experiments in isolated ventricular myocytes from nonfailing transgenic NCX overexpressor mice (OE). In field-stimulated myocytes, ouabain (1-100 μm) increased the amplitude of the Ca2+ transient in OE and wild-type (WT) similarly. Ouabain-mediated spontaneous Ca2+ -activity was significantly more pronounced in OE compared to WT myocytes at higher concentrations (100 μm). Also, at very high concentrations (1000 μm) of ouabain, the number of cells with hypercontraction leading to cell death was higher in OE. Ouabain (10 μm) shortened the action potential duration in both genotypes. Our findings suggest that the proarrhythmic but not the inotropic effects of cardiac glycosides are enhanced by increased NCX expression. This may offer an explanation for the observed lack of prognostic benefit but increased quality of life in HF, which is accompanied by NCX upregulation.
Collapse
Affiliation(s)
- Nils Bögeholz
- Klinik für Kardiologie II: Rhythmologie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149, Münster, Germany
| | - Paul Pauls
- Klinik für Kardiologie II: Rhythmologie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149, Münster, Germany.,Institut für Pharmakologie und Toxikologie, Universität Münster, Domagkstraße 12, 48149, Münster, Germany
| | - Bastian K Bauer
- Klinik für Kardiologie II: Rhythmologie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149, Münster, Germany
| | - Jan S Schulte
- Institut für Pharmakologie und Toxikologie, Universität Münster, Domagkstraße 12, 48149, Münster, Germany
| | - Gerrit Frommeyer
- Klinik für Kardiologie II: Rhythmologie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149, Münster, Germany
| | - Dirk G Dechering
- Klinik für Kardiologie II: Rhythmologie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149, Münster, Germany
| | - Peter Boknik
- Institut für Pharmakologie und Toxikologie, Universität Münster, Domagkstraße 12, 48149, Münster, Germany
| | - Uwe Kirchhefer
- Institut für Pharmakologie und Toxikologie, Universität Münster, Domagkstraße 12, 48149, Münster, Germany
| | - Frank U Müller
- Institut für Pharmakologie und Toxikologie, Universität Münster, Domagkstraße 12, 48149, Münster, Germany
| | - Christian Pott
- Department of Cardiology, Schüchtermann-Klinik, Ulmenallee 5-11, 49214, Bad Rothenfelde, Germany
| | - Lars Eckardt
- Klinik für Kardiologie II: Rhythmologie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149, Münster, Germany
| |
Collapse
|
62
|
Caluori G, Pribyl J, Cmiel V, Pesl M, Potocnak T, Provaznik I, Skladal P, Rotrekl V. Simultaneous study of mechanobiology and calcium dynamics on hESC-derived cardiomyocytes clusters. J Mol Recognit 2018; 32:e2760. [PMID: 30084213 DOI: 10.1002/jmr.2760] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 06/11/2018] [Accepted: 07/07/2018] [Indexed: 12/19/2022]
Abstract
Calcium ions act like ubiquitous second messengers in a wide amount of cellular processes. In cardiac myocytes, Ca2+ handling regulates the mechanical contraction necessary to the heart pump function. The field of intracellular and intercellular Ca2+ handling, employing in vitro models of cardiomyocytes, has become a cornerstone to understand the role and adaptation of calcium signalling in healthy and diseased hearts. Comprehensive in vitro systems and cell-based biosensors are powerful tools to enrich and speed up cardiac phenotypic and drug response evaluation. We have implemented a combined setup to measure contractility and calcium waves in human embryonic stem cells-derived cardiomyocyte 3D clusters, obtained from embryoid body differentiation. A combination of atomic force microscopy to monitor cardiac contractility, and sensitive fast scientific complementary metal-oxide-semiconductor camera for epifluorescence video recording, provided correlated signals in real time. To speed up the integrated data processing, we tested several post-processing algorithms, to improve the automatic detection of relevant functional parameters. The validation of our proposed method was assessed by caffeine stimulation (10mM) and detection/characterization of the induced cardiac response. We successfully report the first simultaneous recording of cardiac contractility and calcium waves on the described cardiac 3D models. The drug stimulation confirmed the automatic detection capabilities of the used algorithms, measuring expected physiological response, such as elongation of contraction time and Ca2+ cytosolic persistence, increased calcium basal fluorescence, and transient peaks. These results contribute to the implementation of novel, integrated, high-information, and reliable experimental systems for cardiac models and drug evaluation.
Collapse
Affiliation(s)
- Guido Caluori
- International Clinical Research Centre of Saint Anne Hospital of Brno (FNUSA-ICRC), Interventional Cardiac Electrophysiology Group, Brno, Czech Republic.,Nanobiotechnology Group, Central European Institute of Technology of Masaryk University (CEITEC-MU), Brno, Czech Republic
| | - Jan Pribyl
- Nanobiotechnology Group, Central European Institute of Technology of Masaryk University (CEITEC-MU), Brno, Czech Republic
| | - Vratislav Cmiel
- Department of Biomedical Engineering, Brno University of Technology, Faculty of Electrical Engineering and Communication, Brno, Czech Republic
| | - Martin Pesl
- International Clinical Research Centre of Saint Anne Hospital of Brno (FNUSA-ICRC), Interventional Cardiac Electrophysiology Group, Brno, Czech Republic.,Department of Biology, Masaryk University, Faculty of Medicine, Brno, Czech Republic
| | - Tomas Potocnak
- Department of Biomedical Engineering, Brno University of Technology, Faculty of Electrical Engineering and Communication, Brno, Czech Republic
| | - Ivo Provaznik
- Department of Biomedical Engineering, Brno University of Technology, Faculty of Electrical Engineering and Communication, Brno, Czech Republic
| | - Petr Skladal
- Nanobiotechnology Group, Central European Institute of Technology of Masaryk University (CEITEC-MU), Brno, Czech Republic
| | - Vladimir Rotrekl
- Department of Biology, Masaryk University, Faculty of Medicine, Brno, Czech Republic
| |
Collapse
|
63
|
Role of the TRPM4 Channel in Cardiovascular Physiology and Pathophysiology. Cells 2018; 7:cells7060062. [PMID: 29914130 PMCID: PMC6025450 DOI: 10.3390/cells7060062] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/11/2018] [Accepted: 06/14/2018] [Indexed: 12/31/2022] Open
Abstract
The transient receptor potential cation channel subfamily M member 4 (TRPM4) channel influences calcium homeostasis during many physiological activities such as insulin secretion, immune response, respiratory reaction, and cerebral vasoconstriction. This calcium-activated, monovalent, selective cation channel also plays a key role in cardiovascular pathophysiology; for example, a mutation in the TRPM4 channel leads to cardiac conduction disease. Recently, it has been suggested that the TRPM4 channel is also involved in the development of cardiac ischemia-reperfusion injury, which causes myocardial infarction. In the present review, we discuss the physiological function of the TRPM4 channel, and assess its role in cardiovascular pathophysiology.
Collapse
|
64
|
Zhai Y, Luo Y, Wu P, Li D. New insights into SERCA2a gene therapy in heart failure: pay attention to the negative effects of B-type natriuretic peptides. J Med Genet 2018; 55:287-296. [PMID: 29478009 DOI: 10.1136/jmedgenet-2017-105120] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/30/2018] [Accepted: 02/05/2018] [Indexed: 12/28/2022]
Abstract
Sarcoplasmic/endoplasmic reticulum calcium ATPase 2a (SERCA2a) is a target of interest in gene therapy for heart failure with reduced ejection fraction (HFrEF). However, the results of an important clinical study, the Calcium Upregulation by Percutaneous Administration of Gene Therapy in Cardiac Disease (CUPID) trial, were controversial. Promising results were observed in the CUPID 1 trial, but the results of the CUPID 2 trial were negative. The factors that caused the controversial results remain unclear. Importantly, enrolled patients were required to have a higher plasma level of B-type natriuretic peptide (BNP) in the CUPID 2 trial. Moreover, BNP was shown to inhibit SERCA2a expression. Therefore, it is possible that high BNP levels interact with treatment effects of SERCA2a gene transfer and accordingly lead to negative results of CUPID 2 trial. From this point of view, effects of SERCA2a gene therapy should be explored in heart failure with preserved ejection fraction, which is characterised by lower BNP levels compared with HFrEF. In this review, we summarise the current knowledge of SERCA2a gene therapy for heart failure, analyse potential interaction between BNP levels and therapeutic effects of SERCA2a gene transfer and provide directions for future research to solve the identified problems.
Collapse
Affiliation(s)
- Yuting Zhai
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuanyuan Luo
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Pei Wu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
65
|
Kubasov IV, Stepanov A, Bobkov D, Radwanski PB, Terpilowski MA, Dobretsov M, Gyorke S. Sub-cellular Electrical Heterogeneity Revealed by Loose Patch Recording Reflects Differential Localization of Sarcolemmal Ion Channels in Intact Rat Hearts. Front Physiol 2018; 9:61. [PMID: 29487533 PMCID: PMC5816904 DOI: 10.3389/fphys.2018.00061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 01/18/2018] [Indexed: 12/31/2022] Open
Abstract
The cardiac action potential (AP) is commonly recoded as an integral signal from isolated myocytes or ensembles of myocytes (with intracellular microelectrodes and extracellular macroelectrodes, respectively). These signals, however, do not provide a direct measure of activity of ion channels and transporters located in two major compartments of a cardiac myocyte: surface sarcolemma and the T-tubule system, which differentially contribute to impulse propagation and excitation-contraction (EC) coupling. In the present study we investigated electrical properties of myocytes within perfused intact rat heart employing loose patch recording with narrow-tip (2 μm diameter) extracellular electrodes. Using this approach, we demonstrated two distinct types of electric signals with distinct waveforms (single peak and multi-peak AP; AP1 and AP2, respectively) during intrinsic pacemaker activity. These two types of waveforms depend on the position of the electrode tip on the myocyte surface. Such heterogeneity of electrical signals was lost when electrodes of larger pipette diameter were used (5 or 10 μm), which indicates that the electric signal was assessed from a region of <5 μm. Importantly, both pharmacological and mathematical simulation based on transverse (T)-tubular distribution suggested that while the AP1 and the initial peak of AP2 are predominantly attributable to the fast, inward Na+ current in myocyte's surface sarcolemma, the late components of AP2 are likely representative of currents associated with L-type Ca2+ channel and Na+/Ca2+ exchanger (NCX) currents which are predominantly located in T-tubules. Thus, loose patch recording with narrow-tip pipette provides a valuable tool for studying cardiac electric activity on the subcellular level in the intact heart.
Collapse
Affiliation(s)
- Igor V. Kubasov
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, Saint-Petersburg, Russia
| | - Andrei Stepanov
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, Saint-Petersburg, Russia
- Institute of Cytology RAS, Saint-Petersburg, Russia
| | - Danila Bobkov
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, Saint-Petersburg, Russia
- Institute of Cytology RAS, Saint-Petersburg, Russia
| | - Przemysław B. Radwanski
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, Ohio State University, Columbus, OH, United States
- Division of Pharmacy Practice and Science, College of Pharmacy, Ohio State University, Columbus, OH, United States
| | - Maxim A. Terpilowski
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, Saint-Petersburg, Russia
| | - Maxim Dobretsov
- Department of Anesthesiology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Sandor Gyorke
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, Ohio State University, Columbus, OH, United States
| |
Collapse
|
66
|
John S, Kim B, Olcese R, Goldhaber JI, Ottolia M. Molecular determinants of pH regulation in the cardiac Na +-Ca 2+ exchanger. J Gen Physiol 2018; 150:245-257. [PMID: 29301861 PMCID: PMC5806679 DOI: 10.1085/jgp.201611693] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 05/25/2017] [Accepted: 11/29/2017] [Indexed: 11/20/2022] Open
Abstract
The cardiac Na+-Ca2+ exchanger (NCX) plays a critical role in the heart by extruding Ca2+ after each contraction and thus regulates cardiac contractility. The activity of NCX is strongly inhibited by cytosolic protons, which suggests that intracellular acidification will have important effects on heart contractility. However, the mechanisms underlying this inhibition remain elusive. It has been suggested that pH regulation originates from the competitive binding of protons to two Ca2+-binding domains within the large cytoplasmic loop of NCX and requires inactivation by intracellular Na+ to fully develop. By combining mutagenesis and electrophysiology, we demonstrate that NCX pH modulation is an allosteric mechanism distinct from Na+ and Ca2+ regulation, and we show that cytoplasmic Na+ can affect the sensitivity of NCX to protons. We further identify two histidines (His 124 and His 165) that are important for NCX proton sensitivity and show that His 165 plays the dominant role. Our results reveal a complex interplay between the different allosteric mechanisms that regulate the activity of NCX. Because of the central role of NCX in cardiac function, these findings are important for our understanding of heart pathophysiology.
Collapse
Affiliation(s)
- Scott John
- Department of Medicine and Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Brian Kim
- Cedars-Sinai Heart Institute, Los Angeles, CA
| | - Riccardo Olcese
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA.,Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Joshua I Goldhaber
- Cedars-Sinai Heart Institute, Los Angeles, CA.,Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Michela Ottolia
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| |
Collapse
|
67
|
Barthmes M, Liao J, Jiang Y, Brüggemann A, Wahl-Schott C. Electrophysiological characterization of the archaeal transporter NCX_Mj using solid supported membrane technology. J Gen Physiol 2017; 147:485-96. [PMID: 27241699 PMCID: PMC4886279 DOI: 10.1085/jgp.201611587] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 05/11/2016] [Indexed: 01/24/2023] Open
Abstract
NCX_Mj is a sodium–calcium exchanger from the archaebacterium Methanococcus jannaschii, whose crystal structure has been solved. Barthmes et al. use solid supported membrane–based electrophysiology to characterize NCX_Mj and reveal its functional similarity to eukaryotic isoforms. Sodium–calcium exchangers (NCXs) are membrane transporters that play an important role in Ca2+ homeostasis and Ca2+ signaling. The recent crystal structure of NCX_Mj, a member of the NCX family from the archaebacterium Methanococcus jannaschii, provided insight into the atomistic details of sodium–calcium exchange. Here, we extend these findings by providing detailed functional data on purified NCX_Mj using solid supported membrane (SSM)–based electrophysiology, a powerful but unexploited tool for functional studies of electrogenic transporter proteins. We show that NCX_Mj is highly selective for Na+, whereas Ca2+ can be replaced by Mg2+ and Sr2+ and that NCX_Mj can be inhibited by divalent ions, particularly Cd2+. By directly comparing the apparent affinities of Na+ and Ca2+ for NCX_Mj with those for human NCX1, we show excellent agreement, indicating a strong functional similarity between NCX_Mj and its eukaryotic isoforms. We also provide detailed instructions to facilitate the adaption of this method to other electrogenic transporter proteins. Our findings demonstrate that NCX_Mj can serve as a model for the NCX family and highlight several possible applications for SSM-based electrophysiology.
Collapse
Affiliation(s)
- Maria Barthmes
- Nanion Technologies, 80636 Munich, Germany Center for Integrated Protein Science (CIPS-M) and Center for Drug Research, Department of Pharmacology, Ludwig Maximilians University and DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, 81377 Munich, Germany
| | - Jun Liao
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390 School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
| | - Youxing Jiang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390 Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | | | - Christian Wahl-Schott
- Center for Integrated Protein Science (CIPS-M) and Center for Drug Research, Department of Pharmacology, Ludwig Maximilians University and DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, 81377 Munich, Germany
| |
Collapse
|
68
|
Shimizu H, Langenbacher AD, Huang J, Wang K, Otto G, Geisler R, Wang Y, Chen JN. The Calcineurin-FoxO-MuRF1 signaling pathway regulates myofibril integrity in cardiomyocytes. eLife 2017; 6:27955. [PMID: 28826496 PMCID: PMC5576919 DOI: 10.7554/elife.27955] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 08/18/2017] [Indexed: 12/21/2022] Open
Abstract
Altered Ca2+ handling is often present in diseased hearts undergoing structural remodeling and functional deterioration. However, whether Ca2+ directly regulates sarcomere structure has remained elusive. Using a zebrafish ncx1 mutant, we explored the impacts of impaired Ca2+ homeostasis on myofibril integrity. We found that the E3 ubiquitin ligase murf1 is upregulated in ncx1-deficient hearts. Intriguingly, knocking down murf1 activity or inhibiting proteasome activity preserved myofibril integrity, revealing a MuRF1-mediated proteasome degradation mechanism that is activated in response to abnormal Ca2+ homeostasis. Furthermore, we detected an accumulation of the murf1 regulator FoxO in the nuclei of ncx1-deficient cardiomyocytes. Overexpression of FoxO in wild type cardiomyocytes induced murf1 expression and caused myofibril disarray, whereas inhibiting Calcineurin activity attenuated FoxO-mediated murf1 expression and protected sarcomeres from degradation in ncx1-deficient hearts. Together, our findings reveal a novel mechanism by which Ca2+ overload disrupts myofibril integrity by activating a Calcineurin-FoxO-MuRF1-proteosome signaling pathway.
Collapse
Affiliation(s)
- Hirohito Shimizu
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, United States
| | - Adam D Langenbacher
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, United States
| | - Jie Huang
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, United States
| | - Kevin Wang
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, United States
| | - Georg Otto
- Genetics and Genomic Medicine, UCL Institute of Child Health, London, United Kingdom
| | - Robert Geisler
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Yibin Wang
- Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States.,Department of Medicine and Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Jau-Nian Chen
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, United States
| |
Collapse
|
69
|
Zhang C, Chen B, Wang Y, Guo A, Tang Y, Khataei T, Shi Y, Kutschke WJ, Zimmerman K, Weiss RM, Liu J, Benson CJ, Hong J, Ma J, Song LS. MG53 is dispensable for T-tubule maturation but critical for maintaining T-tubule integrity following cardiac stress. J Mol Cell Cardiol 2017; 112:123-130. [PMID: 28822805 DOI: 10.1016/j.yjmcc.2017.08.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 08/13/2017] [Accepted: 08/14/2017] [Indexed: 01/28/2023]
Abstract
The cardiac transverse (T)-tubule membrane system is the safeguard for cardiac function and undergoes dramatic remodeling in response to cardiac stress. However, the mechanism by which cardiomyocytes repair damaged T-tubule network remains unclear. In the present study, we tested the hypothesis that MG53, a muscle-specific membrane repair protein, antagonizes T-tubule damage to protect against maladaptive remodeling and thereby loss of excitation-contraction coupling and cardiac function. Using MG53-knockout (MG53-KO) mice, we first established that deficiency of MG53 had no impact on maturation of the T-tubule network in developing hearts. Additionally, MG53 ablation did not influence T-tubule integrity in unstressed adult hearts as late as 10months of age. Following left ventricular pressure overload-induced cardiac stress, MG53 protein levels were increased by approximately three-fold in wild-type mice, indicating that pathological stress induces a significant upregulation of MG53. MG53-deficient mice had worsened T-tubule disruption and pronounced dysregulation of Ca2+ handling properties, including decreased Ca2+ transient amplitude and prolonged time to peak and decay. Moreover, MG53 deficiency exacerbated cardiac hypertrophy and dysfunction and decreased survival following cardiac stress. Our data suggest MG53 is not required for T-tubule development and maintenance in normal physiology. However, MG53 is essential to preserve T-tubule integrity and thereby Ca2+ handling properties and cardiac function under pathological cardiac stress.
Collapse
Affiliation(s)
- Caimei Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Biyi Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Yihui Wang
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Shanghai First People's Hospital, Shanghai Jiaotong University, Shanghai 200080, China
| | - Ang Guo
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Yiqun Tang
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Department of Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Tahsin Khataei
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Yun Shi
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - William J Kutschke
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Kathy Zimmerman
- Department of Veterans Affairs Medical Center, Iowa City, IA 52242, USA
| | - Robert M Weiss
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Jie Liu
- Department of Pathophysiology, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Christopher J Benson
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Department of Veterans Affairs Medical Center, Iowa City, IA 52242, USA
| | - Jiang Hong
- Shanghai First People's Hospital, Shanghai Jiaotong University, Shanghai 200080, China
| | - Jianjie Ma
- Department of Surgery, Ohio State University Medical Center, Columbus, OH 43212, USA
| | - Long-Sheng Song
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Department of Veterans Affairs Medical Center, Iowa City, IA 52242, USA.
| |
Collapse
|
70
|
Cheng D, Wang R, Wang C, Hou L. Mung bean (Phaseolus radiatus L.) polyphenol extract attenuates aluminum-induced cardiotoxicity through an ROS-triggered Ca 2+/JNK/NF-κB signaling pathway in rats. Food Funct 2017; 8:851-859. [PMID: 28128384 DOI: 10.1039/c6fo01817c] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Aluminum (Al) has been linked to the development of some cardiovascular diseases and mung bean is a functional food with the ability to detoxify. We aimed to evaluate the preventive effect and possible underlying mechanisms of the mung bean polyphenol extract (MPE) on Al-induced cardiotoxicity. Control, AlCl3 (171.8 mg Al per kg body weight), MPE + AlCl3 (Al-treatment plus 200 mg MPE per kg body weight), and a group of MPE per se were used. Al intake induced a significant increase of serum CK and LDH activity and the level of Na+, Ca2+, malondialdehyde and advanced oxidation protein products in the AlCl3-treated rats' heart tissue. Administration of MPE significantly improved the integrity and normal ion levels of heart tissue, and attenuated oxidative damage and the accumulation of Al in Al-treated rats. MPE significantly inhibited Al-induced increase of myocardial p-JNK, cytoplasmic NF-κB, cytochrome C, and caspase-9 protein expressions. Therefore, these results showed that MPE has a cardiac protective effect against Al-induced biotoxicity through ROS-JNK and NF-κB-mediated caspase pathways. Furthermore, the stability constant for the vitexin-Al complex was analyzed (log K = log K1 + log K2 = 4.91 + 4.85 = 9.76). We found that MPE-mediated protection against Al-cardiotoxicity is connected both with MPE antioxidant and chelation properties.
Collapse
Affiliation(s)
- Dai Cheng
- Key Laboratory of Food Safety and Sanitation, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin, People's Republic of China.
| | - Ruhua Wang
- Key Laboratory of Food Safety and Sanitation, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin, People's Republic of China.
| | - Chunling Wang
- Key Laboratory of Food Safety and Sanitation, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin, People's Republic of China.
| | - Lihua Hou
- Key Laboratory of Food Safety and Sanitation, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin, People's Republic of China.
| |
Collapse
|
71
|
Ding L, Su XX, Zhang WH, Xu YX, Pan XF. Gene Expressions Underlying Mishandled Calcium Clearance and Elevated Generation of Reactive Oxygen Species in the Coronary Artery Smooth Muscle Cells of Chronic Heart Failure Rats. Chin Med J (Engl) 2017; 130:460-469. [PMID: 28218221 PMCID: PMC5324384 DOI: 10.4103/0366-6999.199825] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The calcium clearance and reactive oxygen species (ROS) generations in the coronary artery smooth muscle cells in chronic heart failure (HF) have not been fully investigated. Therefore, we attempted to understand the gene expressions underlying the mishandling of calcium clearance and the accumulations of ROS. METHODS We initially established an animal model of chronic HF by making the left anterior descending coronary artery ligation (CAL) in rats, and then isolated the coronary artery vascular smooth muscle cells from the ischemic and the nonischemic parts of the coronary artery vessels in 12 weeks after CAL operation. The intracellular calcium concentration and ROS level were measured using flow cytometry, and the gene expressions of sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2a), encoding sarcoplasmic reticulum Ca2+-ATPase 2a, encoding sodium-calcium exchanger (NCX), and p47phox encoding a subunit of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase were examined using real-time quantitative reverse transcription polymerase chain reaction and Western blotting, respectively. RESULTS We found that the calcium accumulation and ROS generation in the coronary artery smooth muscle cells isolated from either the ischemic or the nonischemic part of the CAL coronary artery vessel were significantly increased irrespective of blood supply (all P < 0.01). Moreover, these were accompanied by the increased expressions of NCX and p47phox, the decreased expression of SERCA2a, and the increased amount of phosphorylated forms of p47phox in NADPH oxidase (all P < 0.05). CONCLUSIONS Our results demonstrated that the disordered calcium clearance and the increased ROS generation occurred in the coronary artery smooth muscle cells in rats with chronic HF produced by ligation of the left anterior descending coronary artery (CAL), and which was found to be disassociated from blood supply, and the increased generation of ROS in the cells was found to make concomitancy to the increased activity of NADPH oxidase in cytoplasm.
Collapse
Affiliation(s)
- Liang Ding
- Department of Pharmacology, School of Medicine, Hebei University, Baoding, Hebei 071000, China
| | - Xian-Xiu Su
- Department of Basic Medicine, School of Basic Medicine, Hebei University, Baoding, Hebei 071000, China
| | - Wen-Hui Zhang
- Department of Pharmacology, School of Medicine, Hebei University, Baoding, Hebei 071000, China
| | - Yu-Xiang Xu
- Department of Pharmacology, School of Medicine, Hebei University, Baoding, Hebei 071000, China
| | - Xue-Feng Pan
- Department of Pharmacology, School of Medicine, Hebei University, Baoding, Hebei 071000, China
- Department of Basic Medicine, School of Basic Medicine, Hebei University, Baoding, Hebei 071000, China
- Department of Biological Science, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
72
|
Differential regulation of the Na +-Ca 2+ exchanger 3 (NCX3) by protein kinase PKC and PKA. Cell Calcium 2017; 65:52-62. [PMID: 28233567 DOI: 10.1016/j.ceca.2017.02.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 02/09/2017] [Accepted: 02/09/2017] [Indexed: 11/21/2022]
Abstract
Isoform 3 of the Na+-Ca2+ exchanger (NCX3) participates in the Ca2+ fluxes across the plasma membrane. Among the NCX family, NCX3 carries out a peculiar role due to its specific functions in skeletal muscle and the immune system and to its neuroprotective effect under stress exposure. In this context, proper understanding of the regulation of NCX3 is primordial to consider its potential use as a drug target. In this study, we demonstrated the regulation of NCX3 by protein kinase A (PKA) and C (PKC). Disparity in regulation has been previously reported among the splice variants of NCX3 therefore the activity of Ca2+ uptake and extrusion of the two murine variants was measured using fura-2-based Ca2+ imaging and revealed that both variants are similarly regulated. PKC stimulation diminished the Ca2+ uptake performed by NCX3 in the reverse mode, triggered by a rise in [Ca2+]i or [Na+]i, whereas an opposite response was observed upon PKA stimulation, with a significant increase of the Ca2+ uptake after a rise in [Ca2+]i. The latter stimulation affected similarly the efflux capacity of NCX3 whereas Ca2+ extrusion capacity remained unaffected under activation of PKC. Next, using site-directed mutagenesis, the sensitivity of NCX3 to PKC was abolished by singly mutating its predicted phosphorylation sites T529 or S695. The sensitivity to PKC might be due to the influence of T529 phosphorylation on the Ca2+-binding domain 1. Additionally, we showed that stimulation of NCX3 by PKA occurred through residue S524. This effect may well participate in the fight-or-flight response in skeletal muscle and the long-term potentiation in hippocampus.
Collapse
|
73
|
Averin AS, Kosarsky LS, Tarlachkov SV, Vekhnik VA, Averina IV, Alekseev AE, Fesenko EE, Nakipova OV. The effects of KB-R7943, an inhibitor of reverse Na+/Ca2+ exchange, on the force of contraction of papillary muscles in the heart of the ground squirrel Spermophilus undulatus. Biophysics (Nagoya-shi) 2017. [DOI: 10.1134/s000635091701002x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
74
|
Santana ET, Feliciano RDS, Serra AJ, Brigidio E, Antonio EL, Tucci PJF, Nathanson L, Morris M, Silva JA. Comparative mRNA and MicroRNA Profiling during Acute Myocardial Infarction Induced by Coronary Occlusion and Ablation Radio-Frequency Currents. Front Physiol 2016; 7:565. [PMID: 27932994 PMCID: PMC5123550 DOI: 10.3389/fphys.2016.00565] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 11/07/2016] [Indexed: 12/24/2022] Open
Abstract
The ligation of the left anterior descending coronary artery is the most commonly used experimental model to induce myocardial infarction (MI) in rodents. A high mortality in the acute phase and the heterogeneity of the size of the MI obtained are drawbacks recognized in this model. In an attempt to solve the problem, our group recently developed a new MI experimental model which is based on application of myocardial ablation radio-frequency currents (AB-RF) that yielded MI with homogeneous sizes and significantly reduce acute mortality. In addition, cardiac structural, and functional changes aroused by AB-RF were similar to those seen in animals with MI induced by coronary artery ligation. Herein, we compared mRNA expression of genes that govern post-MI milieu in occlusion and ablation models. We analyzed 48 mRNAs expressions of nine different signal transduction pathways (cell survival and metabolism signs, matrix extracellular, cell cycle, oxidative stress, apoptosis, calcium signaling, hypertrophy markers, angiogenesis, and inflammation) in rat left ventricle 1 week after MI generated by both coronary occlusion and AB-RF. Furthermore, high-throughput miRNA analysis was also assessed in both MI procedures. Interestingly, mRNA expression levels and miRNA expressions showed strong similarities between both models after MI, with few specificities in each model, activating similar signal transduction pathways. To our knowledge, this is the first comparison of genomic alterations of mRNA and miRNA contents after two different MI procedures and identifies key signaling regulators modulating the pathophysiology of these two models that might culminate in heart failure. Furthermore, these analyses may contribute with the current knowledge concerning transcriptional and post-transcriptional changes of AB-RF protocol, arising as an alternative and effective MI method that reproduces most changes seem in coronary occlusion.
Collapse
Affiliation(s)
- Eduardo T Santana
- Rehabilitation Department, Universidade Nove de Julho São Paulo, Brazil
| | - Regiane Dos Santos Feliciano
- Biophotonics Department, Universidade Nove de JulhoSão Paulo, Brazil; Medicine Department, Universidade Nove de JulhoSão Paulo, Brazil
| | - Andrey J Serra
- Biophotonics Department, Universidade Nove de Julho São Paulo, Brazil
| | - Eduardo Brigidio
- Medicine Department, Universidade Nove de Julho São Paulo, Brazil
| | - Ednei L Antonio
- Cardiac Physiology Department, Universidade Federal de São Paulo São Paulo, Brazil
| | - Paulo J F Tucci
- Cardiac Physiology Department, Universidade Federal de São Paulo São Paulo, Brazil
| | - Lubov Nathanson
- Institute for Neuro-Immune Medicine, Nova Southeastern University Fort Lauderdale, FL, USA
| | - Mariana Morris
- Institute for Neuro-Immune Medicine, Nova Southeastern University Fort Lauderdale, FL, USA
| | - José A Silva
- Medicine Department, Universidade Nove de Julho São Paulo, Brazil
| |
Collapse
|
75
|
Epi-reevesioside F inhibits Na+/K+-ATPase, causing cytosolic acidification, Bak activation and apoptosis in glioblastoma. Oncotarget 2016; 6:24032-46. [PMID: 26125228 PMCID: PMC4695168 DOI: 10.18632/oncotarget.4429] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 06/04/2015] [Indexed: 12/17/2022] Open
Abstract
Epi-reevesioside F, a new cardiac glycoside isolated from the root of Reevesia formosana, displayed potent activity against glioblastoma cells. Epi-reevesioside F was more potent than ouabain with IC50 values of 27.3±1.7 vs. 48.7±1.8 nM (P < 0.001) and 45.0±3.4 vs. 81.3±4.3 nM (P < 0.001) in glioblastoma T98 and U87 cells, respectively. However, both Epi-reevesioside F and ouabain were ineffective in A172 cells, a glioblastoma cell line with low Na+/K+-ATPase α3 subunit expression. Epi-reevesioside F induced cell cycle arrest at S and G2 phases and apoptosis. It also induced an increase of intracellular concentration of Na+ but not Ca2+, cleavage and exposure of N-terminus of Bak, loss of mitochondrial membrane potential, inhibition of Akt activity and induction of caspase cascades. Potassium supplements significantly inhibited Epi-reevesioside F-induced effects. Notably, Epi-reevesioside F caused cytosolic acidification that was highly correlated with the anti-proliferative activity. In summary, the data suggest that Epi-reevesioside F inhibits Na+/K+-ATPase, leading to overload of intracellular Na+ and cytosolic acidification, Bak activation and loss of mitochondrial membrane potential. The PI3-kinase/Akt pathway is inhibited and caspase-dependent apoptosis is ultimately triggered in Epi-reevesioside F-treated glioblastoma cells.
Collapse
|
76
|
Blaustein MP, Chen L, Hamlyn JM, Leenen FHH, Lingrel JB, Wier WG, Zhang J. Pivotal role of α2 Na + pumps and their high affinity ouabain binding site in cardiovascular health and disease. J Physiol 2016; 594:6079-6103. [PMID: 27350568 DOI: 10.1113/jp272419] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 06/18/2016] [Indexed: 12/13/2022] Open
Abstract
Reduced smooth muscle (SM)-specific α2 Na+ pump expression elevates basal blood pressure (BP) and increases BP sensitivity to angiotensin II (Ang II) and dietary NaCl, whilst SM-α2 overexpression lowers basal BP and decreases Ang II/salt sensitivity. Prolonged ouabain infusion induces hypertension in rodents, and ouabain-resistant mutation of the α2 ouabain binding site (α2R/R mice) confers resistance to several forms of hypertension. Pressure overload-induced heart hypertrophy and failure are attenuated in cardio-specific α2 knockout, cardio-specific α2 overexpression and α2R/R mice. We propose a unifying hypothesis that reconciles these apparently disparate findings: brain mechanisms, activated by Ang II and high NaCl, regulate sympathetic drive and a novel neurohumoral pathway mediated by both brain and circulating endogenous ouabain (EO). Circulating EO modulates ouabain-sensitive α2 Na+ pump activity and Ca2+ transporter expression and, via Na+ /Ca2+ exchange, Ca2+ homeostasis. This regulates sensitivity to sympathetic activity, Ca2+ signalling and arterial and cardiac contraction.
Collapse
Affiliation(s)
- Mordecai P Blaustein
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| | - Ling Chen
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - John M Hamlyn
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Frans H H Leenen
- Hypertension Unit, University of Ottawa Heart Institute, Ottawa, ON, Canada, K1Y 4W7
| | - Jerry B Lingrel
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267-0524, USA
| | - W Gil Wier
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jin Zhang
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| |
Collapse
|
77
|
Grover AK. Sodium-Calcium Exchanger in Pig Coronary Artery. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 78:145-170. [PMID: 28212796 DOI: 10.1016/bs.apha.2016.06.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review focuses on the sodium-calcium exchangers (NCX) in the left anterior descending coronary artery smooth muscle. Bathing tissues in Na+-substituted solutions caused them to contract. In cultured smooth muscle cells, it increased the cytosolic Ca2+ concentration and extracellular entry of 45Ca2+. All three activities were attributed to NCX since they were inhibited by NCX inhibitors. The tissues also expressed the sarco/endoplasmic reticulum (SER) Ca2+ pump SERCA2b whose activity was much greater than that of NCX. Inhibiting SERCA2b with thapsigargin decreased the NCX-mediated 45Ca2+ accumulation by the cells. The decrease was not observed in cells loaded with the Ca2+-chelator BAPTA. The results are consistent with a limited diffusional space model with a proximity between NCX and SERCA2b. NCX molecules appear to be colocalized with the subsarcolemmal SERCA2b based on studies on membrane flotation experiments and microscopic fluorescence imaging of antibody-labeled cells. Thapsigargin inhibition of SERCA2b moved NCX even closer to SER. This provides a model for the NCX-mediated Ca2+ refilling of SER in the arterial smooth muscle. The model for the NCX-mediated refilling of the depleted SER proposed for smooth muscle did not apply to endothelium in which NCX levels were greater and SERCA levels were lower than in smooth muscle. The effect of thapsigargin on the NCX-mediated Ca2+ accumulation which was observed in smooth muscle was absent in the endothelium. We propose that the coupling between NCX and smooth muscle may be tissue dependent.
Collapse
Affiliation(s)
- A K Grover
- McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
78
|
Acsai K, Ördög B, Varró A, Nánási PP. Role of the dysfunctional ryanodine receptor - Na(+)-Ca(2+)exchanger axis in progression of cardiovascular diseases: What we can learn from pharmacological studies? Eur J Pharmacol 2016; 779:91-101. [PMID: 26970182 DOI: 10.1016/j.ejphar.2016.03.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 03/04/2016] [Accepted: 03/07/2016] [Indexed: 12/28/2022]
Abstract
Abnormal Ca(2+)homeostasis is often associated with chronic cardiovascular diseases, such as hypertension, heart failure or cardiac arrhythmias, and typically contributes to the basic ethiology of the disease. Pharmacological targeting of cardiac Ca(2+)handling has great therapeutic potential offering invaluable options for the prevention, slowing down the progression or suppression of the harmful outcomes like life threatening cardiac arrhythmias. In this review we outline the existing knowledge on the involvement of malfunction of the ryanodine receptor and the Na(+)-Ca(2+)exchanger in disturbances of Ca(2+)homeostasis and discuss important proof of concept pharmacological studies targeting these mechanisms in context of hypertension, heart failure, atrial fibrillation and ventricular arrhythmias. We emphasize the promising results of preclinical studies underpinning the potential benefits of the therapeutic strategies based on ryanodine receptor or Na(+)-Ca(2+)exchanger inhibition.
Collapse
Affiliation(s)
- Károly Acsai
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Szeged, Hungary
| | - Balázs Ördög
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Faculty of Medicine, Szeged, Hungary
| | - András Varró
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Szeged, Hungary; Department of Pharmacology and Pharmacotherapy, University of Szeged, Faculty of Medicine, Szeged, Hungary
| | - Péter P Nánási
- Department of Physiology, University of Debrecen, Debrecen, Hungary; Department of Dentistry, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
79
|
Carvalho FB, Boligon AA, Athayde ML, Rubin MA, Ferreira J, Trevisan G. Inhibitory effect of Scutia buxifolia extracts, fractions, and ursolic acid on Na(+), K(+)-ATPase activity in vitro in membranes purified from rat hearts. JOURNAL OF ETHNOPHARMACOLOGY 2016; 179:45-54. [PMID: 26719288 DOI: 10.1016/j.jep.2015.12.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 12/19/2015] [Accepted: 12/20/2015] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Scutia buxifolia is a tree native to South America and is used as a cardiotonic agent; however, this property has not been associated with a clear mechanism or a specific compound. AIM OF THE STUDY Given the importance of Na(+),K(+)-ATPase as a drug target in the treatment of heart failure, this study aimed to investigate the possible inhibitory effect of S. buxifolia crude extract and fractions (dichloromethane, ethyl acetate, and butanolic fractions), and identified compounds with effects on the activity of Na(+),K(+)-ATPase in vitro. MATERIALS AND METHODS First, we characterized the crude extract and fractions by high-performance liquid chromatography, and then monitored their effects on the activity of Na(+),K(+)-ATPase obtained from heart muscle and brain membranes of adult male Wistar rats. RESULTS We identified gallic acid, chlorogenic acid, caffeic acid, rutin, quercitrin, quercetin, and ursolic acid in S. buxifolia stem bark and leaves; quercitrin and ursolic acid were the main compounds in the ethyl acetate and dichloromethane fractions from leaves and stem bark. The crude extract (3 and 30mg/ml), and the ethyl acetate and dichloromethane fractions (0.1 and 1mg/ml) of both the stem bark and leaves inhibited Na(+),K(+)-ATPase activity in heart and brain samples. We found that, of the identified compounds, only ursolic acid (0.1mg/ml) was able to diminish Na(+), K(+)-ATPase activity in heart and brain samples. CONCLUSIONS These data indicated that the cardiotonic effects of S. buxifolia may be due to the inhibition of Na(+),K(+)-ATPase activity in heart muscle, supporting the popular use of this plant as a treatment for heart failure.
Collapse
Affiliation(s)
- Fabiano B Carvalho
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria (UFSM), 97105-900 Santa Maria, RS, Brazil
| | - Aline A Boligon
- Programa de Pós-graduação em Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal de Santa Maria (UFSM), 97105-900 Santa Maria, RS, Brazil
| | - Margareth L Athayde
- Programa de Pós-graduação em Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal de Santa Maria (UFSM), 97105-900 Santa Maria, RS, Brazil
| | - Maribel A Rubin
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria (UFSM), 97105-900 Santa Maria, RS, Brazil
| | - Juliano Ferreira
- Programa de Pós-graduação em Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), 88049-900 Florianópolis, SC, Brazil
| | - Gabriela Trevisan
- Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (Unesc), 88806-000 Criciúma, SC, Brazil.
| |
Collapse
|
80
|
Burst pacemaker activity of the sinoatrial node in sodium-calcium exchanger knockout mice. Proc Natl Acad Sci U S A 2015. [PMID: 26195795 DOI: 10.1073/pnas.1505670112] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In sinoatrial node (SAN) cells, electrogenic sodium-calcium exchange (NCX) is the dominant calcium (Ca) efflux mechanism. However, the role of NCX in the generation of SAN automaticity is controversial. To investigate the contribution of NCX to pacemaking in the SAN, we performed optical voltage mapping and high-speed 2D laser scanning confocal microscopy (LSCM) of Ca dynamics in an ex vivo intact SAN/atrial tissue preparation from atrial-specific NCX knockout (KO) mice. These mice lack P waves on electrocardiograms, and isolated NCX KO SAN cells are quiescent. Voltage mapping revealed disorganized and arrhythmic depolarizations within the NCX KO SAN that failed to propagate into the atria. LSCM revealed intermittent bursts of Ca transients. Bursts were accompanied by rising diastolic Ca, culminating in long pauses dominated by Ca waves. The L-type Ca channel agonist BayK8644 reduced the rate of Ca transients and inhibited burst generation in the NCX KO SAN whereas the Ca buffer 1,2-Bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (acetoxymethyl ester) (BAPTA AM) did the opposite. These results suggest that cellular Ca accumulation hinders spontaneous depolarization in the NCX KO SAN, possibly by inhibiting L-type Ca currents. The funny current (If) blocker ivabradine also suppressed NCX KO SAN automaticity. We conclude that pacemaker activity is present in the NCX KO SAN, generated by a mechanism that depends upon If. However, the absence of NCX-mediated depolarization in combination with impaired Ca efflux results in intermittent bursts of pacemaker activity, reminiscent of human sinus node dysfunction and "tachy-brady" syndrome.
Collapse
|
81
|
Eykyn TR, Aksentijević D, Aughton KL, Southworth R, Fuller W, Shattock MJ. Multiple quantum filtered (23)Na NMR in the Langendorff perfused mouse heart: Ratio of triple/double quantum filtered signals correlates with [Na]i. J Mol Cell Cardiol 2015. [PMID: 26196304 PMCID: PMC4564289 DOI: 10.1016/j.yjmcc.2015.07.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
We investigate the potential of multiple quantum filtered (MQF) 23Na NMR to probe intracellular [Na]i in the Langendorff perfused mouse heart. In the presence of Tm(DOTP) shift reagent the triple quantum filtered (TQF) signal originated largely from the intracellular sodium pool with a 32 ± 6% contribution of the total TQF signal arising from extracellular sodium, whilst the rank 2 double-quantum filtered signal (DQF), acquired with a 54.7° flip-angle pulse, originated exclusively from the extracellular sodium pool. Given the different cellular origins of the 23Na MQF signals we propose that the TQF/DQF ratio can be used as a semi-quantitative measure of [Na]i in the mouse heart. We demonstrate a good correlation of this ratio with [Na]i measured with shift reagent at baseline and under conditions of elevated [Na]i. We compare the measurements of [Na]i using both shift reagent and TQF/DQF ratio in a cohort of wild type mouse hearts and in a transgenic PLM3SA mouse expressing a non-phosphorylatable form of phospholemman, showing a modest but measurable elevation of baseline [Na]i. MQF filtered 23Na NMR is a potentially useful tool for studying normal and pathophysiological changes in [Na]i, particularly in transgenic mouse models with altered Na regulation. Intracellular Na concentration [Na]i is a key modulator of cardiac cell function. We developed an NMR-compatible Langendorff mouse heart perfusion system. The ratio of triple/double quantum filtered 23Na NMR signals correlates with [Na]i. Intracellular [Na]i can be quantified under physiological perfusion conditions. The PLM3SA transgenic mouse model has a measurable elevation of [Na]i at baseline.
Collapse
Affiliation(s)
- Thomas R Eykyn
- Department of Imaging Chemistry and Biology, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London SE1 7EH, United Kingdom; The British Heart Foundation Centre of Research Excellence, The Rayne Institute, King's College London, St. Thomas' Hospital, London SE1 7EH, United Kingdom.
| | - Dunja Aksentijević
- The British Heart Foundation Centre of Research Excellence, The Rayne Institute, King's College London, St. Thomas' Hospital, London SE1 7EH, United Kingdom
| | - Karen L Aughton
- The British Heart Foundation Centre of Research Excellence, The Rayne Institute, King's College London, St. Thomas' Hospital, London SE1 7EH, United Kingdom
| | - Richard Southworth
- Department of Imaging Chemistry and Biology, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London SE1 7EH, United Kingdom; The British Heart Foundation Centre of Research Excellence, The Rayne Institute, King's College London, St. Thomas' Hospital, London SE1 7EH, United Kingdom
| | - William Fuller
- Division of Cardiovascular and Diabetes Medicine, University of Dundee, Dundee, United Kingdom
| | - Michael J Shattock
- The British Heart Foundation Centre of Research Excellence, The Rayne Institute, King's College London, St. Thomas' Hospital, London SE1 7EH, United Kingdom
| |
Collapse
|
82
|
Feridooni HA, Dibb KM, Howlett SE. How cardiomyocyte excitation, calcium release and contraction become altered with age. J Mol Cell Cardiol 2015; 83:62-72. [PMID: 25498213 DOI: 10.1016/j.yjmcc.2014.12.004] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 12/02/2014] [Accepted: 12/04/2014] [Indexed: 11/29/2022]
Abstract
Cardiovascular disease is the main cause of death globally, accounting for over 17 million deaths each year. As the incidence of cardiovascular disease rises markedly with age, the overall risk of cardiovascular disease is expected to increase dramatically with the aging of the population such that by 2030 it could account for over 23 million deaths per year. It is therefore vitally important to understand how the heart remodels in response to normal aging for at least two reasons: i) to understand why the aged heart is increasingly susceptible to disease; and ii) since it may be possible to modify treatment of disease in older adults if the underlying substrate upon which the disease first develops is fully understood. It is well known that age modulates cardiac function at the level of the individual cardiomyocyte. Generally, in males, aging reduces cell shortening, which is associated with a decrease in the amplitude of the systolic Ca(2+) transient. This may arise due to a decrease in peak L-type Ca(2+) current. Sarcoplasmic reticulum (SR) Ca(2+) load appears to be maintained during normal aging but evidence suggests that SR function is disrupted, such that the rate of sarco/endoplasmic reticulum Ca(2+)-ATPase (SERCA)-mediated Ca(2+) removal is reduced and the properties of SR Ca(2+) release in terms of Ca(2+) sparks are altered. Interestingly, Ca(2+) handling is modulated by age to a lesser degree in females. Here we review how cellular contraction is altered as a result of the aging process by considering expression levels and functional properties of key proteins involved in controlling intracellular Ca(2+). We consider how changes in both electrical properties and intracellular Ca(2+) handling may interact to modulate cardiomyocyte contraction. We also reflect on why cardiovascular risk may differ between the sexes by highlighting sex-specific variation in the age-associated remodeling process. This article is part of a Special Issue entitled CV Aging.
Collapse
Affiliation(s)
- Hirad A Feridooni
- Department of Pharmacology, Dalhousie University, PO Box 15000, 5850 College St, Halifax, NS B3H 4R2, Canada.
| | - Katharine M Dibb
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK.
| | - Susan E Howlett
- Department of Pharmacology, Dalhousie University, PO Box 15000, 5850 College St, Halifax, NS B3H 4R2, Canada; Department of Medicine (Geriatric Medicine), Dalhousie University, PO Box 15000, 5850 College St, Halifax, NS B3H 4R2, Canada; Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK.
| |
Collapse
|
83
|
Interaction of H2S with Calcium Permeable Channels and Transporters. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:323269. [PMID: 26078804 PMCID: PMC4442308 DOI: 10.1155/2015/323269] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 10/14/2014] [Accepted: 11/12/2014] [Indexed: 01/13/2023]
Abstract
A growing amount of evidence has suggested that hydrogen sulfide (H2S), as a gasotransmitter, is involved in intensive physiological and pathological processes. More and more research groups have found that H2S mediates diverse cellular biological functions related to regulating intracellular calcium concentration. These groups have demonstrated the reciprocal interaction between H2S and calcium ion channels and transporters, such as L-type calcium channels (LTCC), T-type calcium channels (TTCC), sodium/calcium exchangers (NCX), transient receptor potential (TRP) channels, β-adrenergic receptors, and N-methyl-D-aspartate receptors (NMDAR) in different cells. However, the understanding of the molecular targets and mechanisms is incomplete. Recently, some research groups demonstrated that H2S modulates the activity of calcium ion channels through protein S-sulfhydration and polysulfide reactions. In this review, we elucidate that H2S controls intracellular calcium homeostasis and the underlying mechanisms.
Collapse
|
84
|
Capel RA, Terrar DA. The importance of Ca(2+)-dependent mechanisms for the initiation of the heartbeat. Front Physiol 2015; 6:80. [PMID: 25859219 PMCID: PMC4373508 DOI: 10.3389/fphys.2015.00080] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/02/2015] [Indexed: 01/01/2023] Open
Abstract
Mechanisms underlying pacemaker activity in the sinus node remain controversial, with some ascribing a dominant role to timing events in the surface membrane (“membrane clock”) and others to uptake and release of calcium from the sarcoplasmic reticulum (SR) (“calcium clock”). Here we discuss recent evidence on mechanisms underlying pacemaker activity with a particular emphasis on the many roles of calcium. There are particular areas of controversy concerning the contribution of calcium spark-like events and the importance of I(f) to spontaneous diastolic depolarisation, though it will be suggested that neither of these is essential for pacemaking. Sodium-calcium exchange (NCX) is most often considered in the context of mediating membrane depolarisation after spark-like events. We present evidence for a broader role of this electrogenic exchanger which need not always depend upon these spark-like events. Short (milliseconds or seconds) and long (minutes) term influences of calcium are discussed including direct regulation of ion channels and NCX, and control of the activity of calcium-dependent enzymes (including CaMKII, AC1, and AC8). The balance between the many contributory factors to pacemaker activity may well alter with experimental and clinical conditions, and potentially redundant mechanisms are desirable to ensure the regular spontaneous heart rate that is essential for life. This review presents evidence that calcium is central to the normal control of pacemaking across a range of temporal scales and seeks to broaden the accepted description of the “calcium clock” to cover these important influences.
Collapse
Affiliation(s)
- Rebecca A Capel
- British Heart Foundation Centre of Research Excellence, Department of Pharmacology, University of Oxford Oxford, UK
| | - Derek A Terrar
- British Heart Foundation Centre of Research Excellence, Department of Pharmacology, University of Oxford Oxford, UK
| |
Collapse
|
85
|
Uryash A, Bassuk J, Kurlansky P, Altamirano F, Lopez JR, Adams JA. Non-invasive technology that improves cardiac function after experimental myocardial infarction: Whole Body Periodic Acceleration (pGz). PLoS One 2015; 10:e0121069. [PMID: 25807532 PMCID: PMC4373845 DOI: 10.1371/journal.pone.0121069] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/27/2015] [Indexed: 01/04/2023] Open
Abstract
Myocardial infarction (MI) may produce significant inflammatory changes and adverse ventricular remodeling leading to heart failure and premature death. Pharmacologic, stem cell transplantation, and exercise have not halted the inexorable rise in the prevalence and great economic costs of heart failure despite extensive investigations of such treatments. New therapeutic modalities are needed. Whole Body Periodic Acceleration (pGz) is a non-invasive technology that increases pulsatile shear stress to the endothelium thereby producing several beneficial cardiovascular effects as demonstrated in animal models, normal humans and patients with heart disease. pGz upregulates endothelial derived nitric oxide synthase (eNOS) and its phosphorylation (p-eNOS) to improve myocardial function in models of myocardial stunning and preconditioning. Here we test whether pGz applied chronically after focal myocardial infarction in rats improves functional outcomes from MI. Focal MI was produced by left coronary artery ligation. One day after ligation animals were randomized to receive daily treatments of pGz for four weeks (MI-pGz) or serve as controls (MI-CONT), with an additional group as non-infarction controls (Sham). Echocardiograms and invasive pressure volume loop analysis were carried out. Infarct transmurality, myocardial fibrosis, and markers of inflammatory and anti-inflammatory cytokines were determined along with protein analysis of eNOS, p-eNOS and inducible nitric oxide synthase (iNOS).At four weeks, survival was 80% in MI-pGz vs 50% in MI-CONT (p< 0.01). Ejection fraction and fractional shortening and invasive pressure volume relation indices of afterload and contractility were significantly better in MI-pGz. The latter where associated with decreased infarct transmurality and decreased fibrosis along with increased eNOS, p-eNOS. Additionally, MI-pGz had significantly lower levels of iNOS, inflammatory cytokines (IL-6, TNF-α), and higher level of anti-inflammatory cytokine (IL-10). pGz improved survival and contractile performance, associated with improved myocardial remodeling. pGz may serve as a simple, safe, non-invasive therapeutic modality to improve myocardial function after MI.
Collapse
Affiliation(s)
- Arkady Uryash
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL, United States of America
| | - Jorge Bassuk
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL, United States of America
| | - Paul Kurlansky
- Columbia Heart Source, Columbia University College of Physicians and Surgeons, New York, NY, United States of America
| | - Francisco Altamirano
- Departments of Molecular Bioscience, School of Veterinary Medicine, University of California Davis, Davis, CA, United States of America
| | - Jose R. Lopez
- Departments of Molecular Bioscience, School of Veterinary Medicine, University of California Davis, Davis, CA, United States of America
| | - Jose A. Adams
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL, United States of America
| |
Collapse
|
86
|
Louch WE, Koivumäki JT, Tavi P. Calcium signalling in developing cardiomyocytes: implications for model systems and disease. J Physiol 2015; 593:1047-63. [PMID: 25641733 PMCID: PMC4358669 DOI: 10.1113/jphysiol.2014.274712] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Accepted: 12/28/2014] [Indexed: 12/15/2022] Open
Abstract
Adult cardiomyocytes exhibit complex Ca(2+) homeostasis, enabling tight control of contraction and relaxation. This intricate regulatory system develops gradually, with progressive maturation of specialized structures and increasing capacity of Ca(2+) sources and sinks. In this review, we outline current understanding of these developmental processes, and draw parallels to pathophysiological conditions where cardiomyocytes exhibit a striking regression to an immature state of Ca(2+) homeostasis. We further highlight the importance of understanding developmental physiology when employing immature cardiomyocyte models such as cultured neonatal cells and stem cells.
Collapse
Affiliation(s)
- William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo0424, Oslo, Norway
- K. G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo0316, Oslo, Norway
| | - Jussi T Koivumäki
- Simula Research Laboratory, Center for Cardiological Innovation and Center for Biomedical ComputingOslo, Norway
| | - Pasi Tavi
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern FinlandKuopio, Finland
| |
Collapse
|
87
|
Ayaz O, Howlett SE. Testosterone modulates cardiac contraction and calcium homeostasis: cellular and molecular mechanisms. Biol Sex Differ 2015; 6:9. [PMID: 25922656 PMCID: PMC4411792 DOI: 10.1186/s13293-015-0027-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 03/27/2015] [Indexed: 02/06/2023] Open
Abstract
The incidence of cardiovascular disease rises dramatically with age in both men and women. Because a woman's risk of cardiovascular disease rises markedly after the onset of menopause, there has been growing interest in the effect of estrogen on the heart and its role in the pathophysiology of these diseases. Much less attention has been paid to the impact of testosterone on the heart, even though the levels of testosterone also decline with age and low-testosterone levels are linked to the development of cardiovascular diseases. The knowledge that receptors for all major sex steroid hormones, including testosterone, are present on individual cardiomyocytes suggests that these hormones may influence the heart at the cellular level. Indeed, it is well established that there are male-female differences in intracellular Ca(2+) release and contraction in isolated ventricular myocytes. Growing evidence suggests that these differences arise from effects of sex steroid hormones on processes involved in intracellular Ca(2+) homeostasis. This review considers how myocardial contractile function is modified by testosterone, with a focus on the impact of testosterone on processes that regulate Ca(2+) handling at the level of the ventricular myocyte. The idea that testosterone regulates Ca(2+) handling in the heart is important, as Ca(2+) dysregulation plays a key role in the pathogenesis of a variety of different cardiovascular diseases. A better understanding of sex hormone regulation of myocardial Ca(2+) homeostasis may reveal new targets for the treatment of cardiovascular diseases in all older adults.
Collapse
Affiliation(s)
- Omar Ayaz
- Department of Pharmacology, Dalhousie University, 5850 College Street, Sir Charles Tupper Medical Building, PO Box 15000, Halifax, NS B3H 4R2 Canada
| | - Susan Ellen Howlett
- Department of Pharmacology, Dalhousie University, 5850 College Street, Sir Charles Tupper Medical Building, PO Box 15000, Halifax, NS B3H 4R2 Canada
- Medicine (Geriatric Medicine), Dalhousie University, 5850 College Street, PO Box 15000, Halifax, NS B3H 4R2 Canada
| |
Collapse
|
88
|
Greiser M, Kerfant BG, Williams GS, Voigt N, Harks E, Dibb KM, Giese A, Meszaros J, Verheule S, Ravens U, Allessie MA, Gammie JS, van der Velden J, Lederer WJ, Dobrev D, Schotten U. Tachycardia-induced silencing of subcellular Ca2+ signaling in atrial myocytes. J Clin Invest 2014; 124:4759-72. [PMID: 25329692 PMCID: PMC4347234 DOI: 10.1172/jci70102] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Accepted: 08/28/2014] [Indexed: 01/06/2023] Open
Abstract
Atrial fibrillation (AF) is characterized by sustained high atrial activation rates and arrhythmogenic cellular Ca2+ signaling instability; however, it is not clear how a high atrial rate and Ca2+ instability may be related. Here, we characterized subcellular Ca2+ signaling after 5 days of high atrial rates in a rabbit model. While some changes were similar to those in persistent AF, we identified a distinct pattern of stabilized subcellular Ca2+ signaling. Ca2+ sparks, arrhythmogenic Ca2+ waves, sarcoplasmic reticulum (SR) Ca2+ leak, and SR Ca2+ content were largely unaltered. Based on computational analysis, these findings were consistent with a higher Ca2+ leak due to PKA-dependent phosphorylation of SR Ca2+ channels (RyR2s), fewer RyR2s, and smaller RyR2 clusters in the SR. We determined that less Ca2+ release per [Ca2+]i transient, increased Ca2+ buffering strength, shortened action potentials, and reduced L-type Ca2+ current contribute to a stunning reduction of intracellular Na+ concentration following rapid atrial pacing. In both patients with AF and in our rabbit model, this silencing led to failed propagation of the [Ca2+]i signal to the myocyte center. We conclude that sustained high atrial rates alone silence Ca2+ signaling and do not produce Ca2+ signaling instability, consistent with an adaptive molecular and cellular response to atrial tachycardia.
Collapse
Affiliation(s)
- Maura Greiser
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Benoît-Gilles Kerfant
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - George S.B. Williams
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Niels Voigt
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Erik Harks
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Katharine M. Dibb
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Anne Giese
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Janos Meszaros
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Sander Verheule
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Ursula Ravens
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Maurits A. Allessie
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - James S. Gammie
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - W. Jonathan Lederer
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Dobromir Dobrev
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Ulrich Schotten
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
89
|
Khananshvili D. Sodium-calcium exchangers (NCX): molecular hallmarks underlying the tissue-specific and systemic functions. Pflugers Arch 2013; 466:43-60. [PMID: 24281864 DOI: 10.1007/s00424-013-1405-y] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Revised: 11/06/2013] [Accepted: 11/09/2013] [Indexed: 12/19/2022]
Abstract
NCX proteins explore the electrochemical gradient of Na(+) to mediate Ca(2+)-fluxes in exchange with Na(+) either in the Ca(2+)-efflux (forward) or Ca(2+)-influx (reverse) mode, whereas the directionality depends on ionic concentrations and membrane potential. Mammalian NCX variants (NCX1-3) and their splice variants are expressed in a tissue-specific manner to modulate the heartbeat rate and contractile force, the brain's long-term potentiation and learning, blood pressure, renal Ca(2+) reabsorption, the immune response, neurotransmitter and insulin secretion, apoptosis and proliferation, mitochondrial bioenergetics, etc. Although the forward mode of NCX represents a major physiological module, a transient reversal of NCX may contribute to EC-coupling, vascular constriction, and synaptic transmission. Notably, the reverse mode of NCX becomes predominant in pathological settings. Since the expression levels of NCX variants are disease-related, the selective pharmacological targeting of tissue-specific NCX variants could be beneficial, thereby representing a challenge. Recent structural and biophysical studies revealed a common module for decoding the Ca(2+)-induced allosteric signal in eukaryotic NCX variants, although the phenotype variances in response to regulatory Ca(2+) remain unclear. The breakthrough discovery of the archaebacterial NCX structure may serve as a template for eukaryotic NCX, although the turnover rates of the transport cycle may differ ~10(3)-fold among NCX variants to fulfill the physiological demands for the Ca(2+) flux rates. Further elucidation of ion-transport and regulatory mechanisms may lead to selective pharmacological targeting of NCX variants under disease conditions.
Collapse
Affiliation(s)
- Daniel Khananshvili
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel-Aviv University, Ramat-Aviv, Tel-Aviv, 69978, Israel,
| |
Collapse
|