1
|
Zhong M, Karma A. Role of ryanodine receptor cooperativity in Ca 2+-wave-mediated triggered activity in cardiomyocytes. J Physiol 2024; 602:6745-6787. [PMID: 39565684 DOI: 10.1113/jp286145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 09/23/2024] [Indexed: 11/22/2024] Open
Abstract
Ca2+ waves are known to trigger delayed after-depolarizations that can cause malignant cardiac arrhythmias. However, modelling Ca2+ waves using physiologically realistic models has remained a major challenge. Existing models with low Ca2+ sensitivity of ryanodine receptors (RyRs) necessitate large release currents, leading to an unrealistically large Ca2+ transient amplitude incompatible with the experimental observations. Consequently, current physiologically detailed models of delayed after-depolarizations resort to unrealistic cell architectures to produce Ca2+ waves with a normal Ca2+ transient amplitude. Here, we address these challenges by incorporating RyR cooperativity into a physiologically detailed model with a realistic cell architecture. We represent RyR cooperativity phenomenologically through a Hill coefficient within the sigmoid function of RyR open probability. Simulations in permeabilized myocytes with high Ca2+ sensitivity reveal that a sufficiently large Hill coefficient is required for Ca2+ wave propagation via the fire-diffuse-fire mechanism. In intact myocytes, propagating Ca2+ waves can occur only within an intermediate Hill coefficient range. Within this range, the spark rate is neither too low, enabling Ca2+ wave propagation, nor too high, allowing for the maintenance of a high sarcoplasmic reticulum load during diastole of the action potential. Moreover, this model successfully replicates other experimentally observed manifestations of Ca2+-wave-mediated triggered activity, including phase 2 and phase 3 early after-depolarizations and high-frequency voltage-Ca2+ oscillations. These oscillations feature an elevated take-off potential with depolarization mediated by the L-type Ca2+ current. The model also sheds light on the roles of luminal gating of RyRs and the mobile buffer ATP in the genesis of these arrhythmogenic phenomena. KEY POINTS: Existing mathematical models of Ca2+ waves use an excessively large Ca2+-release current or unrealistic diffusive coupling between release units. Our physiologically realistic model, using a Hill coefficient in the ryanodine receptor (RyR) gating function to represent RyR cooperativity, addresses these limitations and generates organized Ca2+ waves at Hill coefficients ranging from ∼5 to 10, as opposed to the traditional value of 2. This range of Hill coefficients gives a spark rate neither too low, thereby enabling Ca2+ wave propagation, nor too high, allowing for the maintenance of a high sarcoplasmic reticulum load during the plateau phase of the action potential. Additionally, the model generates Ca2+-wave-mediated phase 2 and phase 3 early after-depolarizations, and coupled membrane voltage with Ca2+ oscillations mediated by the L-type Ca2+ current. This study suggests that pharmacologically targeting RyR cooperativity could be a promising strategy for treating cardiac arrhythmias linked to Ca2+-wave-mediated triggered activity.
Collapse
Affiliation(s)
- Mingwang Zhong
- Physics Department and Center for Interdisciplinary Research in Complex Systems, Northeastern University, Boston, MA, USA
| | - Alain Karma
- Physics Department and Center for Interdisciplinary Research in Complex Systems, Northeastern University, Boston, MA, USA
| |
Collapse
|
2
|
Marabelli C, Santiago DJ, Priori SG. The Structural-Functional Crosstalk of the Calsequestrin System: Insights and Pathological Implications. Biomolecules 2023; 13:1693. [PMID: 38136565 PMCID: PMC10741413 DOI: 10.3390/biom13121693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/14/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Calsequestrin (CASQ) is a key intra-sarcoplasmic reticulum Ca2+-handling protein that plays a pivotal role in the contraction of cardiac and skeletal muscles. Its Ca2+-dependent polymerization dynamics shape the translation of electric excitation signals to the Ca2+-induced contraction of the actin-myosin architecture. Mutations in CASQ are linked to life-threatening pathological conditions, including tubular aggregate myopathy, malignant hyperthermia, and Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT). The variability in the penetrance of these phenotypes and the lack of a clear understanding of the disease mechanisms associated with CASQ mutations pose a major challenge to the development of effective therapeutic strategies. In vitro studies have mainly focused on the polymerization and Ca2+-buffering properties of CASQ but have provided little insight into the complex interplay of structural and functional changes that underlie disease. In this review, the biochemical and structural natures of CASQ are explored in-depth, while emphasizing their direct and indirect consequences for muscle Ca2+ physiology. We propose a novel functional classification of CASQ pathological missense mutations based on the structural stability of the monomer, dimer, or linear polymer conformation. We also highlight emerging similarities between polymeric CASQ and polyelectrolyte systems, emphasizing the potential for the use of this paradigm to guide further research.
Collapse
Affiliation(s)
- Chiara Marabelli
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy;
- Laboratory of Molecular Cardiology, IRCCS ICS Maugeri, 27100 Pavia, Italy
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain;
| | - Demetrio J. Santiago
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain;
| | - Silvia G. Priori
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy;
- Laboratory of Molecular Cardiology, IRCCS ICS Maugeri, 27100 Pavia, Italy
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain;
| |
Collapse
|
3
|
Ma J, Pang X, Wang T, Ning M, Liang Y, Li X, Tian X, Mo Y, Laher I, Li S. Acute aerobic exercise regulation of myocardial calcium homeostasis involves CASQ1, CASQ2, and TRDN. J Appl Physiol (1985) 2023; 135:707-716. [PMID: 37589058 DOI: 10.1152/japplphysiol.00299.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/20/2023] [Accepted: 08/10/2023] [Indexed: 08/18/2023] Open
Abstract
Exercise maintains cardiac calcium homeostasis and promotes cardiovascular health. This study explored temporal changes of calcium-related myocardial transcriptome changes during the recovery phase following a single bout of moderate-intensity aerobic exercise. Healthy male Sprague-Dawley rats were anesthetized with sodium pentobarbital after moderate-intensity aerobic exercise at four time points (0, 12, 24, and 72 h postexercise). The hearts were removed and RNA-seq and bioinformatics analyses were used to examine temporal transcriptional changes in the myocardium. Casq1, Casq2, and Trdn were identified as key genes in the regulation of calcium homeostasis during myocardial recovery. The highest expression of Casq1, Casq2, and Trdn genes and the proteins they encode occurred 24 h after exercise. An in vitro calcium overload heart model using the Langendorff heart perfusion method was used to examine myocardial calcium buffering capacity. Calcium overload caused the least changes in left ventricular developed pressure, infarct area, Lactate dehydrogenase release, and extent of morphological damage to myocardial cells, with the highest protein expressions of CASQ1, CASQ2, and TRDN at 24 h after acute exercise. This study indicates that maximal myocardial Ca2+ buffering capacity occurs 24 h postexercise in rats. Our study provides insights into exercise-mediated improvements in cardiovascular function and exercise preconditioning.NEW & NOTEWORTHY Acute aerobic exercise upregulates myocardial Casq1, Casq2, and Trdn genes and the proteins they encode in rats. Higher protein levels of CASQ1, CASQ2, and TRDN conferred an improved ability of the myocardium to resist calcium overload. Furthermore, 24 h postexercise is the time point with optimal myocardial calcium buffer capacity.
Collapse
Affiliation(s)
- Jiacheng Ma
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, People's Republic of China
| | - Xiaoli Pang
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, People's Republic of China
| | - Tutu Wang
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, People's Republic of China
| | - Miaomiao Ning
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, People's Republic of China
| | - Yu Liang
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, People's Republic of China
| | - Xiaole Li
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, People's Republic of China
| | - Xinyu Tian
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, People's Republic of China
| | - Yurou Mo
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, People's Republic of China
| | - Ismail Laher
- Department of Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shunchang Li
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, People's Republic of China
| |
Collapse
|
4
|
Eisner D, Neher E, Taschenberger H, Smith G. Physiology of intracellular calcium buffering. Physiol Rev 2023; 103:2767-2845. [PMID: 37326298 PMCID: PMC11550887 DOI: 10.1152/physrev.00042.2022] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/08/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023] Open
Abstract
Calcium signaling underlies much of physiology. Almost all the Ca2+ in the cytoplasm is bound to buffers, with typically only ∼1% being freely ionized at resting levels in most cells. Physiological Ca2+ buffers include small molecules and proteins, and experimentally Ca2+ indicators will also buffer calcium. The chemistry of interactions between Ca2+ and buffers determines the extent and speed of Ca2+ binding. The physiological effects of Ca2+ buffers are determined by the kinetics with which they bind Ca2+ and their mobility within the cell. The degree of buffering depends on factors such as the affinity for Ca2+, the Ca2+ concentration, and whether Ca2+ ions bind cooperatively. Buffering affects both the amplitude and time course of cytoplasmic Ca2+ signals as well as changes of Ca2+ concentration in organelles. It can also facilitate Ca2+ diffusion inside the cell. Ca2+ buffering affects synaptic transmission, muscle contraction, Ca2+ transport across epithelia, and the killing of bacteria. Saturation of buffers leads to synaptic facilitation and tetanic contraction in skeletal muscle and may play a role in inotropy in the heart. This review focuses on the link between buffer chemistry and function and how Ca2+ buffering affects normal physiology and the consequences of changes in disease. As well as summarizing what is known, we point out the many areas where further work is required.
Collapse
Affiliation(s)
- David Eisner
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Erwin Neher
- Membrane Biophysics Laboratory, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Holger Taschenberger
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Godfrey Smith
- School of Cardiovascular and Metabolic Health, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
5
|
Magyar ZÉ, Bauer J, Bauerová-Hlinková V, Jóna I, Gaburjakova J, Gaburjakova M, Almássy J. Eu 3+ detects two functionally distinct luminal Ca 2+ binding sites in ryanodine receptors. Biophys J 2023; 122:3516-3531. [PMID: 37533257 PMCID: PMC10502479 DOI: 10.1016/j.bpj.2023.07.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/26/2023] [Accepted: 07/31/2023] [Indexed: 08/04/2023] Open
Abstract
Ryanodine receptors (RyRs) are Ca2+ release channels, gated by Ca2+ in the cytosol and the sarcoplasmic reticulum lumen. Their regulation is impaired in certain cardiac and muscle diseases. Although a lot of data is available on the luminal Ca2+ regulation of RyR, its interpretation is complicated by the possibility that the divalent ions used to probe the luminal binding sites may contaminate the cytoplasmic sites by crossing the channel pore. In this study, we used Eu3+, an impermeable agonist of Ca2+ binding sites, as a probe to avoid this complication and to gain more specific information about the function of the luminal Ca2+ sensor. Single-channel currents were measured from skeletal muscle and cardiac RyRs (RyR1 and RyR2) using the lipid bilayer technique. We show that RyR2 is activated by the luminal addition of Ca2+, whereas RyR1 is inhibited. These results were qualitatively reproducible using Eu3+. The luminal regulation of RyR1 carrying a mutation associated with malignant hyperthermia was not different from that of the wild-type. RyR1 inhibition by Eu3+ was extremely voltage dependent, whereas RyR2 activation did not depend on the membrane potential. These results suggest that the RyR1 inhibition site is in the membrane's electric field (channel pore), whereas the RyR2 activation site is outside. Using in silico analysis and previous results, we predicted putative Ca2+ binding site sequences. We propose that RyR2 bears an activation site, which is missing in RyR1, but both isoforms share the same inhibitory Ca2+ binding site near the channel gate.
Collapse
Affiliation(s)
- Zsuzsanna É Magyar
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Jacob Bauer
- Institute of Molecular Biology, Slovak Academy of Sciences, Bratislava, Slovakia
| | | | - István Jóna
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Jana Gaburjakova
- Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Marta Gaburjakova
- Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia
| | - János Almássy
- Department of Physiology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
6
|
Zhang X, Smith CER, Morotti S, Edwards AG, Sato D, Louch WE, Ni H, Grandi E. Mechanisms of spontaneous Ca 2+ release-mediated arrhythmia in a novel 3D human atrial myocyte model: II. Ca 2+ -handling protein variation. J Physiol 2023; 601:2685-2710. [PMID: 36114707 PMCID: PMC10017376 DOI: 10.1113/jp283602] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/02/2022] [Indexed: 11/08/2022] Open
Abstract
Disruption of the transverse-axial tubule system (TATS) in diseases such as heart failure and atrial fibrillation occurs in combination with changes in the expression and distribution of key Ca2+ -handling proteins. Together this ultrastructural and ionic remodelling is associated with aberrant Ca2+ cycling and electrophysiological instabilities that underlie arrhythmic activity. However, due to the concurrent changes in TATs and Ca2+ -handling protein expression and localization that occur in disease it is difficult to distinguish their individual contributions to the arrhythmogenic state. To investigate this, we applied our novel 3D human atrial myocyte model with spatially detailed Ca2+ diffusion and TATS to investigate the isolated and interactive effects of changes in expression and localization of key Ca2+ -handling proteins and variable TATS density on Ca2+ -handling abnormality driven membrane instabilities. We show that modulating the expression and distribution of the sodium-calcium exchanger, ryanodine receptors and the sarcoplasmic reticulum (SR) Ca2+ buffer calsequestrin have varying pro- and anti-arrhythmic effects depending on the balance of opposing influences on SR Ca2+ leak-load and Ca2+ -voltage relationships. Interestingly, the impact of protein remodelling on Ca2+ -driven proarrhythmic behaviour varied dramatically depending on TATS density, with intermediately tubulated cells being more severely affected compared to detubulated and densely tubulated myocytes. This work provides novel mechanistic insight into the distinct and interactive consequences of TATS and Ca2+ -handling protein remodelling that underlies dysfunctional Ca2+ cycling and electrophysiological instability in disease. KEY POINTS: In our companion paper we developed a 3D human atrial myocyte model, coupling electrophysiology and Ca2+ handling with subcellular spatial details governed by the transverse-axial tubule system (TATS). Here we utilize this model to mechanistically examine the impact of TATS loss and changes in the expression and distribution of key Ca2+ -handling proteins known to be remodelled in disease on Ca2+ homeostasis and electrophysiological stability. We demonstrate that varying the expression and localization of these proteins has variable pro- and anti-arrhythmic effects with outcomes displaying dependence on TATS density. Whereas detubulated myocytes typically appear unaffected and densely tubulated cells seem protected, the arrhythmogenic effects of Ca2+ handling protein remodelling are profound in intermediately tubulated cells. Our work shows the interaction between TATS and Ca2+ -handling protein remodelling that underlies the Ca2+ -driven proarrhythmic behaviour observed in atrial fibrillation and may help to predict the effects of antiarrhythmic strategies at varying stages of ultrastructural remodelling.
Collapse
Affiliation(s)
- Xianwei Zhang
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | | | - Stefano Morotti
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | | | - Daisuke Sato
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Haibo Ni
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - Eleonora Grandi
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| |
Collapse
|
7
|
Díaz del Moral S, Benaouicha M, Villa del Campo C, Torres M, Wagner N, Wagner KD, Muñoz-Chápuli R, Carmona R. Cardiomyocyte-Specific Wt1 Is Involved in Cardiac Metabolism and Response to Damage. J Cardiovasc Dev Dis 2023; 10:211. [PMID: 37233178 PMCID: PMC10219250 DOI: 10.3390/jcdd10050211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/23/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023] Open
Abstract
The Wilms tumor suppressor gene (Wt1) encodes a C2H2-type zinc-finger transcription factor that participates in transcriptional regulation, RNA metabolism, and protein-protein interactions. WT1 is involved in the development of several organs, including the kidneys and gonads, heart, spleen, adrenal glands, liver, diaphragm, and neuronal system. We previously provided evidence of transient WT1 expression in about 25% of cardiomyocytes of mouse embryos. Conditional deletion of Wt1 in the cardiac troponin T lineage caused abnormal cardiac development. A low expression of WT1 has also been reported in adult cardiomyocytes. Therefore, we aimed to explore its function in cardiac homeostasis and in the response to pharmacologically induced damage. Silencing of Wt1 in cultured neonatal murine cardiomyocytes provoked alterations in mitochondrial membrane potential and changes in the expression of genes related to calcium homeostasis. Ablation of WT1 in adult cardiomyocytes by crossing αMHCMerCreMer mice with homozygous WT1-floxed mice induced hypertrophy, interstitial fibrosis, altered metabolism, and mitochondrial dysfunction. In addition, conditional deletion of WT1 in adult cardiomyocytes increased doxorubicin-induced damage. These findings suggest a novel role of WT1 in myocardial physiology and protection against damage.
Collapse
Affiliation(s)
- Sandra Díaz del Moral
- Department of Animal Biology, Faculty of Science, University of Málaga, 29071 Málaga, Spain; (S.D.d.M.); (R.M.-C.)
| | - Maha Benaouicha
- Department of Cell Biology, Genetics and Physiology, Faculty of Science, University of Málaga, 29071 Málaga, Spain;
| | - Cristina Villa del Campo
- Cardiovascular Development Program, Centro Nacional de Investigaciones Cardiovasculares, CNIC, 28029 Madrid, Spain; (C.V.d.C.); (M.T.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Miguel Torres
- Cardiovascular Development Program, Centro Nacional de Investigaciones Cardiovasculares, CNIC, 28029 Madrid, Spain; (C.V.d.C.); (M.T.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Nicole Wagner
- Université Côte d’Azur, CNRS, INSERM, iBV, 06108 Nice, France; (N.W.); (K.-D.W.)
| | - Kay-Dietrich Wagner
- Université Côte d’Azur, CNRS, INSERM, iBV, 06108 Nice, France; (N.W.); (K.-D.W.)
| | - Ramón Muñoz-Chápuli
- Department of Animal Biology, Faculty of Science, University of Málaga, 29071 Málaga, Spain; (S.D.d.M.); (R.M.-C.)
| | - Rita Carmona
- Department of Human Anatomy, Legal Medicine and History of Science, Faculty of Medicine, University of Málaga, 29071 Málaga, Spain
| |
Collapse
|
8
|
Moore J, Ewoldt J, Venturini G, Pereira AC, Padilha K, Lawton M, Lin W, Goel R, Luptak I, Perissi V, Seidman CE, Seidman J, Chin MT, Chen C, Emili A. Multi-Omics Profiling of Hypertrophic Cardiomyopathy Reveals Altered Mechanisms in Mitochondrial Dynamics and Excitation-Contraction Coupling. Int J Mol Sci 2023; 24:4724. [PMID: 36902152 PMCID: PMC10002553 DOI: 10.3390/ijms24054724] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Hypertrophic cardiomyopathy is one of the most common inherited cardiomyopathies and a leading cause of sudden cardiac death in young adults. Despite profound insights into the genetics, there is imperfect correlation between mutation and clinical prognosis, suggesting complex molecular cascades driving pathogenesis. To investigate this, we performed an integrated quantitative multi-omics (proteomic, phosphoproteomic, and metabolomic) analysis to illuminate the early and direct consequences of mutations in myosin heavy chain in engineered human induced pluripotent stem-cell-derived cardiomyocytes relative to late-stage disease using patient myectomies. We captured hundreds of differential features, which map to distinct molecular mechanisms modulating mitochondrial homeostasis at the earliest stages of pathobiology, as well as stage-specific metabolic and excitation-coupling maladaptation. Collectively, this study fills in gaps from previous studies by expanding knowledge of the initial responses to mutations that protect cells against the early stress prior to contractile dysfunction and overt disease.
Collapse
Affiliation(s)
- Jarrod Moore
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jourdan Ewoldt
- Department of Biomedical Engineering, Boston University, Boston, MA 02218, USA
| | | | | | - Kallyandra Padilha
- Laboratory of Genetics and Molecular Cardiology, Clinical Hospital, Faculty of Medicine, University of São Paulo, Sao Paulo 05508-000, Brazil
| | - Matthew Lawton
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Weiwei Lin
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Raghuveera Goel
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Ivan Luptak
- Myocardial Biology Unit, Boston University School of Medicine, Boston, MA 02118, USA
| | - Valentina Perissi
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Christine E. Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Jonathan Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Michael T. Chin
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02145, USA
| | - Christopher Chen
- Department of Biomedical Engineering, Boston University, Boston, MA 02218, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Andrew Emili
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
9
|
Quantification of the calcium signaling deficit in muscles devoid of triadin. PLoS One 2022; 17:e0264146. [PMID: 35213584 PMCID: PMC8880904 DOI: 10.1371/journal.pone.0264146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 02/03/2022] [Indexed: 11/24/2022] Open
Abstract
Triadin, a protein of the sarcoplasmic reticulum (SR) of striated muscles, anchors the calcium-storing protein calsequestrin to calcium release RyR channels at the junction with t-tubules, and modulates these channels by conformational effects. Triadin ablation induces structural SR changes and alters the expression of other proteins. Here we quantify alterations of calcium signaling in single skeletal myofibers of constitutive triadin-null mice. We find higher resting cytosolic and lower SR-luminal [Ca2+], 40% lower calsequestrin expression, and more CaV1.1, RyR1 and SERCA1. Despite the increased CaV1.1, the mobile intramembrane charge was reduced by ~20% in Triadin-null fibers. The initial peak of calcium release flux by pulse depolarization was minimally altered in the null fibers (revealing an increase in peak calcium permeability). The “hump” phase that followed, attributable to calcium detaching from calsequestrin, was 25% lower, a smaller change than expected from the reduced calsequestrin content and calcium saturation. The exponential decay rate of calcium transients was 25% higher, consistent with the higher SERCA1 content. Recovery of calcium flux after a depleting depolarization was faster in triadin-null myofibers, consistent with the increased uptake rate and lower SR calsequestrin content. In sum, the triadin knockout determines an increased RyR1 channel openness, which depletes the SR, a substantial loss of calsequestrin and gains in other couplon proteins. Powerful functional compensations ensue: activation of SOCE that increases [Ca2+]cyto; increased SERCA1 activity, which limits the decrease in [Ca2+]SR and a restoration of SR calcium storage of unknown substrate. Together, they effectively limit the functional loss in skeletal muscles.
Collapse
|
10
|
The function and regulation of calsequestrin-2: implications in calcium-mediated arrhythmias. Biophys Rev 2022; 14:329-352. [PMID: 35340602 PMCID: PMC8921388 DOI: 10.1007/s12551-021-00914-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/14/2021] [Indexed: 01/09/2023] Open
Abstract
Cardiac arrhythmias are life-threatening events in which the heart develops an irregular rhythm. Mishandling of Ca2+ within the myocytes of the heart has been widely demonstrated to be an underlying mechanism of arrhythmogenesis. This includes altered function of the ryanodine receptor (RyR2)-the primary Ca2+ release channel located to the sarcoplasmic reticulum (SR). The spontaneous leak of SR Ca2+ via RyR2 is a well-established contributor in the development of arrhythmic contractions. This leak is associated with increased channel activity in response to changes in SR Ca2+ load. RyR2 activity can be regulated through several avenues, including interactions with numerous accessory proteins. One such protein is calsequestrin-2 (CSQ2), which is the primary Ca2+-buffering protein within the SR. The capacity of CSQ2 to buffer Ca2+ is tightly associated with the ability of the protein to polymerise in response to changing Ca2+ levels. CSQ2 can itself be regulated through phosphorylation and glycosylation modifications, which impact protein polymerisation and trafficking. Changes in CSQ2 modifications are implicated in cardiac pathologies, while mutations in CSQ2 have been identified in arrhythmic patients. Here, we review the role of CSQ2 in arrhythmogenesis including evidence for the indirect and direct regulation of RyR2 by CSQ2, and the consequences of a loss of functional CSQ2 in Ca2+ homeostasis and Ca2+-mediated arrhythmias. Supplementary Information The online version contains supplementary material available at 10.1007/s12551-021-00914-6.
Collapse
|
11
|
Hoang-Trong TM, Ullah A, Lederer WJ, Jafri MS. A Stochastic Spatiotemporal Model of Rat Ventricular Myocyte Calcium Dynamics Demonstrated Necessary Features for Calcium Wave Propagation. MEMBRANES 2021; 11:989. [PMID: 34940490 PMCID: PMC8706945 DOI: 10.3390/membranes11120989] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/12/2021] [Accepted: 12/15/2021] [Indexed: 11/16/2022]
Abstract
Calcium (Ca2+) plays a central role in the excitation and contraction of cardiac myocytes. Experiments have indicated that calcium release is stochastic and regulated locally suggesting the possibility of spatially heterogeneous calcium levels in the cells. This spatial heterogeneity might be important in mediating different signaling pathways. During more than 50 years of computational cell biology, the computational models have been advanced to incorporate more ionic currents, going from deterministic models to stochastic models. While periodic increases in cytoplasmic Ca2+ concentration drive cardiac contraction, aberrant Ca2+ release can underly cardiac arrhythmia. However, the study of the spatial role of calcium ions has been limited due to the computational expense of using a three-dimensional stochastic computational model. In this paper, we introduce a three-dimensional stochastic computational model for rat ventricular myocytes at the whole-cell level that incorporate detailed calcium dynamics, with (1) non-uniform release site placement, (2) non-uniform membrane ionic currents and membrane buffers, (3) stochastic calcium-leak dynamics and (4) non-junctional or rogue ryanodine receptors. The model simulates spark-induced spark activation and spark-induced Ca2+ wave initiation and propagation that occur under conditions of calcium overload at the closed-cell condition, but not when Ca2+ levels are normal. This is considered important since the presence of Ca2+ waves contribute to the activation of arrhythmogenic currents.
Collapse
Affiliation(s)
- Tuan Minh Hoang-Trong
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA 22030, USA; (T.M.H.-T.); (A.U.)
| | - Aman Ullah
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA 22030, USA; (T.M.H.-T.); (A.U.)
| | - William Jonathan Lederer
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Mohsin Saleet Jafri
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA 22030, USA; (T.M.H.-T.); (A.U.)
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| |
Collapse
|
12
|
Sleiman Y, Lacampagne A, Meli AC. "Ryanopathies" and RyR2 dysfunctions: can we further decipher them using in vitro human disease models? Cell Death Dis 2021; 12:1041. [PMID: 34725342 PMCID: PMC8560800 DOI: 10.1038/s41419-021-04337-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/08/2021] [Accepted: 10/14/2021] [Indexed: 12/23/2022]
Abstract
The regulation of intracellular calcium (Ca2+) homeostasis is fundamental to maintain normal functions in many cell types. The ryanodine receptor (RyR), the largest intracellular calcium release channel located on the sarco/endoplasmic reticulum (SR/ER), plays a key role in the intracellular Ca2+ handling. Abnormal type 2 ryanodine receptor (RyR2) function, associated to mutations (ryanopathies) or pathological remodeling, has been reported, not only in cardiac diseases, but also in neuronal and pancreatic disorders. While animal models and in vitro studies provided valuable contributions to our knowledge on RyR2 dysfunctions, the human cell models derived from patients’ cells offer new hope for improving our understanding of human clinical diseases and enrich the development of great medical advances. We here discuss the current knowledge on RyR2 dysfunctions associated with mutations and post-translational remodeling. We then reviewed the novel human cellular technologies allowing the correlation of patient’s genome with their cellular environment and providing approaches for personalized RyR-targeted therapeutics.
Collapse
Affiliation(s)
- Yvonne Sleiman
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Alain Lacampagne
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Albano C Meli
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France.
| |
Collapse
|
13
|
Filomena MC, Yamamoto DL, Carullo P, Medvedev R, Ghisleni A, Piroddi N, Scellini B, Crispino R, D'Autilia F, Zhang J, Felicetta A, Nemska S, Serio S, Tesi C, Catalucci D, Linke WA, Polishchuk R, Poggesi C, Gautel M, Bang ML. Myopalladin knockout mice develop cardiac dilation and show a maladaptive response to mechanical pressure overload. eLife 2021; 10:e58313. [PMID: 34558411 PMCID: PMC8547954 DOI: 10.7554/elife.58313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 09/23/2021] [Indexed: 12/13/2022] Open
Abstract
Myopalladin (MYPN) is a striated muscle-specific immunoglobulin domain-containing protein located in the sarcomeric Z-line and I-band. MYPN gene mutations are causative for dilated (DCM), hypertrophic, and restrictive cardiomyopathy. In a yeast two-hybrid screening, MYPN was found to bind to titin in the Z-line, which was confirmed by microscale thermophoresis. Cardiac analyses of MYPN knockout (MKO) mice showed the development of mild cardiac dilation and systolic dysfunction, associated with decreased myofibrillar isometric tension generation and increased resting tension at longer sarcomere lengths. MKO mice exhibited a normal hypertrophic response to transaortic constriction (TAC), but rapidly developed severe cardiac dilation and systolic dysfunction, associated with fibrosis, increased fetal gene expression, higher intercalated disc fold amplitude, decreased calsequestrin-2 protein levels, and increased desmoplakin and SORBS2 protein levels. Cardiomyocyte analyses showed delayed Ca2+ release and reuptake in unstressed MKO mice as well as reduced Ca2+ spark amplitude post-TAC, suggesting that altered Ca2+ handling may contribute to the development of DCM in MKO mice.
Collapse
Affiliation(s)
- Maria Carmela Filomena
- Institute of Genetic and Biomedical Research (IRGB) - National Research Council (CNR), Milan unitMilanItaly
- IRCCS Humanitas Research HospitalMilanItaly
| | - Daniel L Yamamoto
- Institute of Genetic and Biomedical Research (IRGB) - National Research Council (CNR), Milan unitMilanItaly
| | - Pierluigi Carullo
- Institute of Genetic and Biomedical Research (IRGB) - National Research Council (CNR), Milan unitMilanItaly
- IRCCS Humanitas Research HospitalMilanItaly
| | - Roman Medvedev
- IRCCS Humanitas Research HospitalMilanItaly
- Department of Cardiac Surgery, University of VeronaVeronaItaly
| | - Andrea Ghisleni
- Randall Centre for Cell and Molecular Biophysics, King's College London BHF Centre of Research ExcellenceLondonUnited Kingdom
| | - Nicoletta Piroddi
- Department of Experimental and Clinical Medicine, University of FlorenceFlorenceItaly
| | - Beatrice Scellini
- Department of Experimental and Clinical Medicine, University of FlorenceFlorenceItaly
| | - Roberta Crispino
- Telethon Institute of Genetics and Medicine (TIGEM)PozzuoliItaly
| | | | - Jianlin Zhang
- Department of Medicine, University of California, San DiegoLa JollaUnited States
| | - Arianna Felicetta
- IRCCS Humanitas Research HospitalMilanItaly
- Humanitas UniversityPieve EmanueleItaly
| | | | | | - Chiara Tesi
- Department of Experimental and Clinical Medicine, University of FlorenceFlorenceItaly
| | | | - Wolfgang A Linke
- Institute of Physiology II, University of MuensterMuensterGermany
| | - Roman Polishchuk
- Telethon Institute of Genetics and Medicine (TIGEM)PozzuoliItaly
| | - Corrado Poggesi
- Department of Experimental and Clinical Medicine, University of FlorenceFlorenceItaly
| | - Mathias Gautel
- Randall Centre for Cell and Molecular Biophysics, King's College London BHF Centre of Research ExcellenceLondonUnited Kingdom
| | - Marie-Louise Bang
- Institute of Genetic and Biomedical Research (IRGB) - National Research Council (CNR), Milan unitMilanItaly
- IRCCS Humanitas Research HospitalMilanItaly
| |
Collapse
|
14
|
Cely-Ortiz A, Felice JI, Díaz-Zegarra LA, Valverde CA, Federico M, Palomeque J, Wehrens XHT, Kranias EG, Aiello EA, Lascano EC, Negroni JA, Mattiazzi A. Determinants of Ca2+ release restitution: Insights from genetically altered animals and mathematical modeling. J Gen Physiol 2021; 152:152125. [PMID: 32986800 PMCID: PMC7594441 DOI: 10.1085/jgp.201912512] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 07/27/2020] [Accepted: 08/21/2020] [Indexed: 01/07/2023] Open
Abstract
Each heartbeat is followed by a refractory period. Recovery from refractoriness is known as Ca2+ release restitution (CRR), and its alterations are potential triggers of Ca2+ arrhythmias. Although the control of CRR has been associated with SR Ca2+ load and RYR2 Ca2+ sensitivity, the relative role of some of the determinants of CRR remains largely undefined. An intriguing point, difficult to dissect and previously neglected, is the possible independent effect of SR Ca2+ content versus the velocity of SR Ca2+ refilling on CRR. To assess these interrogations, we used isolated myocytes with phospholamban (PLN) ablation (PLNKO), knock-in mice with pseudoconstitutive CaMKII phosphorylation of RYR2 S2814 (S2814D), S2814D crossed with PLNKO mice (SDKO), and a previously validated human cardiac myocyte model. Restitution of cytosolic Ca2+ (Fura-2 AM) and L-type calcium current (ICaL; patch-clamp) was evaluated with a two-pulse (S1/S2) protocol. CRR and ICaL restitution increased as a function of the (S2-S1) coupling interval, following an exponential curve. When SR Ca2+ load was increased by increasing extracellular [Ca2+] from 2.0 to 4.0 mM, CRR and ICaL restitution were enhanced, suggesting that ICaL restitution may contribute to the faster CRR observed at 4.0 mM [Ca2+]. In contrast, ICaL restitution did not differ among the different mouse models. For a given SR Ca2+ load, CRR was accelerated in S2814D myocytes versus WT, but not in PLNKO and SDKO myocytes versus WT and S2814D, respectively. The model mimics all experimental data. Moreover, when the PLN ablation-induced decrease in RYR2 expression was corrected, the model revealed that CRR was accelerated in PLNKO and SDKO versus WT and S2814D myocytes, consistent with the enhanced velocity of refilling, SR [Ca2+] recovery, and CRR. We speculate that refilling rate might enhance CRR independently of SR Ca2+ load.
Collapse
Affiliation(s)
- Alejandra Cely-Ortiz
- Centro de Investigaciones Cardiovasculares, Centro Científico Tecnológico-La Plata, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Juan I Felice
- Centro de Investigaciones Cardiovasculares, Centro Científico Tecnológico-La Plata, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Leandro A Díaz-Zegarra
- Centro de Investigaciones Cardiovasculares, Centro Científico Tecnológico-La Plata, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Carlos A Valverde
- Centro de Investigaciones Cardiovasculares, Centro Científico Tecnológico-La Plata, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Marilén Federico
- Centro de Investigaciones Cardiovasculares, Centro Científico Tecnológico-La Plata, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Julieta Palomeque
- Centro de Investigaciones Cardiovasculares, Centro Científico Tecnológico-La Plata, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Xander H T Wehrens
- Departments of Molecular Physiology and Biophysics, Medicine (in Cardiology), Neuroscience, Pediatrics, Center for Space Medicine, Baylor College of Medicine, Cardiovascular Research Institute, Houston, TX
| | - Evangelia G Kranias
- Department of Pharmacology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Ernesto A Aiello
- Centro de Investigaciones Cardiovasculares, Centro Científico Tecnológico-La Plata, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Elena C Lascano
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Favaloro, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Jorge A Negroni
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Favaloro, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Alicia Mattiazzi
- Centro de Investigaciones Cardiovasculares, Centro Científico Tecnológico-La Plata, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| |
Collapse
|
15
|
The structure-based cancer-related single amino acid variation prediction. Sci Rep 2021; 11:13599. [PMID: 34193921 PMCID: PMC8245468 DOI: 10.1038/s41598-021-92793-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/16/2021] [Indexed: 11/09/2022] Open
Abstract
Single amino acid variation (SAV) is an amino acid substitution of the protein sequence that can potentially influence the entire protein structure or function, as well as its binding affinity. Protein destabilization is related to diseases, including several cancers, although using traditional experiments to clarify the relationship between SAVs and cancer uses much time and resources. Some SAV prediction methods use computational approaches, with most predicting SAV-induced changes in protein stability. In this investigation, all SAV characteristics generated from protein sequences, structures and the microenvironment were converted into feature vectors and fed into an integrated predicting system using a support vector machine and genetic algorithm. Critical features were used to estimate the relationship between their properties and cancers caused by SAVs. We describe how we developed a prediction system based on protein sequences and structure that is capable of distinguishing if the SAV is related to cancer or not. The five-fold cross-validation performance of our system is 89.73% for the accuracy, 0.74 for the Matthews correlation coefficient, and 0.81 for the F1 score. We have built an online prediction server, CanSavPre ( http://bioinfo.cmu.edu.tw/CanSavPre/ ), which is expected to become a useful, practical tool for cancer research and precision medicine.
Collapse
|
16
|
Precision Medicine in Catecholaminergic Polymorphic Ventricular Tachycardia: JACC Focus Seminar 5/5. J Am Coll Cardiol 2021; 77:2592-2612. [PMID: 34016269 DOI: 10.1016/j.jacc.2020.12.073] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/14/2020] [Accepted: 12/21/2020] [Indexed: 11/20/2022]
Abstract
In this final of a 5-part Focus Seminar series on precision medicine, we focus on catecholaminergic polymorphic ventricular tachycardia (CPVT). This focus on CPVT allows us to take a "deep dive" and explore the full extent of the precision medicine opportunities for a single cardiovascular condition at a level that was not possible in the preceding articles. As a new paradigm presented in this article, it has become clear that CPVT can occur as either a typical or atypical form. Although there is a degree of overlap between the typical and atypical forms, it is notable that they arise due to different underlying genetic changes, likely exhibiting differing mechanisms of action, and presenting with different phenotypic features. The recognition of these differing forms of CPVT and their different etiologies and mechanisms is an important step toward implementing rapidly emerging precision medicine approaches that will tailor novel therapies to specific gene defects.
Collapse
|
17
|
Pathways of calcium regulation, electron transport, and mitochondrial protein translation are molecular signatures of susceptibility to recurrent exertional rhabdomyolysis in Thoroughbred racehorses. PLoS One 2021; 16:e0244556. [PMID: 33566847 PMCID: PMC7875397 DOI: 10.1371/journal.pone.0244556] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/13/2020] [Indexed: 12/13/2022] Open
Abstract
Recurrent exertional rhabdomyolysis (RER) is a chronic muscle disorder of unknown etiology in racehorses. A potential role of intramuscular calcium (Ca2+) dysregulation in RER has led to the use of dantrolene to prevent episodes of rhabdomyolysis. We examined differentially expressed proteins (DEP) and gene transcripts (DEG) in gluteal muscle of Thoroughbred race-trained mares after exercise among three groups of 5 horses each; 1) horses susceptible to, but not currently experiencing rhabdomyolysis, 2) healthy horses with no history of RER (control), 3) RER-susceptible horses treated with dantrolene pre-exercise (RER-D). Tandem mass tag LC/MS/MS quantitative proteomics and RNA-seq analysis (FDR <0.05) was followed by gene ontology (GO) and semantic similarity of enrichment terms. Of the 375 proteins expressed, 125 were DEP in RER-susceptible versus control, with 52 ↑DEP mainly involving Ca2+ regulation (N = 11) (e.g. RYR1, calmodulin, calsequestrin, calpain), protein degradation (N = 6), antioxidants (N = 4), plasma membranes (N = 3), glyco(geno)lysis (N = 3) and 21 DEP being blood-borne. ↓DEP (N = 73) were largely mitochondrial (N = 45) impacting the electron transport system (28), enzymes (6), heat shock proteins (4), and contractile proteins (12) including Ca2+ binding proteins. There were 812 DEG in RER-susceptible versus control involving the electron transfer system, the mitochondrial transcription/translational response and notably the pro-apoptotic Ca2+-activated mitochondrial membrane transition pore (SLC25A27, BAX, ATP5 subunits). Upregulated mitochondrial DEG frequently had downregulation of their encoded DEP with semantic similarities highlighting signaling mechanisms regulating mitochondrial protein translation. RER-susceptible horses treated with dantrolene, which slows sarcoplasmic reticulum Ca2+ release, showed no DEG compared to control horses. We conclude that RER-susceptibility is associated with alterations in proteins, genes and pathways impacting myoplasmic Ca2+ regulation, the mitochondrion and protein degradation with opposing effects on mitochondrial transcriptional/translational responses and mitochondrial protein content. RER could potentially arise from excessive sarcoplasmic reticulum Ca2+ release and subsequent mitochondrial buffering of excessive myoplasmic Ca2+.
Collapse
|
18
|
Blatter LA, Kanaporis G, Martinez-Hernandez E, Oropeza-Almazan Y, Banach K. Excitation-contraction coupling and calcium release in atrial muscle. Pflugers Arch 2021; 473:317-329. [PMID: 33398498 DOI: 10.1007/s00424-020-02506-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/03/2020] [Accepted: 12/16/2020] [Indexed: 01/02/2023]
Abstract
In cardiac muscle, the process of excitation-contraction coupling (ECC) describes the chain of events that links action potential induced myocyte membrane depolarization, surface membrane ion channel activation, triggering of Ca2+ induced Ca2+ release from the sarcoplasmic reticulum (SR) Ca2+ store to activation of the contractile machinery that is ultimately responsible for the pump function of the heart. Here we review similarities and differences of structural and functional attributes of ECC between atrial and ventricular tissue. We explore a novel "fire-diffuse-uptake-fire" paradigm of atrial ECC and Ca2+ release that assigns a novel role to the SR SERCA pump and involves a concerted "tandem" activation of the ryanodine receptor Ca2+ release channel by cytosolic and luminal Ca2+. We discuss the contribution of the inositol 1,4,5-trisphosphate (IP3) receptor Ca2+ release channel as an auxiliary pathway to Ca2+ signaling, and we review IP3 receptor-induced Ca2+ release involvement in beat-to-beat ECC, nuclear Ca2+ signaling, and arrhythmogenesis. Finally, we explore the topic of electromechanical and Ca2+ alternans and its ramifications for atrial arrhythmia.
Collapse
Affiliation(s)
- L A Blatter
- Department of Physiology & Biophysics, Rush University Medical Center, 1750 W. Harrison Street, Chicago, IL, 60612, USA.
| | - G Kanaporis
- Department of Physiology & Biophysics, Rush University Medical Center, 1750 W. Harrison Street, Chicago, IL, 60612, USA
| | - E Martinez-Hernandez
- Department of Physiology & Biophysics, Rush University Medical Center, 1750 W. Harrison Street, Chicago, IL, 60612, USA
| | - Y Oropeza-Almazan
- Department of Physiology & Biophysics, Rush University Medical Center, 1750 W. Harrison Street, Chicago, IL, 60612, USA
| | - K Banach
- Department of Internal Medicine/Cardiology, Rush University Medical Center, Chicago, IL, 60612, USA
| |
Collapse
|
19
|
Zhang H, Fu Q, Shi X, Pan Z, Yang W, Huang Z, Tang T, He X, Zhang R. Human A-to-I RNA editing SNP loci are enriched in GWAS signals for autoimmune diseases and under balancing selection. Genome Biol 2020; 21:288. [PMID: 33256812 PMCID: PMC7702712 DOI: 10.1186/s13059-020-02205-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Adenosine-to-inosine (A-to-I) RNA editing plays important roles in diversifying the transcriptome and preventing MDA5 sensing of endogenous dsRNA as nonself. To date, few studies have investigated the population genomic signatures of A-to-I editing due to the lack of editing sites overlapping with SNPs. RESULTS In this study, we applied a pipeline to robustly identify SNP editing sites from population transcriptomic data and combined functional genomics, GWAS, and population genomics approaches to study the function and evolution of A-to-I editing. We find that the G allele, which is equivalent to edited I, is overrepresented in editing SNPs. Functionally, A/G editing SNPs are highly enriched in GWAS signals of autoimmune and immune-related diseases. Evolutionarily, derived allele frequency distributions of A/G editing SNPs for both A and G alleles as the ancestral alleles are skewed toward intermediate frequency alleles relative to neutral SNPs, a hallmark of balancing selection, suggesting that both A and G alleles are functionally important. The signal of balancing selection is confirmed by a number of additional population genomic analyses. CONCLUSIONS We uncovered a hidden layer of A-to-I RNA editing SNP loci as a common target of balancing selection, and we propose that the maintenance of such editing SNP variations may be at least partially due to constraints on the resolution of the balance between immune activity and self-tolerance.
Collapse
Affiliation(s)
- Hui Zhang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, People's Republic of China
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, People's Republic of China
| | - Qiang Fu
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Xinrui Shi
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Ziqing Pan
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Wenbing Yang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Zichao Huang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Tian Tang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Xionglei He
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Rui Zhang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, People's Republic of China.
- RNA Biomedical Institute, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
20
|
Bodin A, Labas V, Bisson A, Teixeira-Gomes AP, Blasco H, Tomas D, Combes-Soia L, Marcelo P, Miquelestorena-Standley E, Baron C, Angoulvant D, Babuty D, Clementy N. Acute pathophysiological myocardial changes following intra-cardiac electrical shocks using a proteomic approach in a sheep model. Sci Rep 2020; 10:20252. [PMID: 33219330 PMCID: PMC7679418 DOI: 10.1038/s41598-020-77346-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 11/04/2020] [Indexed: 12/18/2022] Open
Abstract
Implantable cardioverter-defibrillators (ICD) are meant to fight life-threatening ventricular arrhythmias and reduce overall mortality. Ironically, life-saving shocks themselves have been shown to be independently associated with an increased mortality. We sought to identify myocardial changes at the protein level immediately after ICD electrical shocks using a proteomic approach. ICD were surgically implanted in 10 individuals of a healthy male sheep model: a control group (N = 5) without any shock delivery and a shock group (N = 5) with the delivery of 5 consecutive shocks at 41 J. Myocardial tissue samples were collected at the right-ventricle apex near to the lead coil and at the right ventricle basal free wall region. Global quantitative proteomics experiments on myocardial tissue samples were performed using mass spectrometry techniques. Proteome was significantly modified after electrical shock and several mechanisms were associated: protein, DNA and membrane damages due to extreme physical conditions induced by ICD-shock but also due to regulated cell death; metabolic remodeling; oxidative stress; calcium dysregulation; inflammation and fibrosis. These proteome modifications were seen in myocardium both "near" and "far" from electrical shock region. N-term acetylated troponin C was an interesting tissular biomarker, significantly decreased after electrical shock in the "far" region (AUC: 0.93). Our data support an acute shock-induced myocardial tissue injury which might be involved in acute paradoxical deleterious effects such as heart failure and ventricular arrhythmias.
Collapse
Affiliation(s)
- Alexandre Bodin
- Service de Cardiologie, Centre Hospitalier Universitaire Trousseau Et EA7505, Faculté de Médecine, Université François Rabelais, Tours, France.
- INRAE, CNRS, IFCE, UMR PRC, Université de Tours, 37380, Nouzilly, France.
| | - Valérie Labas
- INRAE, CNRS, IFCE, UMR PRC, Université de Tours, 37380, Nouzilly, France
- INRAE, CHU de Tours, Plate-Forme de Chirurgie Et D'Imagerie Pour La Recherche Et L'Enseignement, Université de Tours, 37380, Nouzilly, France
| | - Arnaud Bisson
- Service de Cardiologie, Centre Hospitalier Universitaire Trousseau Et EA7505, Faculté de Médecine, Université François Rabelais, Tours, France
| | | | - Hélène Blasco
- Imagerie Et Cerveau - UMR 1253, Université de Tours, Tours, France
| | - Daniel Tomas
- INRAE, CNRS, IFCE, UMR PRC, Université de Tours, 37380, Nouzilly, France
- INRAE, CHU de Tours, Plate-Forme de Chirurgie Et D'Imagerie Pour La Recherche Et L'Enseignement, Université de Tours, 37380, Nouzilly, France
| | - Lucie Combes-Soia
- INRAE, CNRS, IFCE, UMR PRC, Université de Tours, 37380, Nouzilly, France
- INRAE, CHU de Tours, Plate-Forme de Chirurgie Et D'Imagerie Pour La Recherche Et L'Enseignement, Université de Tours, 37380, Nouzilly, France
| | - Paulo Marcelo
- Plate-Forme ICAP, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, 80054, Amiens, France
| | | | - Christophe Baron
- Transplantation, Immunologie et Inflammation T2i - EA 4245, Université de Tours, Tours, France
| | - Denis Angoulvant
- Service de Cardiologie, Centre Hospitalier Universitaire Trousseau Et EA7505, Faculté de Médecine, Université François Rabelais, Tours, France
- Transplantation, Immunologie et Inflammation T2i - EA 4245, Université de Tours, Tours, France
| | - Dominique Babuty
- Service de Cardiologie, Centre Hospitalier Universitaire Trousseau Et EA7505, Faculté de Médecine, Université François Rabelais, Tours, France
| | - Nicolas Clementy
- Service de Cardiologie, Centre Hospitalier Universitaire Trousseau Et EA7505, Faculté de Médecine, Université François Rabelais, Tours, France
- Transplantation, Immunologie et Inflammation T2i - EA 4245, Université de Tours, Tours, France
| |
Collapse
|
21
|
Effects of High-Fat Diet Induced Obesity and Fructooligosaccharide Supplementation on Cardiac Protein Expression. Nutrients 2020; 12:nu12113404. [PMID: 33167590 PMCID: PMC7694524 DOI: 10.3390/nu12113404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/29/2020] [Accepted: 11/03/2020] [Indexed: 01/12/2023] Open
Abstract
The mechanism by which high fat-diet induced obesity affects cardiac protein expression is unclear, and the extent to which this is modulated by prebiotic treatment is not known. These outcomes were assessed in rats initially fed a high-fat diet, then the top 40% weight gain group were randomly allocated to control (CON), high-fat (HF) and HF supplemented with fructooligosaccharide (32 g; HF-FOS) treatments for 12 weeks (n = 10/group). At sacrifice, left ventricles were either frozen or preserved in formalin. Serum was stored for glucose and insulin measurements. Protein spectra was obtained using an Orbitrap analyzer, processed with Sequest and fold changes assessed with Scaffold Q +. Treatment effects for body weights, glucose and insulin were assessed using one-way ANOVA, and the differential protein expression was assessed by a Mann-Whitney U test. The Database for Annotation, Visualization and Integrated Discovery and the Kyoto Encyclopedia of Genes and Genomes identified pathways containing overrepresented proteins. Hematoxylin and eosin sections were graded for hypertrophy and also quantified; differences were identified using Chi-square analyses and Mann-Whitney U tests. HF diet fed rats were significantly (p < 0.05) heavier than CON, and 23 proteins involved in mitochondrial function and lipid metabolism were differentially expressed between HF and CON. Between HF-FOS and HF, 117 proteins involved in contractility, lipid and carbohydrate metabolism were differentially expressed. HF cardiomyocytes were significantly (p < 0.05) more hypertrophic than CON. We conclude that high-fat feeding and FOS are associated with subcellular deviations in cardiac metabolism and contractility, which may influence myocardial function and alter the risk of cardiovascular disease.
Collapse
|
22
|
Zhang XH, Morad M. Ca 2+ signaling of human pluripotent stem cells-derived cardiomyocytes as compared to adult mammalian cardiomyocytes. Cell Calcium 2020; 90:102244. [PMID: 32585508 PMCID: PMC7483365 DOI: 10.1016/j.ceca.2020.102244] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 12/23/2022]
Abstract
Human induced pluripotent stem cells derived cardiomyocytes (hiPSC-CMs) have been extensively used for in vitro modeling of human cardiovascular disease, drug screening and pharmacotherapy, but little rigorous studies have been reported on their biophysical or Ca2+ signaling properties. There is also considerable concern as to the level of their maturity and whether they can serve as reliable models for adult human cardiac myocytes. Ultrastructural difference such as lack of t-tubular network, their polygonal shapes, disorganized sarcomeric myofilament, and their rhythmic automaticity, among others, have been cited as evidence for immaturity of hiPSC-CMs. In this review, we will deal with Ca2+ signaling, its regulation, and its stage of maturity as compared to the mammalian adult cardiomyocytes. We shall summarize the data on functional aspects of Ca2+signaling and its parameters that include: L-type calcium channel (Cav1.2), ICa-induced Ca2+release, CICR, and its parameters, cardiac Na/Ca exchanger (NCX1), the ryanodine receptors (RyR2), sarco-reticular Ca2+pump, SERCA2a/PLB, and the contribution of mitochondrial Ca2+ to hiPSC-CMs excitation-contraction (EC)-coupling as compared with adult mammalian cardiomyocytes. The comparative studies suggest that qualitatively hiPSC-CMs have similar Ca2+signaling properties as those of adult cardiomyocytes, but quantitative differences do exist. This review, we hope, will allow the readers to judge for themselves to what extent Ca2+signaling of hiPSC-CMs represents the adult form of this signaling pathway, and whether these cells can be used as good models of human cardiomyocytes.
Collapse
Affiliation(s)
- Xiao-Hua Zhang
- Cardiac Signaling Center of University of South Carolina, Medical University of South Carolina, Clemson University, Charleston SC, United States
| | - Martin Morad
- Cardiac Signaling Center of University of South Carolina, Medical University of South Carolina, Clemson University, Charleston SC, United States.
| |
Collapse
|
23
|
A Novel Fluorescent Reporter System Identifies Laminin-511/521 as Potent Regulators of Cardiomyocyte Maturation. Sci Rep 2020; 10:4249. [PMID: 32144297 PMCID: PMC7060274 DOI: 10.1038/s41598-020-61163-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/21/2020] [Indexed: 12/31/2022] Open
Abstract
Pluripotent stem cell-derived cardiomyocytes (PSC-CMs) hold great promise for disease modeling and drug discovery. However, PSC-CMs exhibit immature phenotypes in culture, and the lack of maturity limits their broad applications. While physical and functional analyses are generally used to determine the status of cardiomyocyte maturation, they could be time-consuming and often present challenges in comparing maturation-enhancing strategies. Therefore, there is a demand for a method to assess cardiomyocyte maturation rapidly and reproducibly. In this study, we found that Myomesin-2 (Myom2), encoding M-protein, is upregulated postnatally, and based on this, we targeted TagRFP to the Myom2 locus in mouse embryonic stem cells. Myom2-RFP+ PSC-CMs exhibited more mature phenotypes than RFP- cells in morphology, function and transcriptionally, conductive to sarcomere shortening assays. Using this system, we screened extracellular matrices (ECMs) and identified laminin-511/521 as potent enhancers of cardiomyocyte maturation. Together, we developed and characterized a novel fluorescent reporter system for the assessment of cardiomyocyte maturation and identified potent maturation-enhancing ECMs through this simple and rapid assay. This system is expected to facilitate use of PSC-CMs in a variety of scientific and medical investigations.
Collapse
|
24
|
Gaburjakova J, Almassy J, Gaburjakova M. Luminal addition of non-permeant Eu 3+ interferes with luminal Ca 2+ regulation of the cardiac ryanodine receptor. Bioelectrochemistry 2020; 132:107449. [PMID: 31918058 DOI: 10.1016/j.bioelechem.2019.107449] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/17/2019] [Accepted: 12/17/2019] [Indexed: 10/25/2022]
Abstract
Dysregulation of the cardiac ryanodine receptor (RYR2) by luminal Ca2+ has been implicated in a life-threatening, stress-induced arrhythmogenic disease. The mechanism of luminal Ca2+-mediated RYR2 regulation is under debate, and it has been attributed to Ca2+ binding on the cytosolic face (the Ca2+ feedthrough mechanism) and/or the luminal face of the RYR2 channel (the true luminal mechanism). The molecular nature and location of the luminal Ca2+ site is unclear. At the single-channel level, we directly probed the RYR2 luminal face by Eu3+, considering the non-permeant nature of trivalent cations and their high binding affinities for Ca2+ sites. Without affecting essential determinants of the Ca2+ feedthrough mechanism, we found that luminal Eu3+ competitively antagonized the activation effect of luminal Ca2+ on RYR2 responsiveness to cytosolic caffeine, and no appreciable effect was observed for luminal Ba2+ (mimicking the absence of luminal Ca2+). Importantly, luminal Eu3+ caused no changes in RYR2 gating. Our results indicate that two distinct Ca2+ sites (available for luminal Ca2+ even when the channel is closed) are likely involved in the true luminal mechanism. One site facing the lumen regulates channel responsiveness to caffeine, while the other site, presumably positioned in the channel pore, governs the gating behavior.
Collapse
Affiliation(s)
- Jana Gaburjakova
- Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dubravska Cesta 9, 840 05 Bratislava, Slovak Republic.
| | - Janos Almassy
- Department of Physiology, Faculty of Medicine, University of Debrecen, PO Box 400, Debrecen 4002, Hungary.
| | - Marta Gaburjakova
- Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dubravska Cesta 9, 840 05 Bratislava, Slovak Republic.
| |
Collapse
|
25
|
Sarco-Endoplasmic Reticulum Calcium Release Model Based on Changes in the Luminal Calcium Content. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:337-370. [DOI: 10.1007/978-3-030-12457-1_14] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
26
|
Chakravarty H, Bal C, Yadav M, Jena N, Bal NC, Sharon A. First Insight on Small Molecules as Cardiac Calsequestrin Stabilizers. ACS OMEGA 2019; 4:11508-11514. [PMID: 31460256 PMCID: PMC6682146 DOI: 10.1021/acsomega.9b01113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 06/19/2019] [Indexed: 06/10/2023]
Abstract
Catecholaminergic polymorphic ventricular tachycardia (CPVT) is caused by mutations of cardiac calsequestrin (CASQ2) that impair its characteristic ability of Ca2+-induced polymerization-depolymerization. However, stabilizing the CASQ2 polymer by pharmacological agents to treat CPVT has not been reported so far. Here, we tested whether small molecules can stabilize CASQ2 polymers. We synthesized 24 glycinate/alaninate/acetate α-pyranone analogs and conducted the CASQ2 depolymerization assay. Most of the molecules of this class of compounds inhibited the depolymerization of the protein upon Ca2+ chelation by ethylene glycol tetraacetic acid. Structure-activity relationship studies revealed that the compounds with the 4-fluoro-phenyl group at the C-6 position of the pyranone ring and open-chain primary amine at C-4 are the most active of the class. This is the first report of an α-pyranone class of compounds with the ability to stabilize CASQ2 polymers and opens up the possibility to target Ca2+-release disorders via modulation of CASQ2 polymerization.
Collapse
Affiliation(s)
| | - Chandralata Bal
- Department
of Chemistry, Birla Institute of Technology,
Mesra, Ranchi 835215, India
| | - Monika Yadav
- Department
of Chemistry, Birla Institute of Technology,
Mesra, Ranchi 835215, India
| | - Nivedita Jena
- KIIT Technology Business Incubator and KIIT School of Biotechnology, KIIT University, Bhubaneswar 751021 India
| | - Naresh C. Bal
- KIIT Technology Business Incubator and KIIT School of Biotechnology, KIIT University, Bhubaneswar 751021 India
| | - Ashoke Sharon
- Department
of Chemistry, Birla Institute of Technology,
Mesra, Ranchi 835215, India
| |
Collapse
|
27
|
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: 54] [Impact Index Per Article: 9.0] [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
|
28
|
Jones PP, MacQuaide N, Louch WE. Dyadic Plasticity in Cardiomyocytes. Front Physiol 2018; 9:1773. [PMID: 30618792 PMCID: PMC6298195 DOI: 10.3389/fphys.2018.01773] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 11/23/2018] [Indexed: 11/13/2022] Open
Abstract
Contraction of cardiomyocytes is dependent on sub-cellular structures called dyads, where invaginations of the surface membrane (t-tubules) form functional junctions with the sarcoplasmic reticulum (SR). Within each dyad, Ca2+ entry through t-tubular L-type Ca2+ channels (LTCCs) elicits Ca2+ release from closely apposed Ryanodine Receptors (RyRs) in the SR membrane. The efficiency of this process is dependent on the density and macroscale arrangement of dyads, but also on the nanoscale organization of LTCCs and RyRs within them. We presently review accumulating data demonstrating the remarkable plasticity of these structures. Dyads are known to form gradually during development, with progressive assembly of both t-tubules and junctional SR terminals, and precise trafficking of LTCCs and RyRs. While dyads can exhibit compensatory remodeling when required, dyadic degradation is believed to promote impaired contractility and arrythmogenesis in cardiac disease. Recent data indicate that this plasticity of dyadic structure/function is dependent on the regulatory proteins junctophilin-2, amphiphysin-2 (BIN1), and caveolin-3, which critically arrange dyadic membranes while stabilizing the position and activity of LTCCs and RyRs. Indeed, emerging evidence indicates that clustering of both channels enables "coupled gating", implying that nanoscale localization and function are intimately linked, and may allow fine-tuning of LTCC-RyR crosstalk. We anticipate that improved understanding of dyadic plasticity will provide greater insight into the processes of cardiac compensation and decompensation, and new opportunities to target the basic mechanisms underlying heart disease.
Collapse
Affiliation(s)
- Peter P. Jones
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- HeartOtago, University of Otago, Dunedin, New Zealand
| | - Niall MacQuaide
- Institute of Cardiovascular Sciences, University of Glasgow, Glasgow, United Kingdom
- Clyde Biosciences, Glasgow, United Kingdom
| | - William E. Louch
- Institute for Experimental Medical Research, Oslo University Hospital, University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| |
Collapse
|
29
|
Zhong M, Rees CM, Terentyev D, Choi BR, Koren G, Karma A. NCX-Mediated Subcellular Ca 2+ Dynamics Underlying Early Afterdepolarizations in LQT2 Cardiomyocytes. Biophys J 2018; 115:1019-1032. [PMID: 30173888 DOI: 10.1016/j.bpj.2018.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/18/2018] [Accepted: 08/02/2018] [Indexed: 12/12/2022] Open
Abstract
Long QT syndrome type 2 (LQT2) is a congenital disease characterized by loss of function mutations in hERG potassium channels (IKr). LQT2 is associated with fatal ventricular arrhythmias promoted by triggered activity in the form of early afterdepolarizations (EADs). We previously demonstrated that intracellular Ca2+ handling is remodeled in LQT2 myocytes. Remodeling leads to aberrant late RyR-mediated Ca2+ releases that drive forward-mode Na+-Ca2+ exchanger (NCX) current and slow repolarization to promote reopening of L-type calcium channels and EADs. Forward-mode NCX was found to be enhanced despite the fact that these late releases do not significantly alter the whole-cell cytosolic calcium concentration during a vulnerable period of phase 2 of the action potential corresponding to the onset of EADs. Here, we use a multiscale ventricular myocyte model to explain this finding. We show that because the local NCX current is a saturating nonlinear function of the local submembrane calcium concentration, a larger number of smaller-amplitude discrete Ca2+ release events can produce a large increase in whole-cell forward-mode NCX current without increasing significantly the whole-cell cytosolic calcium concentration. Furthermore, we develop novel insights, to our knowledge, into how alterations of stochastic RyR activity at the single-channel level cause late aberrant Ca2+ release events. Experimental measurements in transgenic LTQ2 rabbits confirm the critical arrhythmogenic role of NCX and identify this current as a potential target for antiarrhythmic therapies in LQT2.
Collapse
Affiliation(s)
- Mingwang Zhong
- Physics Department and Center for Interdisciplinary Research in Complex Systems, Northeastern University, Boston, Massachusetts
| | - Colin M Rees
- Physics Department and Center for Interdisciplinary Research in Complex Systems, Northeastern University, Boston, Massachusetts
| | - Dmitry Terentyev
- Cardiovascular Research Centre, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Bum-Rak Choi
- Cardiovascular Research Centre, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Gideon Koren
- Cardiovascular Research Centre, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Alain Karma
- Physics Department and Center for Interdisciplinary Research in Complex Systems, Northeastern University, Boston, Massachusetts.
| |
Collapse
|
30
|
Liu B, Walton SD, Ho HT, Belevych AE, Tikunova SB, Bonilla I, Shettigar V, Knollmann BC, Priori SG, Volpe P, Radwański PB, Davis JP, Györke S. Gene Transfer of Engineered Calmodulin Alleviates Ventricular Arrhythmias in a Calsequestrin-Associated Mouse Model of Catecholaminergic Polymorphic Ventricular Tachycardia. J Am Heart Assoc 2018; 7:JAHA.117.008155. [PMID: 29720499 PMCID: PMC6015318 DOI: 10.1161/jaha.117.008155] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background Catecholaminergic polymorphic ventricular tachycardia (CPVT) is a familial arrhythmogenic syndrome characterized by sudden death. There are several genetic forms of CPVT associated with mutations in genes encoding the cardiac ryanodine receptor (RyR2) and its auxiliary proteins including calsequestrin (CASQ2) and calmodulin (CaM). It has been suggested that impairment of the ability of RyR2 to stay closed (ie, refractory) during diastole may be a common mechanism for these diseases. Here, we explore the possibility of engineering CaM variants that normalize abbreviated RyR2 refractoriness for subsequent viral‐mediated delivery to alleviate arrhythmias in non–CaM‐related CPVT. Methods and Results To that end, we have designed a CaM protein (GSH‐M37Q; dubbed as therapeutic CaM or T‐CaM) that exhibited a slowed N‐terminal Ca dissociation rate and prolonged RyR2 refractoriness in permeabilized myocytes derived from CPVT mice carrying the CASQ2 mutation R33Q. This T‐CaM was introduced to the heart of R33Q mice through recombinant adeno‐associated viral vector serotype 9. Eight weeks postinfection, we performed confocal microscopy to assess Ca handling and recorded surface ECGs to assess susceptibility to arrhythmias in vivo. During catecholamine stimulation with isoproterenol, T‐CaM reduced isoproterenol‐promoted diastolic Ca waves in isolated CPVT cardiomyocytes. Importantly, T‐CaM exposure abolished ventricular tachycardia in CPVT mice challenged with catecholamines. Conclusions Our results suggest that gene transfer of T‐CaM by adeno‐associated viral vector serotype 9 improves myocyte Ca handling and alleviates arrhythmias in a calsequestrin‐associated CPVT model, thus supporting the potential of a CaM‐based antiarrhythmic approach as a therapeutic avenue for genetically distinct forms of CPVT.
Collapse
Affiliation(s)
- Bin Liu
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH.,Department of Biological Sciences, Mississippi State University, Starkville, MI
| | - Shane D Walton
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH
| | - Hsiang-Ting Ho
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH
| | - Andriy E Belevych
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH
| | - Svetlana B Tikunova
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH
| | - Ingrid Bonilla
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH
| | - Vikram Shettigar
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH
| | - Bjorn C Knollmann
- Division of Clinical Pharmacology, Vanderbilt University School of Medicine, Vanderbilt, TN
| | - Silvia G Priori
- Division of Cardiology and Molecular Cardiology, Maugeri Foundation-University of Pavia, Italy
| | - Pompeo Volpe
- Department of Biomedical Sciences, University of Padova, Italy
| | - Przemysław B Radwański
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH
| | - Jonathan P Davis
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH
| | - Sándor Györke
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH
| |
Collapse
|
31
|
Meissner G. The structural basis of ryanodine receptor ion channel function. J Gen Physiol 2017; 149:1065-1089. [PMID: 29122978 PMCID: PMC5715910 DOI: 10.1085/jgp.201711878] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 10/12/2017] [Indexed: 01/25/2023] Open
Abstract
Large-conductance Ca2+ release channels known as ryanodine receptors (RyRs) mediate the release of Ca2+ from an intracellular membrane compartment, the endo/sarcoplasmic reticulum. There are three mammalian RyR isoforms: RyR1 is present in skeletal muscle; RyR2 is in heart muscle; and RyR3 is expressed at low levels in many tissues including brain, smooth muscle, and slow-twitch skeletal muscle. RyRs form large protein complexes comprising four 560-kD RyR subunits, four ∼12-kD FK506-binding proteins, and various accessory proteins including calmodulin, protein kinases, and protein phosphatases. RyRs share ∼70% sequence identity, with the greatest sequence similarity in the C-terminal region that forms the transmembrane, ion-conducting domain comprising ∼500 amino acids. The remaining ∼4,500 amino acids form the large regulatory cytoplasmic "foot" structure. Experimental evidence for Ca2+, ATP, phosphorylation, and redox-sensitive sites in the cytoplasmic structure have been described. Exogenous effectors include the two Ca2+ releasing agents caffeine and ryanodine. Recent work describing the near atomic structures of mammalian skeletal and cardiac muscle RyRs provides a structural basis for the regulation of the RyRs by their multiple effectors.
Collapse
Affiliation(s)
- Gerhard Meissner
- Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
32
|
Paul M, Kemparaju K, Girish KS. Inhibition of constitutive NF-κB activity induces platelet apoptosis via ER stress. Biochem Biophys Res Commun 2017; 493:1471-1477. [PMID: 28986259 DOI: 10.1016/j.bbrc.2017.10.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 10/03/2017] [Indexed: 12/15/2022]
Abstract
Platelets are anucleate cells, known for their pivotal roles in hemostasis, inflammation, immunity, and disease progression. Being anuclear, platelets are known to express several transcriptional factors which exert nongenomic functions, including the positive and negative regulation of platelet activation. NF-κB is one such transcriptional factor involved in the regulation of genes for survival, proliferation, inflammation and immunity. Although, the role NF-κB in platelet activation and aggregation is partially known, its function in management of platelet survival and apoptosis remain unexplored. Therefore, two unrelated inhibitors of NF-κB activation, BAY 11-7082 and MLN4924 were used to determine the role of NF-κB in platelets. Inhibition of NF-κB caused decreased SERCA activity and increased cytosolic Ca2+ level causing ER stress which was determined by the phosphorylation of eIF2-α. Further, there was increased BAX and decreased BCl-2 levels, incidence of mitochondrial membrane potential depolarization, release of cytochrome c into cytosol, caspase activation, PS externalization and cell death in BAY 11-7082 and MLN4924 treated platelets. The obtained results demonstrate the critical role played by NF-κB in Ca2+ homeostasis and survival of platelets. In addition, the study demonstrates the potential side effects associated with NF-κB inhibitors employed during inflammation and cancer therapy.
Collapse
Affiliation(s)
- Manoj Paul
- DOS in Biochemistry, University of Mysore, Manasagangothri, Mysuru 570 006, India
| | - Kempaiah Kemparaju
- DOS in Biochemistry, University of Mysore, Manasagangothri, Mysuru 570 006, India.
| | - Kesturu S Girish
- DOS in Biochemistry, University of Mysore, Manasagangothri, Mysuru 570 006, India; Department of Studies and Research in Biochemistry, Tumkur University, Tumakuru 572 103, India.
| |
Collapse
|
33
|
Blatter LA. The intricacies of atrial calcium cycling during excitation-contraction coupling. J Gen Physiol 2017; 149:857-865. [PMID: 28798277 PMCID: PMC5583713 DOI: 10.1085/jgp.201711809] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 07/12/2017] [Indexed: 12/20/2022] Open
Abstract
Blatter discusses the initiation and spread of Ca release, Ca store depletion, and release termination in atrial myocytes.
Collapse
Affiliation(s)
- Lothar A Blatter
- Department of Physiology and Biophysics, Rush University Medical Center, Chicago, IL
| |
Collapse
|
34
|
Jones PP, Guo W, Chen SRW. Control of cardiac ryanodine receptor by sarcoplasmic reticulum luminal Ca 2. J Gen Physiol 2017; 149:867-875. [PMID: 28798281 PMCID: PMC5583710 DOI: 10.1085/jgp.201711805] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/25/2017] [Accepted: 07/18/2017] [Indexed: 12/22/2022] Open
Abstract
Jones et al. propose that SR luminal Ca2+ regulates RyR2 activity via a luminal Ca2+ sensor distinct from the cytosolic Ca2+ sensor.
Collapse
Affiliation(s)
- Peter P Jones
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, Otago, New Zealand .,HeartOtago, University of Otago, Dunedin, Otago, New Zealand
| | - Wenting Guo
- Libin Cardiovascular Institute of Alberta, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - S R Wayne Chen
- Libin Cardiovascular Institute of Alberta, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
35
|
Sobie EA, Williams GSB, Lederer WJ. Ambiguous interactions between diastolic and SR Ca 2+ in the regulation of cardiac Ca 2+ release. J Gen Physiol 2017; 149:847-855. [PMID: 28798276 PMCID: PMC5583714 DOI: 10.1085/jgp.201711814] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/21/2017] [Indexed: 01/20/2023] Open
Abstract
Sobie et al. highlight unresolved issues concerning the regulation of sarcoplasmic reticulum calcium release in cardiac myocytes.
Collapse
Affiliation(s)
- Eric A Sobie
- Department of Pharmacological Sciences, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - George S B Williams
- BioMET, Center for Biomedical Engineering and Technology, University of Maryland, Baltimore, MD
| | - W J Lederer
- BioMET, Center for Biomedical Engineering and Technology, University of Maryland, Baltimore, MD
| |
Collapse
|
36
|
Ríos E. Perspectives on "Control of Ca release from within the cardiac sarcoplasmic reticulum". J Gen Physiol 2017; 149:833-836. [PMID: 28798278 PMCID: PMC5583715 DOI: 10.1085/jgp.201711847] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 07/24/2017] [Indexed: 12/11/2022] Open
Abstract
Five groups of experts unravel the complex modulation of a function crucial for the beating heart.
Collapse
Affiliation(s)
- Eduardo Ríos
- Section of Cellular Signaling, Department of Physiology and Biophysics, Rush University, Chicago, IL
| |
Collapse
|
37
|
Cannell MB, Kong CHT. Quenching the spark: Termination of CICR in the submicroscopic space of the dyad. J Gen Physiol 2017; 149:837-845. [PMID: 28798280 PMCID: PMC5583711 DOI: 10.1085/jgp.201711807] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 07/21/2017] [Indexed: 12/20/2022] Open
Abstract
Cannell and Kong discuss the different termination mechanisms proposed for CICR in cardiac myocytes.
Collapse
Affiliation(s)
- Mark B Cannell
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, England, UK
| | - Cherrie H T Kong
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, England, UK
| |
Collapse
|
38
|
Györke S, Belevych AE, Liu B, Kubasov IV, Carnes CA, Radwański PB. The role of luminal Ca regulation in Ca signaling refractoriness and cardiac arrhythmogenesis. J Gen Physiol 2017; 149:877-888. [PMID: 28798279 PMCID: PMC5583712 DOI: 10.1085/jgp.201711808] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Revised: 06/19/2017] [Accepted: 07/12/2017] [Indexed: 01/05/2023] Open
Abstract
Györke et al. discuss the role of sarcoplasmic reticulum Ca2+ in cardiac refractoriness and pathological implications.
Collapse
Affiliation(s)
- Sándor Györke
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH .,Davis Heart and Lung Research Institute, Columbus, OH
| | - Andriy E Belevych
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH.,Davis Heart and Lung Research Institute, Columbus, OH
| | - Bin Liu
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH.,Davis Heart and Lung Research Institute, Columbus, OH
| | - Igor V Kubasov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Cynthia A Carnes
- College of Pharmacy, The Ohio State University, Columbus, OH.,Davis Heart and Lung Research Institute, Columbus, OH
| | - Przemysław B Radwański
- College of Pharmacy, The Ohio State University, Columbus, OH.,Davis Heart and Lung Research Institute, Columbus, OH
| |
Collapse
|
39
|
Maxwell JT, Blatter LA. A novel mechanism of tandem activation of ryanodine receptors by cytosolic and SR luminal Ca 2+ during excitation-contraction coupling in atrial myocytes. J Physiol 2017; 595:3835-3845. [PMID: 28028837 DOI: 10.1113/jp273611] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 12/01/2016] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS In atrial myocytes excitation-contraction coupling is strikingly different from ventricle because atrial myocytes lack a transverse tubule membrane system: Ca2+ release starts in the cell periphery and propagates towards the cell centre by Ca2+ -induced Ca2+ release from the sarcoplasmic reticulum (SR) Ca2+ store. The cytosolic Ca2+ sensitivity of the ryanodine receptor (RyRs) Ca2+ release channel is low and it is unclear how Ca2+ release can be activated in the interior of atrial cells. Simultaneous confocal imaging of cytosolic and intra-SR calcium revealed a transient elevation of store Ca2+ that we termed 'Ca2+ sensitization signal'. We propose a novel paradigm of atrial ECC that is based on tandem activation of the RyRs by cytosolic and luminal Ca2+ through a 'fire-diffuse-uptake-fire' (or FDUF) mechanism: Ca2+ uptake by SR Ca2+ pumps at the propagation front elevates Ca2+ inside the SR locally, leading to luminal RyR sensitization and lowering of the cytosolic Ca2+ activation threshold. ABSTRACT In atrial myocytes Ca2+ release during excitation-contraction coupling (ECC) is strikingly different from ventricular myocytes. In many species atrial myocytes lack a transverse tubule system, dividing the sarcoplasmic reticulum (SR) Ca2+ store into the peripheral subsarcolemmnal junctional (j-SR) and the much more abundant central non-junctional (nj-SR) SR. Action potential (AP)-induced Ca2+ entry activates Ca2+ -induced Ca2+ release (CICR) from j-SR ryanodine receptor (RyR) Ca2+ release channels. Peripheral elevation of [Ca2+ ]i initiates CICR from nj-SR and sustains propagation of CICR to the cell centre. Simultaneous confocal measurements of cytosolic ([Ca2+ ]i ; with the fluorescent Ca2+ indicator rhod-2) and intra-SR ([Ca2+ ]SR ; fluo-5N) Ca2+ in rabbit atrial myocytes revealed that Ca2+ release from j-SR resulted in a cytosolic Ca2+ transient of higher amplitude compared to release from nj-SR; however, the degree of depletion of j-SR [Ca2+ ]SR was smaller than nj-SR [Ca2+ ]SR . Similarly, Ca2+ signals from individual release sites of the j-SR showed a larger cytosolic amplitude (Ca2+ sparks) but smaller depletion (Ca2+ blinks) than release from nj-SR. During AP-induced Ca2+ release the rise of [Ca2+ ]i detected at individual release sites of the nj-SR preceded the depletion of [Ca2+ ]SR , and during this latency period a transient elevation of [Ca2+ ]SR occurred. We propose that Ca2+ release from nj-SR is activated by cytosolic and luminal Ca2+ (tandem RyR activation) via a novel 'fire-diffuse-uptake-fire' (FDUF) mechanism. This novel paradigm of atrial ECC predicts that Ca2+ uptake by sarco-endoplasmic reticulum Ca2+ -ATPase (SERCA) at the propagation front elevates local [Ca2+ ]SR , leading to luminal RyR sensitization and lowering of the activation threshold for cytosolic CICR.
Collapse
Affiliation(s)
- Joshua T Maxwell
- Department of Molecular Biophysics and Physiology, Rush University Medical Centre, Chicago, IL, 60612, USA
| | - Lothar A Blatter
- Department of Molecular Biophysics and Physiology, Rush University Medical Centre, Chicago, IL, 60612, USA
| |
Collapse
|
40
|
Calsequestrin depolymerizes when calcium is depleted in the sarcoplasmic reticulum of working muscle. Proc Natl Acad Sci U S A 2017; 114:E638-E647. [PMID: 28069951 DOI: 10.1073/pnas.1620265114] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Calsequestrin, the only known protein with cyclical storage and supply of calcium as main role, is proposed to have other functions, which remain unproven. Voluntary movement and the heart beat require this calcium flow to be massive and fast. How does calsequestrin do it? To bind large amounts of calcium in vitro, calsequestrin must polymerize and then depolymerize to release it. Does this rule apply inside the sarcoplasmic reticulum (SR) of a working cell? We answered using fluorescently tagged calsequestrin expressed in muscles of mice. By FRAP and imaging we monitored mobility of calsequestrin as [Ca2+] in the SR--measured with a calsequestrin-fused biosensor--was lowered. We found that calsequestrin is polymerized within the SR at rest and that it depolymerized as [Ca2+] went down: fully when calcium depletion was maximal (a condition achieved with an SR calcium channel opening drug) and partially when depletion was limited (a condition imposed by fatiguing stimulation, long-lasting depolarization, or low drug concentrations). With fluorescence and electron microscopic imaging we demonstrated massive movements of calsequestrin accompanied by drastic morphological SR changes in fully depleted cells. When cells were partially depleted no remodeling was found. The present results support the proposed role of calsequestrin in termination of calcium release by conformationally inducing closure of SR channels. A channel closing switch operated by calsequestrin depolymerization will limit depletion, thereby preventing full disassembly of the polymeric calsequestrin network and catastrophic structural changes in the SR.
Collapse
|
41
|
Abstract
Cardiac arrhythmias can follow disruption of the normal cellular electrophysiological processes underlying excitable activity and their tissue propagation as coherent wavefronts from the primary sinoatrial node pacemaker, through the atria, conducting structures and ventricular myocardium. These physiological events are driven by interacting, voltage-dependent, processes of activation, inactivation, and recovery in the ion channels present in cardiomyocyte membranes. Generation and conduction of these events are further modulated by intracellular Ca2+ homeostasis, and metabolic and structural change. This review describes experimental studies on murine models for known clinical arrhythmic conditions in which these mechanisms were modified by genetic, physiological, or pharmacological manipulation. These exemplars yielded molecular, physiological, and structural phenotypes often directly translatable to their corresponding clinical conditions, which could be investigated at the molecular, cellular, tissue, organ, and whole animal levels. Arrhythmogenesis could be explored during normal pacing activity, regular stimulation, following imposed extra-stimuli, or during progressively incremented steady pacing frequencies. Arrhythmic substrate was identified with temporal and spatial functional heterogeneities predisposing to reentrant excitation phenomena. These could arise from abnormalities in cardiac pacing function, tissue electrical connectivity, and cellular excitation and recovery. Triggering events during or following recovery from action potential excitation could thereby lead to sustained arrhythmia. These surface membrane processes were modified by alterations in cellular Ca2+ homeostasis and energetics, as well as cellular and tissue structural change. Study of murine systems thus offers major insights into both our understanding of normal cardiac activity and its propagation, and their relationship to mechanisms generating clinical arrhythmias.
Collapse
Affiliation(s)
- Christopher L-H Huang
- Physiological Laboratory and the Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
42
|
Handhle A, Ormonde CE, Thomas NL, Bralesford C, Williams AJ, Lai FA, Zissimopoulos S. Calsequestrin interacts directly with the cardiac ryanodine receptor luminal domain. J Cell Sci 2016; 129:3983-3988. [PMID: 27609834 PMCID: PMC5117208 DOI: 10.1242/jcs.191643] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 09/05/2016] [Indexed: 11/20/2022] Open
Abstract
Cardiac muscle contraction requires sarcoplasmic reticulum (SR) Ca2+ release mediated by the quaternary complex comprising the ryanodine receptor 2 (RyR2), calsequestrin 2 (CSQ2), junctin (encoded by ASPH) and triadin. Here, we demonstrate that a direct interaction exists between RyR2 and CSQ2. Topologically, CSQ2 binding occurs at the first luminal loop of RyR2. Co-expression of RyR2 and CSQ2 in a human cell line devoid of the other quaternary complex proteins results in altered Ca2+-release dynamics compared to cells expressing RyR2 only. These findings provide a new perspective for understanding the SR luminal Ca2+ sensor and its involvement in cardiac physiology and disease.
Collapse
Affiliation(s)
- Ahmed Handhle
- Sir Geraint Evans Wales Heart Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK.,Medical Biochemistry Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Chloe E Ormonde
- Sir Geraint Evans Wales Heart Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - N Lowri Thomas
- Sir Geraint Evans Wales Heart Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Catherine Bralesford
- Sir Geraint Evans Wales Heart Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Alan J Williams
- Sir Geraint Evans Wales Heart Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - F Anthony Lai
- Sir Geraint Evans Wales Heart Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Spyros Zissimopoulos
- Sir Geraint Evans Wales Heart Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| |
Collapse
|
43
|
Oxidative stress and ca(2+) release events in mouse cardiomyocytes. Biophys J 2016; 107:2815-2827. [PMID: 25517148 DOI: 10.1016/j.bpj.2014.10.054] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 09/30/2014] [Accepted: 10/24/2014] [Indexed: 12/31/2022] Open
Abstract
Cellular oxidative stress, associated with a variety of common cardiac diseases, is well recognized to affect the function of several key proteins involved in Ca(2+) signaling and excitation-contraction coupling, which are known to be exquisitely sensitive to reactive oxygen species. These include the Ca(2+) release channels of the sarcoplasmic reticulum (ryanodine receptors or RyR2s) and the Ca(2+)/calmodulin-dependent protein kinase II (CaMKII). Oxidation of RyR2s was found to increase the open probability of the channel, whereas CaMKII can be activated independent of Ca(2+) through oxidation. Here, we investigated how oxidative stress affects RyR2 function and SR Ca(2+) signaling in situ, by analyzing Ca(2+) sparks in permeabilized mouse cardiomyocytes under a broad range of oxidative conditions. The results show that with increasing oxidative stress Ca(2+) spark duration is prolonged. In addition, long and very long-lasting (up to hundreds of milliseconds) localized Ca(2+) release events started to appear, eventually leading to sarcoplasmic reticulum (SR) Ca(2+) depletion. These changes of release duration could be prevented by the CaMKII inhibitor KN93 and did not occur in mice lacking the CaMKII-specific S2814 phosphorylation site on RyR2. The appearance of long-lasting Ca(2+) release events was paralleled by an increase of RyR2 oxidation, but also by RyR-S2814 phosphorylation, and by CaMKII oxidation. Our results suggest that in a strongly oxidative environment oxidation-dependent activation of CaMKII leads to RyR2 phosphorylation and thereby contributes to the massive prolongation of SR Ca(2+) release events.
Collapse
|
44
|
Petrovič P, Valent I, Cocherová E, Pavelková J, Zahradníková A. Ryanodine receptor gating controls generation of diastolic calcium waves in cardiac myocytes. ACTA ACUST UNITED AC 2016; 145:489-511. [PMID: 26009544 PMCID: PMC4442793 DOI: 10.1085/jgp.201411281] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Calcium waves can form and propagate at low frequencies of spontaneous calcium sparks if the calcium dependence of spark frequency is sufficiently steep, or the number of open RyRs is sufficiently large. The role of cardiac ryanodine receptor (RyR) gating in the initiation and propagation of calcium waves was investigated using a mathematical model comprising a stochastic description of RyR gating and a deterministic description of calcium diffusion and sequestration. We used a one-dimensional array of equidistantly spaced RyR clusters, representing the confocal scanning line, to simulate the formation of calcium sparks. Our model provided an excellent description of the calcium dependence of the frequency of diastolic calcium sparks and of the increased tendency for the production of calcium waves after a decrease in cytosolic calcium buffering. We developed a hypothesis relating changes in the propensity to form calcium waves to changes of RyR gating and tested it by simulation. With a realistic RyR gating model, increased ability of RyR to be activated by Ca2+ strongly increased the propensity for generation of calcium waves at low (0.05–0.1-µM) calcium concentrations but only slightly at high (0.2–0.4-µM) calcium concentrations. Changes in RyR gating altered calcium wave formation by changing the calcium sensitivity of spontaneous calcium spark activation and/or the average number of open RyRs in spontaneous calcium sparks. Gating changes that did not affect RyR activation by Ca2+ had only a weak effect on the propensity to form calcium waves, even if they strongly increased calcium spark frequency. Calcium waves induced by modulating the properties of the RyR activation site could be suppressed by inhibiting the spontaneous opening of the RyR. These data can explain the increased tendency for production of calcium waves under conditions when RyR gating is altered in cardiac diseases.
Collapse
Affiliation(s)
- Pavol Petrovič
- Department of Physical and Theoretical Chemistry, Faculty of Natural Sciences, Comenius University, 842 15 Bratislava, Slovak Republic
| | - Ivan Valent
- Department of Physical and Theoretical Chemistry, Faculty of Natural Sciences, Comenius University, 842 15 Bratislava, Slovak Republic Department of Muscle Cell Research, Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences, 833 34 Bratislava, Slovak Republic
| | - Elena Cocherová
- Department of Muscle Cell Research, Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences, 833 34 Bratislava, Slovak Republic Institute of Electronics and Photonics, Faculty of Electrical Engineering and Information Technology, Slovak University of Technology, 812 19 Bratislava, Slovak Republic
| | - Jana Pavelková
- Department of Muscle Cell Research, Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences, 833 34 Bratislava, Slovak Republic
| | - Alexandra Zahradníková
- Department of Muscle Cell Research, Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences, 833 34 Bratislava, Slovak Republic
| |
Collapse
|
45
|
Liu B, Ho HT, Brunello L, Unudurthi SD, Lou Q, Belevych AE, Qian L, Kim DH, Cho C, Janssen PML, Hund TJ, Knollmann BC, Kranias EG, Györke S. Ablation of HRC alleviates cardiac arrhythmia and improves abnormal Ca handling in CASQ2 knockout mice prone to CPVT. Cardiovasc Res 2015; 108:299-311. [PMID: 26410369 DOI: 10.1093/cvr/cvv222] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 09/17/2015] [Indexed: 01/02/2023] Open
Abstract
AIMS Cardiac calsequestrin (CASQ2) and histidine-rich Ca-binding protein (HRC) are sarcoplasmic reticulum (SR) Ca-binding proteins that regulate SR Ca release in mammalian heart. Deletion of either CASQ2 or HRC results in relatively mild phenotypes characterized by preserved cardiac structure and function, although CASQ2 knockout (KO), or Cnull, shows increased arrhythmia burden under conditions of catecholaminergic stress. We hypothesized that given the apparent overlap of functions of CASQ2 and HRC, simultaneous ablation of both would deteriorate the cardiac phenotype compared with the single knockouts. METHODS AND RESULTS In contrast to this expectation, double knockout (DKO) mice lacking both CASQ2 and HRC exhibited normal cardiac ejection fraction and ultrastructure. Moreover, the predisposition to catecholamine-dependent arrhythmia that characterizes the Cnull phenotype was alleviated in the DKO mice. At the myocyte level, DKO mice displayed Ca transients of normal amplitude; additionally, the frequency of spontaneous Ca waves and sparks in the presence of isoproterenol were decreased markedly compared with Cnull. Furthermore, restitution of SR Ca release was slowed in DKO myocytes compared with Cnull cells. CONCLUSION Our results suggest that rather than being functionally redundant, CASQ2 and HRC modulate cardiac ryanodine receptor-mediated (RyR2) Ca release in an opposing manner. In particular, while CASQ2 stabilizes RyR2 rendering it refractory in the diastolic phase, HRC enhances RyR2 activity facilitating RyR2 recovery from refractoriness.
Collapse
Affiliation(s)
- Bin Liu
- Department of Physiology and Cell Biology, College of Medicine, and Davis Heart and Lung Research Institute, The Ohio State University, 473 W. 12th Avenue, Columbus, OH 43210, USA
| | - Hsiang-Ting Ho
- Department of Physiology and Cell Biology, College of Medicine, and Davis Heart and Lung Research Institute, The Ohio State University, 473 W. 12th Avenue, Columbus, OH 43210, USA
| | - Lucia Brunello
- Department of Physiology and Cell Biology, College of Medicine, and Davis Heart and Lung Research Institute, The Ohio State University, 473 W. 12th Avenue, Columbus, OH 43210, USA
| | - Sathya D Unudurthi
- Department of Physiology and Cell Biology, College of Medicine, and Davis Heart and Lung Research Institute, The Ohio State University, 473 W. 12th Avenue, Columbus, OH 43210, USA
| | - Qing Lou
- Department of Physiology and Cell Biology, College of Medicine, and Davis Heart and Lung Research Institute, The Ohio State University, 473 W. 12th Avenue, Columbus, OH 43210, USA
| | - Andriy E Belevych
- Department of Physiology and Cell Biology, College of Medicine, and Davis Heart and Lung Research Institute, The Ohio State University, 473 W. 12th Avenue, Columbus, OH 43210, USA
| | - Lan Qian
- Department of Physiology and Cell Biology, College of Medicine, and Davis Heart and Lung Research Institute, The Ohio State University, 473 W. 12th Avenue, Columbus, OH 43210, USA
| | - Do Han Kim
- School of Life Sciences and Systems Biology Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Chunghee Cho
- School of Life Sciences and Systems Biology Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Paul M L Janssen
- Department of Physiology and Cell Biology, College of Medicine, and Davis Heart and Lung Research Institute, The Ohio State University, 473 W. 12th Avenue, Columbus, OH 43210, USA
| | - Thomas J Hund
- Department of Physiology and Cell Biology, College of Medicine, and Davis Heart and Lung Research Institute, The Ohio State University, 473 W. 12th Avenue, Columbus, OH 43210, USA
| | - Bjorn C Knollmann
- Division of Clinical Pharmacology, Vanderbilt University Medical School, Nashville, TN 37232, USA
| | - Evangelia G Kranias
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Sándor Györke
- Department of Physiology and Cell Biology, College of Medicine, and Davis Heart and Lung Research Institute, The Ohio State University, 473 W. 12th Avenue, Columbus, OH 43210, USA
| |
Collapse
|
46
|
Cytosolic Ca²⁺ buffering determines the intra-SR Ca²⁺ concentration at which cardiac Ca²⁺ sparks terminate. Cell Calcium 2015; 58:246-53. [PMID: 26095947 DOI: 10.1016/j.ceca.2015.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/04/2015] [Accepted: 06/06/2015] [Indexed: 11/21/2022]
Abstract
Single ryanodine receptor (RyR) Ca(2+) flux amplitude (i(Ca-RyR)) decreases as intra-sarcoplasmic reticulum (SR) Ca(2+) levels fall during a cardiac Ca(2+) spark. Since i(Ca-RyR) drives the inter-RyR Ca(2+)-induced Ca(2+) release (CICR) that underlies the spark, decreasing i(Ca-RyR) may contribute to spark termination because RyRs that spontaneously close may stay closed. To test this possibility, we simultaneously measured local cytosolic and intra-SR ([Ca(2+)]cyto and [Ca(2+)]SR) during Ca(2+) sparks in permeabilized rabbit ventricular myocytes. Local cytosolic or intra-SR Ca(2+) dynamics were manipulated using Ca(2+) buffers. Buffer manipulations applied in cells had no effect on individual RyR channels reconstituted in planar lipid bilayers. Presence of a fast cytosolic Ca(2+) buffer (BAPTA) significantly suppressed Ca(2+) spark activity and sparks terminated earlier at a higher than usual [Ca(2+)]SR level (∼80% vs. ∼62%). When cytosolic Ca(2+) buffer power was reduced (i.e. cytosolic EGTA level decreased), sparks terminated later and at a lower than usual [Ca(2+)]SR level (∼45% vs. ∼62%). When intra-SR Ca(2+) buffer power was increased, sparks also terminated later and at a lower than usual [Ca(2+)]SR (∼48% vs. ∼62%). These results suggest that cytosolic local control of inter-RyR CICR by i(Ca-RyR) plays a substantial role during the spark termination process. Thus, alterations in local cytosolic Ca(2+) handling dynamics in the dyadic cleft (Ca(2+) buffering, extrusion, etc.) likely influence Ca(2+) spark termination.
Collapse
|
47
|
Tsai WC, Lu YY, Chen YC, Chang CJ, Kao YH, Lin YK, Chen YH, Chen SA, Yang LY, Chen YJ. Ablation of androgen receptor gene triggers right ventricular outflow tract ventricular tachycardia. Int J Cardiol 2015; 189:172-81. [DOI: 10.1016/j.ijcard.2015.04.080] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 04/08/2015] [Accepted: 04/10/2015] [Indexed: 10/23/2022]
|
48
|
Nonaka M, Morimoto S, Murayama T, Kurebayashi N, Li L, Wang YY, Arioka M, Yoshihara T, Takahashi-Yanaga F, Sasaguri T. Stage-dependent benefits and risks of pimobendan in mice with genetic dilated cardiomyopathy and progressive heart failure. Br J Pharmacol 2015; 172:2369-82. [PMID: 25560565 DOI: 10.1111/bph.13062] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 12/16/2014] [Accepted: 12/18/2014] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND AND PURPOSE The Ca(2+) sensitizer pimobendan is a unique inotropic agent that improves cardiac contractility with less of an increase in oxygen consumption and potentially fewer adverse effects on myocardial remodelling and arrhythmia, compared with traditional inotropes. However, clinical trials report contradictory effects of pimobendan in patients with heart failure (HF). We provide mechanistic experimental evidence of the efficacy of pimobendan using a novel mouse model of progressive HF. EXPERIMENTAL APPROACH A knock-in mouse model of human genetic dilated cardiomyopathy, which shows a clear transition from compensatory to end-stage HF at a fixed time during growth, was used to evaluate the efficacy of pimobendan and explore the underlying molecular and cellular mechanisms. KEY RESULTS Pimobendan prevented myocardial remodelling in compensated HF and significantly extended life span in both compensated and end-stage HF, but dose-dependently increased sudden death in end-stage HF. In cardiomyocytes isolated from end-stage HF mice, pimobendan induced triggered activity probably because of early or delayed afterdepolarizations. The L-type Ca(2+) channel blocker verapamil decreased the incidence of triggered activity, suggesting that this was from over-elevated cytoplasmic Ca(2+) through increased Ca(2+) entry by PDE3 inhibition under diminished sarcoplasmic reticulum Ca(2+) reuptake and increased Ca(2+) leakage from sarcoplasmic reticulum in end-stage HF. CONCLUSIONS AND IMPLICATIONS Pimobendan was beneficial regardless of HF stage, but increased sudden cardiac death in end-stage HF with extensive remodelling of Ca(2+) handling. Reduction of cytoplasmic Ca(2+) elevated by PDE3 inhibition might decrease this risk of sudden cardiac death.
Collapse
Affiliation(s)
- Miki Nonaka
- Department of Clinical Pharmacology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Herraiz-Martínez A, Álvarez-García J, Llach A, Molina CE, Fernandes J, Ferrero-Gregori A, Rodríguez C, Vallmitjana A, Benítez R, Padró JM, Martínez-González J, Cinca J, Hove-Madsen L. Ageing is associated with deterioration of calcium homeostasis in isolated human right atrial myocytes. Cardiovasc Res 2015; 106:76-86. [PMID: 25712961 PMCID: PMC4362404 DOI: 10.1093/cvr/cvv046] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Aims Ageing-related cardiac disorders such as heart failure and atrial fibrillation often present with intracellular calcium homeostasis dysfunction. However, knowledge of the intrinsic effects of ageing on cellular calcium handling in the human heart is sparse. Therefore, this study aimed to analyse how ageing affects key mechanisms that regulate intracellular calcium in human atrial myocytes. Methods and results Whole membrane currents and intracellular calcium transients were measured in isolated human right atrial myocytes from 80 patients with normal left atrial dimensions and no history of atrial fibrillation. Patients were categorized as young (<55 years, n = 21), middle aged (55–74 years, n = 42), and old (≥75 years, n = 17). Protein levels were determined by western blot. Ageing was associated with the following electrophysiological changes: (i) a 3.2-fold decrease in the calcium transient (P < 0.01); (ii) reduction of the L-type calcium current (ICa) amplitude (2.4 ± 0.3 pA/pF vs. 1.4 ± 0.2 pA/pF, P < 0.01); (iii) lower levels of L-type calcium channel alpha-subunit (P < 0.05); (iv) lower rates of both fast (14.5 ± 0.9 ms vs. 20.9 ± 1.9, P < 0.01) and slow (73 ± 3 vs. 120 ± 12 ms, P < 0.001) ICa inactivation; and (v) a decrease in the sarcoplasmic reticulum calcium content (10.1 ± 0.8 vs. 6.4 ± 0.6 amol/pF, P < 0.005) associated with a significant decrease in both SERCA2 (P < 0.05) and calsequestrin-2 (P < 0.05) protein levels. In contrast, ageing did not affect spontaneous sarcoplasmic reticulum calcium release. Conclusion Ageing is associated with depression of SR calcium content, L-type calcium current, and calcium transient amplitude that may favour a progressive decline in right atrial contractile function with age.
Collapse
Affiliation(s)
- Adela Herraiz-Martínez
- Cardiovascular Research Centre CSIC-ICCC and IIB-Sant Pau, Hospital de la Santa Creu i Sant Pau, St Antoni Mª Claret 167, Barcelona 08025, Spain
| | - Jesus Álvarez-García
- Department of Cardiology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anna Llach
- Department of Cardiology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Cristina E Molina
- Cardiovascular Research Centre CSIC-ICCC and IIB-Sant Pau, Hospital de la Santa Creu i Sant Pau, St Antoni Mª Claret 167, Barcelona 08025, Spain
| | - Jacqueline Fernandes
- Department of Cardiology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Andreu Ferrero-Gregori
- Department of Cardiology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Cristina Rodríguez
- Cardiovascular Research Centre CSIC-ICCC and IIB-Sant Pau, Hospital de la Santa Creu i Sant Pau, St Antoni Mª Claret 167, Barcelona 08025, Spain
| | - Alexander Vallmitjana
- Department of Automatic Control, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Raúl Benítez
- Department of Automatic Control, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Josep M Padró
- Department of Cardiac Surgery, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - José Martínez-González
- Cardiovascular Research Centre CSIC-ICCC and IIB-Sant Pau, Hospital de la Santa Creu i Sant Pau, St Antoni Mª Claret 167, Barcelona 08025, Spain
| | - Juan Cinca
- Department of Cardiology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Leif Hove-Madsen
- Cardiovascular Research Centre CSIC-ICCC and IIB-Sant Pau, Hospital de la Santa Creu i Sant Pau, St Antoni Mª Claret 167, Barcelona 08025, Spain
| |
Collapse
|
50
|
Zhang JZ, Waddell HMM, Jones PP. Regulation of RYR2 by sarcoplasmic reticulum Ca(2+). Clin Exp Pharmacol Physiol 2015; 42:720-6. [PMID: 25603835 DOI: 10.1111/1440-1681.12364] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Revised: 09/17/2014] [Accepted: 10/09/2014] [Indexed: 11/28/2022]
Abstract
Ca(2+) is arguably the most important ion involved in the contraction of the heart. The cardiac ryanodine receptor (RyR2), the major Ca(2+) release channel located in the sarcoplasmic reticulum (SR) membrane, is responsible for releasing the bulk of Ca(2+) required for contraction. Moreover, RyR2 is also crucial for maintaining SR Ca(2+) homeostasis by releasing Ca(2+) from the SR when it becomes overloaded with Ca(2+) . During normal contraction, RyR2 is activated by cytosolic Ca(2+) , whereas during store overload conditions, the opening of RyR2 is governed by SR Ca(2+) . Although the process of the cytosolic control of RyR2 is well established, the molecular mechanism by which SR luminal Ca(2+) regulates RyR2 has only recently been elucidated and remains controversial. In addition to the activation of RyR2, SR luminal Ca(2+) also determines when the RyR2 channel closes. RyR2-mediated Ca(2+) release from the SR does not continue until the SR is completely depleted. Rather, it ceases when SR luminal Ca(2+) falls below a certain level. Given the importance of SR Ca(2+) , it is not surprising that the SR luminal Ca(2+) level is tightly controlled by SR Ca(2+) -buffering proteins. Consequently, the opening and closing of RyR2 is heavily influenced by the presence of such proteins, particularly those associated with RyR2, such as calsequestrin and the histidine-rich Ca(2+) -binding protein. These proteins appear to indirectly alter RyR2 activity by modifying the microdomain SR Ca(2+) level surrounding RyR2.
Collapse
Affiliation(s)
- Joe Z Zhang
- Department of Physiology and HeartOtago, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - Helen M M Waddell
- Department of Physiology and HeartOtago, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - Peter P Jones
- Department of Physiology and HeartOtago, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|