1
|
Vázquez‐Sánchez S, Blasco A, Fernández‐Corredoira P, Cantolla P, Mercado‐García E, Rodríguez‐Sánchez E, González‐Lafuente L, Poveda J, González‐Moreno D, Matutano A, Peribañez S, García‐Consuegra I, Volpe M, Fernández‐Velasco M, Ruilope LM, Ruiz‐Hurtado G. Recombinant Klotho administration after myocardial infarction reduces ischaemic injury and arrhythmias by blocking intracellular calcium mishandling and CaMKII activation. J Pathol 2025; 265:342-356. [PMID: 39815421 PMCID: PMC11794962 DOI: 10.1002/path.6388] [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: 05/08/2024] [Revised: 10/04/2024] [Accepted: 12/03/2024] [Indexed: 01/18/2025]
Abstract
Ischaemic heart disease (IHD) remains a major cause of death and morbidity. Klotho is a well-known anti-ageing factor with relevant cardioprotective actions, at least when renal dysfunction is present, but its actions are much less known when renal function is preserved. This study investigated Klotho as a biomarker and potential novel treatment of IHD-associated complications after myocardial infarction (MI) under preserved renal function. Association between circulating Klotho levels and cardiac injury was investigated in patients after ST-elevation MI (STEMI). Biochemical, in vivo and in vitro cardiac function and histological and molecular studies were performed to determine the effect of recombinant Klotho in the failing hearts of mice after MI. We demonstrated that STEMI patients showed lower systemic Klotho levels, with the lowest Klotho tertile in those patients with higher N-terminal pro B-type natriuretic peptide (NT-proBNP) levels. Mice also showed a decrease in systemic Klotho levels after MI induction. Furthermore, recombinant Klotho administration in mice reduced infarct area and attenuated cardiac hypertrophy and fibrosis. We also demonstrated that Klotho treatment prevented reduction in ejection fraction and MI-related ECG changes, including prolonged QRS, JT, QTc, and TpeakTend intervals and premature ventricular contractions. In adult mouse cardiomyocytes, Klotho treatment restricted systolic calcium (Ca2+) release and cell shortening disturbances after MI. Klotho prevented increased diastolic Ca2+ leak and pro-arrhythmogenic events in PMI mice by blocking activation of the Ca2+/calmodulin-dependent kinase type II (CaMKII) pathway, preventing ryanodine receptor type 2 (RyR2) hyperphosphorylation. In conclusion, Klotho supplementation protected against functional and structural cardiac remodelling and ameliorated ventricular arrhythmic events by preventing intracardiomyocyte Ca2+ mishandling in mice following MI. These data uncover a new cardioprotective role of Klotho, emerging as a biomarker of ventricular injury and potential treatment for patients after MI. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Sara Vázquez‐Sánchez
- Cardiorenal Translational LaboratoryImas12 Research Institute, Hospital Universitario 12 de OctubreMadridSpain
- RICORS2040‐RenalMadridSpain
| | - Ana Blasco
- Acute Cardiac Care UnitsCardiology Service. Hospital Universitario Puerta de Hierro‐MajadahondaMadridSpain
- Research Ethics CommitteeInstituto de Investigación Puerta de Hierro‐Segovia de AranaMadridSpain
| | | | - Paula Cantolla
- Clinical and Invasive Cardiology Group, Instituto de Investigación Sanitaria del Hospital La Paz (IdiPAZ)Hospital Universitario La PazMadridSpain
| | - Elisa Mercado‐García
- Cardiorenal Translational LaboratoryImas12 Research Institute, Hospital Universitario 12 de OctubreMadridSpain
| | - Elena Rodríguez‐Sánchez
- Cardiorenal Translational LaboratoryImas12 Research Institute, Hospital Universitario 12 de OctubreMadridSpain
- RICORS2040‐RenalMadridSpain
| | - Laura González‐Lafuente
- Cardiorenal Translational LaboratoryImas12 Research Institute, Hospital Universitario 12 de OctubreMadridSpain
- RICORS2040‐RenalMadridSpain
| | - Jonay Poveda
- Cardiorenal Translational LaboratoryImas12 Research Institute, Hospital Universitario 12 de OctubreMadridSpain
| | - Daniel González‐Moreno
- Cardiorenal Translational LaboratoryImas12 Research Institute, Hospital Universitario 12 de OctubreMadridSpain
- RICORS2040‐RenalMadridSpain
| | - Andrea Matutano
- Acute Cardiac Care UnitsCardiology Service. Hospital Universitario Puerta de Hierro‐MajadahondaMadridSpain
- Research Ethics CommitteeInstituto de Investigación Puerta de Hierro‐Segovia de AranaMadridSpain
| | - Sonia Peribañez
- Cardiology ServiceHospital Universitario Miguel ServetZaragozaSpain
| | - Inés García‐Consuegra
- Proteomics UnitInstitute of Research Imas12, Hospital Universitario 12 de OctubreMadridSpain
| | - Massimo Volpe
- Department of Clinical and Molecular MedicineSapienza University of RomeRomeItaly
- IRCCS San RaffaeleRomeItaly
| | - María Fernández‐Velasco
- Clinical and Invasive Cardiology Group, Instituto de Investigación Sanitaria del Hospital La Paz (IdiPAZ)Hospital Universitario La PazMadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER‐CV)Instituto de Salud Carlos IIIMadridSpain
| | - Luis M. Ruilope
- Cardiorenal Translational LaboratoryImas12 Research Institute, Hospital Universitario 12 de OctubreMadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER‐CV)Instituto de Salud Carlos IIIMadridSpain
- European University of MadridMadridSpain
| | - Gema Ruiz‐Hurtado
- Cardiorenal Translational LaboratoryImas12 Research Institute, Hospital Universitario 12 de OctubreMadridSpain
- RICORS2040‐RenalMadridSpain
- Department of Physiology, School of MedicineUniversidad Autónoma de MadridMadridSpain
| |
Collapse
|
2
|
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
|
3
|
Yang B, Wang SQ, Yang HQ. β-adrenergic regulation of Ca 2+ signaling in heart cells. BIOPHYSICS REPORTS 2024; 10:274-282. [PMID: 39539286 PMCID: PMC11554573 DOI: 10.52601/bpr.2024.240906] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/15/2024] [Indexed: 11/16/2024] Open
Abstract
β-adrenergic receptors (βARs) play significant roles in regulating Ca2+ signaling in cardiac myocytes, thus holding a key function in modulating heart performance. βARs regulate the influx of extracellular Ca2+ and the release and uptake of Ca2+ from the sarcoplasmic reticulum (SR) by activating key components such as L-type calcium channels (LTCCs), ryanodine receptors (RyRs) and phospholamban (PLN), mediated by the phosphorylation actions by protein kinase A (PKA). In cardiac myocytes, the presence of β2AR provides a protective mechanism against potential overstimulation of β1AR, which may aid in the restoration of cardiac dysfunctions. Understanding the Ca2+ regulatory signaling pathways of βARs in cardiac myocytes and the differences among various βAR subtypes are crucial in cardiology and hold great potential for developing treatments for heart diseases.
Collapse
Affiliation(s)
- Bo Yang
- Cyrus Tang Medical Institute, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| | - Shi-Qiang Wang
- State Key Lab of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Hua-Qian Yang
- Cyrus Tang Medical Institute, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
4
|
Medvedev RY, Turner DGP, DeGuire FC, Leonov V, Lang D, Gorelik J, Alvarado FJ, Bondarenko VE, Glukhov AV. Caveolae-associated cAMP/Ca 2+-mediated mechano-chemical signal transduction in mouse atrial myocytes. J Mol Cell Cardiol 2023; 184:75-87. [PMID: 37805125 PMCID: PMC10842990 DOI: 10.1016/j.yjmcc.2023.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/11/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Caveolae are tiny invaginations in the sarcolemma that buffer extra membrane and contribute to mechanical regulation of cellular function. While the role of caveolae in membrane mechanosensation has been studied predominantly in non-cardiomyocyte cells, caveolae contribution to cardiac mechanotransduction remains elusive. Here, we studied the role of caveolae in the regulation of Ca2+ signaling in atrial cardiomyocytes. In Langendorff-perfused mouse hearts, atrial pressure/volume overload stretched atrial myocytes and decreased caveolae density. In isolated cells, caveolae were disrupted through hypotonic challenge that induced a temporal (<10 min) augmentation of Ca2+ transients and caused a rise in Ca2+ spark activity. Similar changes in Ca2+ signaling were observed after chemical (methyl-β-cyclodextrin) and genetic ablation of caveolae in cardiac-specific conditional caveolin-3 knock-out mice. Acute disruption of caveolae, both mechanical and chemical, led to the elevation of cAMP level in the cell interior, and cAMP-mediated augmentation of protein kinase A (PKA)-phosphorylated ryanodine receptors (at Ser2030 and Ser2808). Caveolae-mediated stimulatory effects on Ca2+ signaling were abolished via inhibition of cAMP production by adenyl cyclase antagonists MDL12330 and SQ22536, or reduction of PKA activity by H-89. A compartmentalized mathematical model of mouse atrial myocytes linked the observed changes to a microdomain-specific decrease in phosphodiesterase activity, which disrupted cAMP signaling and augmented PKA activity. Our findings add a new dimension to cardiac mechanobiology and highlight caveolae-associated cAMP/PKA-mediated phosphorylation of Ca2+ handling proteins as a novel component of mechano-chemical feedback in atrial myocytes.
Collapse
Affiliation(s)
- Roman Y Medvedev
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Daniel G P Turner
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Frank C DeGuire
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Vladislav Leonov
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Di Lang
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Julia Gorelik
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Francisco J Alvarado
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Vladimir E Bondarenko
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA, USA
| | - Alexey V Glukhov
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
5
|
Hernández Mesa M, van den Brink J, Louch WE, McCabe KJ, Rangamani P. Nanoscale organization of ryanodine receptor distribution and phosphorylation pattern determines the dynamics of calcium sparks. PLoS Comput Biol 2022; 18:e1010126. [PMID: 35666763 PMCID: PMC9203011 DOI: 10.1371/journal.pcbi.1010126] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 06/16/2022] [Accepted: 04/20/2022] [Indexed: 11/18/2022] Open
Abstract
Super-resolution imaging techniques have provided a better understanding of the relationship between the nanoscale organization and function of ryanodine receptors (RyRs) in cardiomyocytes. Recent data have indicated that this relationship is disrupted in heart failure (HF), as RyRs are dispersed into smaller and more numerous clusters. However, RyRs are also hyperphosphorylated in this condition, and this is reported to occur preferentially within the cluster centre. Thus, the combined impact of RyR relocalization and sensitization on Ca2+ spark generation in failing cardiomyocytes is likely complex and these observations suggest that both the nanoscale organization of RyRs and the pattern of phosphorylated RyRs within clusters could be critical determinants of Ca2+ spark dynamics. To test this hypothesis, we used computational modeling to quantify the relationships between RyR cluster geometry, phosphorylation patterns, and sarcoplasmic reticulum (SR) Ca2+ release. We found that RyR cluster disruption results in a decrease in spark fidelity and longer sparks with a lower amplitude. Phosphorylation of some RyRs within the cluster can play a compensatory role, recovering healthy spark dynamics. Interestingly, our model predicts that such compensation is critically dependent on the phosphorylation pattern, as phosphorylation localized within the cluster center resulted in longer Ca2+ sparks and higher spark fidelity compared to a uniformly distributed phosphorylation pattern. Our results strongly suggest that both the phosphorylation pattern and nanoscale RyR reorganization are critical determinants of Ca2+ dynamics in HF.
Collapse
Affiliation(s)
- María Hernández Mesa
- Department of Computational Physiology, Simula Research Laboratory, Oslo, Norway
| | - Jonas van den Brink
- Department of Computational Physiology, Simula Research Laboratory, Oslo, Norway
| | - William E. Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Kimberly J. McCabe
- Department of Computational Physiology, Simula Research Laboratory, Oslo, Norway
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
6
|
Therapeutic Approaches of Ryanodine Receptor-Associated Heart Diseases. Int J Mol Sci 2022; 23:ijms23084435. [PMID: 35457253 PMCID: PMC9031589 DOI: 10.3390/ijms23084435] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 01/08/2023] Open
Abstract
Cardiac diseases are the leading causes of death, with a growing number of cases worldwide, posing a challenge for both healthcare and research. Therefore, the most relevant aim of cardiac research is to unravel the molecular pathomechanisms and identify new therapeutic targets. Cardiac ryanodine receptor (RyR2), the Ca2+ release channel of the sarcoplasmic reticulum, is believed to be a good therapeutic target in a group of certain heart diseases, collectively called cardiac ryanopathies. Ryanopathies are associated with the impaired function of the RyR, leading to heart diseases such as congestive heart failure (CHF), catecholaminergic polymorphic ventricular tachycardia (CPVT), arrhythmogenic right ventricular dysplasia type 2 (ARVD2), and calcium release deficiency syndrome (CRDS). The aim of the current review is to provide a short insight into the pathological mechanisms of ryanopathies and discuss the pharmacological approaches targeting RyR2.
Collapse
|
7
|
Regulation of cardiac ryanodine receptor function by the cyclic-GMP dependent protein kinase G. Curr Res Physiol 2022; 5:171-178. [PMID: 35356048 PMCID: PMC8958330 DOI: 10.1016/j.crphys.2022.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/20/2022] [Accepted: 03/21/2022] [Indexed: 11/21/2022] Open
Abstract
Background The cGMP-dependent protein kinase G (PKG) phosphorylates the cardiac ryanodine receptor (RyR2) in vitro. We aimed to determine whether modulation of endogenous PKG alters RyR2-mediated spontaneous Ca2+ release and whether this effect is linked to a change in RyR2 phosphorylation. Methods & Results: Human embryonic kidney (HEK293) cells with inducible RyR2 expression were treated with the cGMP analogue 8-Br-cGMP (100 μM) to activate endogenous PKG. In cells transfected with luminal Ca2+ sensor, D1ER, PKG activation significantly reduced the threshold for RyR2-mediated spontaneous Ca2+ release (93.9 ± 0.4% of store size with vehicle vs. 91.7 ± 0.8% with 8-Br-cGMP, P = 0.04). Mutation of the proposed PKG phosphorylation sites, S2808 and S2030, either individually or as a combination, prevented the decrease in Ca2+ release threshold induced by endogenous PKG activation. Interestingly, despite a functional dependence on expression of RyR2 phosphorylation sites, 8-Br-cGMP activation of PKG did not promote a detectable change in S2808 phosphorylation (P = 0.9). Paradoxically, pharmacological inhibition of PKG with KT 5823 (1 μM) also reduced the threshold for spontaneous Ca2+ release through RyR2 without affecting S2808 phosphorylation. Silencing RNA knockdown of endogenous PKG expression also had no quantifiable effect on RyR2 S2808 phosphorylation (P = 0.9). However, unlike PKG inhibition with KT 5823, PKG knockdown did not alter spontaneous Ca2+ release propensity or luminal Ca2+ handling. Conclusion In an intact cell model, activation of endogenous PKG reduces the threshold for RyR2-mediated spontaneous Ca2+ release in a manner dependent on the RyR2 phosphorylation sites S2808 and S2030. This study clarifies the regulation of RyR2 Ca2+ release by endogenous PKG and functionally implicates the role of RyR2 phosphorylation. PKG regulation of RyR2 has been under-researched relative to other kinases. Endogenous PKG activation reduced the threshold for spontaneous RyR2 Ca2+ release. Regulation of RyR2 by PKG required expression of serine residues 2808 and 2030. Knockdown of PKG found minimal basal regulation of RyR2 by PKG.
Collapse
|
8
|
MicroRNAs and Calcium Signaling in Heart Disease. Int J Mol Sci 2021; 22:ijms221910582. [PMID: 34638924 PMCID: PMC8508866 DOI: 10.3390/ijms221910582] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 01/02/2023] Open
Abstract
In hearts, calcium (Ca2+) signaling is a crucial regulatory mechanism of muscle contraction and electrical signals that determine heart rhythm and control cell growth. Ca2+ signals must be tightly controlled for a healthy heart, and the impairment of Ca2+ handling proteins is a key hallmark of heart disease. The discovery of microRNA (miRNAs) as a new class of gene regulators has greatly expanded our understanding of the controlling module of cardiac Ca2+ cycling. Furthermore, many studies have explored the involvement of miRNAs in heart diseases. In this review, we aim to summarize cardiac Ca2+ signaling and Ca2+-related miRNAs in pathological conditions, including cardiac hypertrophy, heart failure, myocardial infarction, and atrial fibrillation. We also discuss the therapeutic potential of Ca2+-related miRNAs as a new target for the treatment of heart diseases.
Collapse
|
9
|
Val‐Blasco A, Gil‐Fernández M, Rueda A, Pereira L, Delgado C, Smani T, Ruiz Hurtado G, Fernández‐Velasco M. Ca 2+ mishandling in heart failure: Potential targets. Acta Physiol (Oxf) 2021; 232:e13691. [PMID: 34022101 DOI: 10.1111/apha.13691] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/14/2022]
Abstract
Ca2+ mishandling is a common feature in several cardiovascular diseases such as heart failure (HF). In many cases, impairment of key players in intracellular Ca2+ homeostasis has been identified as the underlying mechanism of cardiac dysfunction and cardiac arrhythmias associated with HF. In this review, we summarize primary novel findings related to Ca2+ mishandling in HF progression. HF research has increasingly focused on the identification of new targets and the contribution of their role in Ca2+ handling to the progression of the disease. Recent research studies have identified potential targets in three major emerging areas implicated in regulation of Ca2+ handling: the innate immune system, bone metabolism factors and post-translational modification of key proteins involved in regulation of Ca2+ handling. Here, we describe their possible contributions to the progression of HF.
Collapse
Affiliation(s)
| | | | - Angélica Rueda
- Department of Biochemistry Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV‐IPN) México City Mexico
| | - Laetitia Pereira
- INSERM UMR‐S 1180 Laboratory of Ca Signaling and Cardiovascular Physiopathology University Paris‐Saclay Châtenay‐Malabry France
| | - Carmen Delgado
- Instituto de Investigaciones Biomédicas Alberto Sols Madrid Spain
- Department of Metabolism and Cell Signalling Biomedical Research Institute "Alberto Sols" CSIC‐UAM Madrid Spain
| | - Tarik Smani
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV) Madrid Spain
- Department of Medical Physiology and Biophysics University of Seville Seville Spain
- Group of Cardiovascular Pathophysiology Institute of Biomedicine of Seville University Hospital of Virgen del Rocío, University of Seville, CSIC Seville Spain
| | - Gema Ruiz Hurtado
- Cardiorenal Translational Laboratory Institute of Research i+12 University Hospital 12 de Octubre Madrid Spain
- CIBER‐CV University Hospita1 12 de Octubre Madrid Spain
| | - Maria Fernández‐Velasco
- La Paz University Hospital Health Research Institute IdiPAZ Madrid Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV) Madrid Spain
| |
Collapse
|
10
|
Sato D, Uchinoumi H, Bers DM. Increasing SERCA function promotes initiation of calcium sparks and breakup of calcium waves. J Physiol 2021; 599:3267-3278. [PMID: 33963531 PMCID: PMC8249358 DOI: 10.1113/jp281579] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/04/2021] [Indexed: 01/16/2023] Open
Abstract
KEY POINTS Increasing sarcoplasmic/endoplasmic reticulum calcium ATPase (SERCA) pump activity enhances sarcoplasmic reticulum calcium (Ca) load, which increases both ryanodine receptor opening and driving force of Ca release flux. Both of these effects promote Ca spark formation and wave propagation. However, increasing SERCA activity also accelerates local cytosolic Ca decay as the wave front travels to the next cluster, which limits wave propagation. As a result, increasing SERCA pump activity has a biphasic effect on the propensity of arrhythmogenic Ca waves, but a monotonic effect to increase Ca spark frequency and amplitude. ABSTRACT Waves of sarcoplasmic reticulum (SR) calcium (Ca) release can cause arrhythmogenic afterdepolarizations in cardiac myocytes. Ca waves propagate when Ca sparks at one Ca release unit (CRU) recruit new Ca sparks in neighbouring CRUs. Under normal conditions, Ca sparks are too small to recruit neighbouring Ca sparks where Ca sensitivity is also low. However, under pathological conditions such as a Ca overload or ryanodine receptor (RyR) sensitization, Ca sparks can be larger and propagate more readily as macro-sparks or full Ca waves. Increasing SERCA pump activity promotes SR Ca load, which promotes RyR opening and increases driving force of the Ca release flux from SR to cytosol, promoting Ca waves. However, high sarcoplasmic/endoplasmic reticulum calcium ATPase (SERCA) activity can also decrease local cytosolic [Ca] as it approaches the next CRU, thereby reducing wave appearance and propagation. In this study, we use a physiologically detailed model of subcellular Ca cycling and experiments in phospholamban-knockout mice, to show how Ca waves are initiated and propagate and how different conditions contribute to the generation and propagation of Ca waves. We show that reducing diffusive coupling between Ca sparks by increasing SERCA activity prevents Ca waves by reducing [Ca] at the next CRU, as do Ca buffers, low intra-SR Ca diffusion and distance between CRUs. Increasing SR Ca uptake rate has a biphasic effect on Ca wave propagation; initially it enhances Ca spark probability and amplitude and CRU coupling, thereby promoting arrhythmogenic Ca wave propagation, but at higher levels SR Ca uptake can abort those arrhythmogenic Ca waves.
Collapse
Affiliation(s)
- Daisuke Sato
- Department of Pharmacology, University of California, Davis School of Medicine, California, USA
| | - Hitoshi Uchinoumi
- Department of Pharmacology, University of California, Davis School of Medicine, California, USA.,Department of Cardiology, Yamaguchi University School of Medicine, Yamaguchi, Japan
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis School of Medicine, California, USA
| |
Collapse
|
11
|
Multisite phosphorylation of the cardiac ryanodine receptor: a random or coordinated event? Pflugers Arch 2020; 472:1793-1807. [PMID: 33078311 DOI: 10.1007/s00424-020-02473-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/03/2020] [Accepted: 10/02/2020] [Indexed: 10/23/2022]
Abstract
Many proteins are phosphorylated at more than one phosphorylation site to achieve precise tuning of protein function and/or integrate a multitude of signals into the activity of one protein. Increasing the number of phosphorylation sites significantly broadens the complexity of molecular mechanisms involved in processing multiple phosphorylation sites by one or more distinct kinases. The cardiac ryanodine receptor (RYR2) is a well-established multiple phospho-target of kinases activated in response to β-adrenergic stimulation because this Ca2+ channel is a critical component of Ca2+ handling machinery which is responsible for β-adrenergic enhancement of cardiac contractility. Our review presents a selective overview of the extensive, often conflicting, literature which focuses on identifying reliable lines of evidence to establish if multiple RYR2 phosphorylation is achieved randomly or in a specific sequence, and whether phosphorylation at individual sites is functionally specific and additive or similar and can therefore be substituted.
Collapse
|
12
|
Evans AM. On a Magical Mystery Tour with 8-Bromo-Cyclic ADP-Ribose: From All-or-None Block to Nanojunctions and the Cell-Wide Web. Molecules 2020; 25:E4768. [PMID: 33081414 PMCID: PMC7587525 DOI: 10.3390/molecules25204768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 09/08/2020] [Indexed: 11/16/2022] Open
Abstract
A plethora of cellular functions are controlled by calcium signals, that are greatly coordinated by calcium release from intracellular stores, the principal component of which is the sarco/endooplasmic reticulum (S/ER). In 1997 it was generally accepted that activation of various G protein-coupled receptors facilitated inositol-1,4,5-trisphosphate (IP3) production, activation of IP3 receptors and thus calcium release from S/ER. Adding to this, it was evident that S/ER resident ryanodine receptors (RyRs) could support two opposing cellular functions by delivering either highly localised calcium signals, such as calcium sparks, or by carrying propagating, global calcium waves. Coincidentally, it was reported that RyRs in mammalian cardiac myocytes might be regulated by a novel calcium mobilising messenger, cyclic adenosine diphosphate-ribose (cADPR), that had recently been discovered by HC Lee in sea urchin eggs. A reputedly selective and competitive cADPR antagonist, 8-bromo-cADPR, had been developed and was made available to us. We used 8-bromo-cADPR to further explore our observation that S/ER calcium release via RyRs could mediate two opposing functions, namely pulmonary artery dilation and constriction, in a manner seemingly independent of IP3Rs or calcium influx pathways. Importantly, the work of others had shown that, unlike skeletal and cardiac muscles, smooth muscles might express all three RyR subtypes. If this were the case in our experimental system and cADPR played a role, then 8-bromo-cADPR would surely block one of the opposing RyR-dependent functions identified, or the other, but certainly not both. The latter seemingly implausible scenario was confirmed. How could this be, do cells hold multiple, segregated SR stores that incorporate different RyR subtypes in receipt of spatially segregated signals carried by cADPR? The pharmacological profile of 8-bromo-cADPR action supported not only this, but also indicated that intracellular calcium signals were delivered across intracellular junctions formed by the S/ER. Not just one, at least two. This article retraces the steps along this journey, from the curious pharmacological profile of 8-bromo-cADPR to the discovery of the cell-wide web, a diverse network of cytoplasmic nanocourses demarcated by S/ER nanojunctions, which direct site-specific calcium flux and may thus coordinate the full panoply of cellular processes.
Collapse
Grants
- 01/A/S/07453 Biotechnology and Biological Sciences Research Council
- WT046374 , WT056423, WT070772, WT074434, WT081195AIA, WT212923, WT093147 Wellcome Trust
- PG/10/95/28657 British Heart Foundation
- FS/03/033/15432, FS/05/050, PG/05/128/19884, RG/12/14/29885, PG/10/95/28657 British Heart Foundation
- RG/12/14/29885 British Heart Foundation
Collapse
Affiliation(s)
- A Mark Evans
- Centre for Discovery Brain Sciences and Cardiovascular Science, Edinburgh Medical School, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, UK
| |
Collapse
|
13
|
Njegic A, Wilson C, Cartwright EJ. Targeting Ca 2 + Handling Proteins for the Treatment of Heart Failure and Arrhythmias. Front Physiol 2020; 11:1068. [PMID: 33013458 PMCID: PMC7498719 DOI: 10.3389/fphys.2020.01068] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/04/2020] [Indexed: 12/18/2022] Open
Abstract
Diseases of the heart, such as heart failure and cardiac arrhythmias, are a growing socio-economic burden. Calcium (Ca2+) dysregulation is key hallmark of the failing myocardium and has long been touted as a potential therapeutic target in the treatment of a variety of cardiovascular diseases (CVD). In the heart, Ca2+ is essential for maintaining normal cardiac function through the generation of the cardiac action potential and its involvement in excitation contraction coupling. As such, the proteins which regulate Ca2+ cycling and signaling play a vital role in maintaining Ca2+ homeostasis. Changes to the expression levels and function of Ca2+-channels, pumps and associated intracellular handling proteins contribute to altered Ca2+ homeostasis in CVD. The remodeling of Ca2+-handling proteins therefore results in impaired Ca2+ cycling, Ca2+ leak from the sarcoplasmic reticulum and reduced Ca2+ clearance, all of which contributes to increased intracellular Ca2+. Currently, approved treatments for targeting Ca2+ handling dysfunction in CVD are focused on Ca2+ channel blockers. However, whilst Ca2+ channel blockers have been successful in the treatment of some arrhythmic disorders, they are not universally prescribed to heart failure patients owing to their ability to depress cardiac function. Despite the progress in CVD treatments, there remains a clear need for novel therapeutic approaches which are able to reverse pathophysiology associated with heart failure and arrhythmias. Given that heart failure and cardiac arrhythmias are closely associated with altered Ca2+ homeostasis, this review will address the molecular changes to proteins associated with both Ca2+-handling and -signaling; their potential as novel therapeutic targets will be discussed in the context of pre-clinical and, where available, clinical data.
Collapse
Affiliation(s)
- Alexandra Njegic
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, United Kingdom.,Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Claire Wilson
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, United Kingdom.,Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Elizabeth J Cartwright
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
14
|
Federico M, Valverde CA, Mattiazzi A, Palomeque J. Unbalance Between Sarcoplasmic Reticulum Ca 2 + Uptake and Release: A First Step Toward Ca 2 + Triggered Arrhythmias and Cardiac Damage. Front Physiol 2020; 10:1630. [PMID: 32038301 PMCID: PMC6989610 DOI: 10.3389/fphys.2019.01630] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/24/2019] [Indexed: 12/19/2022] Open
Abstract
The present review focusses on the regulation and interplay of cardiac SR Ca2+ handling proteins involved in SR Ca2+ uptake and release, i.e., SERCa2/PLN and RyR2. Both RyR2 and SERCA2a/PLN are highly regulated by post-translational modifications and/or different partners' proteins. These control mechanisms guarantee a precise equilibrium between SR Ca2+ reuptake and release. The review then discusses how disruption of this balance alters SR Ca2+ handling and may constitute a first step toward cardiac damage and malignant arrhythmias. In the last part of the review, this concept is exemplified in different cardiac diseases, like prediabetic and diabetic cardiomyopathy, digitalis intoxication and ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Marilén Federico
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", CCT-La Plata/CONICET, Facultad de Cs. Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Carlos A Valverde
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", CCT-La Plata/CONICET, Facultad de Cs. Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Alicia Mattiazzi
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", CCT-La Plata/CONICET, Facultad de Cs. Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Julieta Palomeque
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", CCT-La Plata/CONICET, Facultad de Cs. Médicas, Universidad Nacional de La Plata, La Plata, Argentina.,Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Buenos Aires, Argentina
| |
Collapse
|
15
|
Asghari P, Scriven DR, Ng M, Panwar P, Chou KC, van Petegem F, Moore ED. Cardiac ryanodine receptor distribution is dynamic and changed by auxiliary proteins and post-translational modification. eLife 2020; 9:51602. [PMID: 31916935 PMCID: PMC6994221 DOI: 10.7554/elife.51602] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 01/09/2020] [Indexed: 12/14/2022] Open
Abstract
The effects of the immunophilins, FKBP12 and FKBP12.6, and phosphorylation on type II ryanodine receptor (RyR2) arrangement and function were examined using correlation microscopy (line scan confocal imaging of Ca2+ sparks and dual-tilt electron tomography) and dSTORM imaging of permeabilized Wistar rat ventricular myocytes. Saturating concentrations (10 µmol/L) of either FKBP12 or 12.6 significantly reduced the frequency, spread, amplitude and Ca2+ spark mass relative to control, while the tomograms revealed both proteins shifted the tetramers into a largely side-by-side configuration. Phosphorylation of immunophilin-saturated RyR2 resulted in structural and functional changes largely comparable to phosphorylation alone. dSTORM images of myocyte surfaces demonstrated that both FKBP12 and 12.6 significantly reduced RyR2 cluster sizes, while phosphorylation, even of immunophilin-saturated RyR2, increased them. We conclude that both RyR2 cluster size and the arrangement of tetramers within clusters is dynamic and respond to changes in the cellular environment. Further, these changes affect Ca2+ spark formation.
Collapse
Affiliation(s)
- Parisa Asghari
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - David Rl Scriven
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Myles Ng
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Pankaj Panwar
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Keng C Chou
- Department of Chemistry, University of British Columbia, Vancouver, Canada
| | - Filip van Petegem
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Edwin Dw Moore
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| |
Collapse
|
16
|
Kulkarni K, Merchant FM, Kassab MB, Sana F, Moazzami K, Sayadi O, Singh JP, Heist EK, Armoundas AA. Cardiac Alternans: Mechanisms and Clinical Utility in Arrhythmia Prevention. J Am Heart Assoc 2019; 8:e013750. [PMID: 31617437 PMCID: PMC6898836 DOI: 10.1161/jaha.119.013750] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Kanchan Kulkarni
- Cardiovascular Research CenterMassachusetts General HospitalBostonMA
| | | | - Mohamad B. Kassab
- Cardiovascular Research CenterMassachusetts General HospitalBostonMA
| | - Furrukh Sana
- Cardiovascular Research CenterMassachusetts General HospitalBostonMA
| | - Kasra Moazzami
- Cardiovascular Research CenterMassachusetts General HospitalBostonMA
| | - Omid Sayadi
- Cardiovascular Research CenterMassachusetts General HospitalBostonMA
| | - Jagmeet P. Singh
- Cardiology DivisionCardiac Arrhythmia ServiceMassachusetts General HospitalBostonMA
| | - E. Kevin Heist
- Cardiology DivisionCardiac Arrhythmia ServiceMassachusetts General HospitalBostonMA
| | - Antonis A. Armoundas
- Cardiovascular Research CenterMassachusetts General HospitalBostonMA
- Institute for Medical Engineering and ScienceMassachusetts Institute of TechnologyCambridgeMA
| |
Collapse
|
17
|
Potenza DM, Janicek R, Fernandez-Tenorio M, Camors E, Ramos-Mondragón R, Valdivia HH, Niggli E. Phosphorylation of the ryanodine receptor 2 at serine 2030 is required for a complete β-adrenergic response. J Gen Physiol 2018; 151:131-145. [PMID: 30541771 PMCID: PMC6363414 DOI: 10.1085/jgp.201812155] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/12/2018] [Accepted: 10/22/2018] [Indexed: 11/20/2022] Open
Abstract
Phosphorylation is thought to play a role in modulation of the ryanodine receptor 2 channel. Using a S2030A knock-in mouse model, Potenza et al. reveal that phosphorylation of RyR2-S2030 mediates channel regulation during the β-adrenergic response. During physical exercise or stress, the sympathetic system stimulates cardiac contractility via β-adrenergic receptor (β-AR) activation, resulting in protein kinase A (PKA)–mediated phosphorylation of the cardiac ryanodine receptor RyR2. PKA-dependent “hyperphosphorylation” of the RyR2 channel has been proposed as a major impairment that contributes to progression of heart failure. However, the sites of PKA phosphorylation and their phosphorylation status in cardiac diseases are not well defined. Among the known RyR2 phosphorylation sites, serine 2030 (S2030) remains highly controversial as a site of functional impact. We examined the contribution of RyR2-S2030 to Ca2+ signaling and excitation–contraction coupling (ECC) in a transgenic mouse with an ablated RyR2-S2030 phosphorylation site (RyR2-S2030A+/+). We assessed ECC gain by using whole-cell patch–clamp recordings and confocal Ca2+ imaging during β-ARs stimulation with isoproterenol (Iso) and consistent SR Ca2+ loading and L-type Ca2+ current (ICa) triggering. Under these conditions, ECC gain is diminished in mutant compared with WT cardiomyocytes. Resting Ca2+ spark frequency (CaSpF) with Iso is also reduced by mutation of S2030. In permeabilized cells, when SR Ca2+ pump activity is kept constant (using 2D12 antibody against phospholamban), cAMP does not change CaSpF in S2030A+/+ myocytes. Using Ca2+ spark recovery analysis, we found that mutant RyR Ca2+ sensitivity is not enhanced by Iso application, contrary to WT RyRs. Furthermore, ablation of RyR2-S2030 prevents acceleration of Ca2+ waves and increases latency to the first spontaneous Ca2+ release after a train of stimulations during Iso treatment. Together, these results suggest that phosphorylation at S2030 may represent an important step in the modulation of RyR2 activity during β-adrenergic stimulation and a potential target for the development of new antiarrhythmic drugs.
Collapse
Affiliation(s)
| | | | | | - Emmanuel Camors
- Center for Arrhythmia Research, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Roberto Ramos-Mondragón
- Center for Arrhythmia Research, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Héctor H Valdivia
- Department of Medicine, Wisconsin Institutes for Medical Research, University of Wisconsin, Madison, WI.,Center for Arrhythmia Research, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Ernst Niggli
- Department of Physiology, University of Bern, Bern, Switzerland
| |
Collapse
|
18
|
Niu Y, Kane AD, Lusby CM, Allison BJ, Chua YY, Kaandorp JJ, Nevin-Dolan R, Ashmore TJ, Blackmore HL, Derks JB, Ozanne SE, Giussani DA. Maternal Allopurinol Prevents Cardiac Dysfunction in Adult Male Offspring Programmed by Chronic Hypoxia During Pregnancy. Hypertension 2018; 72:971-978. [PMID: 30354714 PMCID: PMC6135482 DOI: 10.1161/hypertensionaha.118.11363] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Integrating functional and molecular levels, we investigated the effects of maternal treatment with a xanthine oxidase inhibitor on the programming of cardiac dysfunction in adult offspring using an established rat model of hypoxic pregnancy. Female Wistar rats were divided into normoxic or hypoxic (13% O2) pregnancy±maternal allopurinol treatment (30 mg kg-1 d-1). At 4 months, hearts were isolated from 1 male per litter per outcome variable to determine cardiac function and responses to ischemia-reperfusion in a Langendorff preparation. Sympathetic dominance, perfusate CK (creatine kinase) and LDH (lactate dehydrogenase) and the cardiac protein expression of the β1-adrenergic receptor, the M2 Ach receptor (muscarinic type-2 acetylcholine receptor), and the SERCA2a (sarcoplasmic reticulum Ca2+ ATPase 2a) were determined. Relative to controls, offspring from hypoxic pregnancy showed elevated left ventricular end diastolic pressure (+34.7%), enhanced contractility (dP/dtmax, +41.6%), reduced coronary flow rate (-21%) and an impaired recovery to ischemia-reperfusion (left ventricular diastolic pressure, area under the curve recovery -19.1%; all P<0.05). Increased sympathetic reactivity (heart rate, +755.5%; left ventricular diastolic pressure, +418.9%) contributed to the enhanced myocardial contractility ( P<0.05). Perfusate CK (+431%) and LDH (+251.3%) and the cardiac expression of SERCA2a (+71.4%) were also elevated ( P<0.05), further linking molecular markers of cardiac stress and injury to dysfunction. Maternal allopurinol restored all functional and molecular indices of cardiac pathology. The data support a link between xanthine oxidase-derived oxidative stress in hypoxic pregnancy and cardiac dysfunction in the adult offspring, providing a target for early intervention in the developmental programming of heart disease.
Collapse
Affiliation(s)
- Youguo Niu
- From the Department of Physiology, Development, and Neuroscience, University of Cambridge, United Kingdom (Y.N., A.D.K., C.M.L., B.J.A., Y.Y.C., R.N.-D., D.A.G.)
- Cambridge Cardiovascular Strategic Research Initiative (Y.N., S.E.O., D.A.G.)
| | - Andrew D. Kane
- From the Department of Physiology, Development, and Neuroscience, University of Cambridge, United Kingdom (Y.N., A.D.K., C.M.L., B.J.A., Y.Y.C., R.N.-D., D.A.G.)
| | - Ciara M. Lusby
- From the Department of Physiology, Development, and Neuroscience, University of Cambridge, United Kingdom (Y.N., A.D.K., C.M.L., B.J.A., Y.Y.C., R.N.-D., D.A.G.)
| | - Beth J. Allison
- From the Department of Physiology, Development, and Neuroscience, University of Cambridge, United Kingdom (Y.N., A.D.K., C.M.L., B.J.A., Y.Y.C., R.N.-D., D.A.G.)
| | - Yi Yi Chua
- From the Department of Physiology, Development, and Neuroscience, University of Cambridge, United Kingdom (Y.N., A.D.K., C.M.L., B.J.A., Y.Y.C., R.N.-D., D.A.G.)
| | - Joepe J. Kaandorp
- University Medical Center, Utrecht, the Netherlands (J.J.K., J.B.D.)
| | - Rhiannon Nevin-Dolan
- From the Department of Physiology, Development, and Neuroscience, University of Cambridge, United Kingdom (Y.N., A.D.K., C.M.L., B.J.A., Y.Y.C., R.N.-D., D.A.G.)
| | - Thomas J. Ashmore
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom (T.J.A., H.L.B., S.E.O.)
| | - Heather L. Blackmore
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom (T.J.A., H.L.B., S.E.O.)
| | - Jan B. Derks
- University Medical Center, Utrecht, the Netherlands (J.J.K., J.B.D.)
| | - Susan E. Ozanne
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom (T.J.A., H.L.B., S.E.O.)
- Cambridge Cardiovascular Strategic Research Initiative (Y.N., S.E.O., D.A.G.)
| | - Dino A. Giussani
- From the Department of Physiology, Development, and Neuroscience, University of Cambridge, United Kingdom (Y.N., A.D.K., C.M.L., B.J.A., Y.Y.C., R.N.-D., D.A.G.)
- Cambridge Cardiovascular Strategic Research Initiative (Y.N., S.E.O., D.A.G.)
| |
Collapse
|
19
|
Chen M, Xu D, Wu AZ, Kranias E, Lin SF, Chen PS, Chen Z. Phospholamban regulates nuclear Ca 2+ stores and inositol 1,4,5-trisphosphate mediated nuclear Ca 2+ cycling in cardiomyocytes. J Mol Cell Cardiol 2018; 123:185-197. [PMID: 30261161 DOI: 10.1016/j.yjmcc.2018.09.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/04/2018] [Accepted: 09/21/2018] [Indexed: 01/15/2023]
Abstract
AIMS Phospholamban (PLB) is the key regulator of the cardiac Ca2+ pump (SERCA2a)-mediated sarcoplasmic reticulum Ca2+ stores. We recently reported that PLB is highly concentrated in the nuclear envelope (NE) from where it can modulate perinuclear Ca2+ handling of the cardiomyocytes (CMs). Since inositol 1,4,5-trisphosphate (IP3) receptor (IP3R) mediates nuclear Ca2+ release, we examined whether the nuclear pool of PLB regulates IP3-induced nuclear Ca2+ handling. METHODS AND RESULTS Fluo-4 based confocal Ca2+ imaging was performed to measure Ca2+ dynamics across both nucleus and cytosol in saponin-permeabilized CMs isolated from wild-type (WT) or PLB-knockout (PLB-KO) mice. At diastolic intracellular Ca2+ ([Ca2+]i = 100 nM), the Fab fragment of the monoclonal PLB antibody (anti-PLB Fab) facilitated the formation and increased the length of spontaneous Ca2+ waves (SCWs) originating from the nuclear region in CMs from WT but not from PLB-KO mice. We next examined nuclear Ca2+ activities at basal condition and after sequential addition of IP3, anti-PLB Fab, and the IP3R inhibitor 2-aminoethoxydiphenyl borate (2-APB) at a series of [Ca2+]i. In WT mice, at 10 nM [Ca2+]i where ryanodine receptor (RyR2) based spontaneous Ca2+ sparks rarely occurred, IP3 increased fluorescence amplitude (F/F0) of overall nuclear region to 1.19 ± 0.02. Subsequent addition of anti-PLB Fab significantly decreased F/F0 to 1.09 ± 0.02. At 50 nM [Ca2+]i, anti-PLB Fab not only decreased the overall nuclear F/F0 previously elevated by IP3, but also increased the amplitude and duration of spark-like nuclear Ca2+ release events. These nuclear Ca2+ releases were blocked by 2-APB. At 100 nM [Ca2+]i, IP3 induced short SCWs originating from nucleus. Anti-PLB Fab transformed those short waves into long SCWs with propagation from the nucleus into the cytosol. In contrast, neither nuclear nor cytosolic Ca2+ dynamics was affected by anti-PLB Fab in CMs from PLB-KO mice in all these conditions. Furthermore, in WT CMs pretreated with RyR2 blocker tetracaine, IP3 and anti-PLB Fab still increased the magnitude of nuclear Ca2+ release but failed to regenerate SCWs. Finally, anti-PLB Fab increased low Ca2+ affinity mag-fluo 4 fluorescence intensity in the lumen of NE of nuclei isolated from WT but not in PLB-KO mice. CONCLUSION PLB regulates nuclear Ca2+ handling. By increasing Ca2+ uptake into lumen of the NE and perhaps other perinuclear membranes, the acute reversal of PLB inhibition decreases global Ca2+ concentration at rest in the nucleoplasm, and increases Ca2+ release into the nucleus, through mechanisms involving IP3R and RyR2 in the vicinity.
Collapse
Affiliation(s)
- Mu Chen
- Krannert Institute of Cardiology, Indiana University, Indianapolis, IN, USA; Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dongzhu Xu
- Krannert Institute of Cardiology, Indiana University, Indianapolis, IN, USA; Cardiovascular Division, Institute of Clinical Medicine, Faculty of Medicine, University of Tsukuba, Japan
| | - Adonis Z Wu
- Krannert Institute of Cardiology, Indiana University, Indianapolis, IN, USA
| | - Evangelia Kranias
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Shien-Fong Lin
- Krannert Institute of Cardiology, Indiana University, Indianapolis, IN, USA; Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsin-Chu, Taiwan
| | - Peng-Sheng Chen
- Krannert Institute of Cardiology, Indiana University, Indianapolis, IN, USA
| | - Zhenhui Chen
- Krannert Institute of Cardiology, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
20
|
Gonano LA, Jones PP. FK506-binding proteins 12 and 12.6 (FKBPs) as regulators of cardiac Ryanodine Receptors: Insights from new functional and structural knowledge. Channels (Austin) 2017. [PMID: 28636428 DOI: 10.1080/19336950.2017.1344799] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Ryanodine Receptors (RyRs) are intracellular Ca2+ channels that mediate Ca2+ flux from the sarco(endo)plasmic reticulum in many cell types. The interaction of RyRs with FK506-binding proteins (FKBPs) has been proposed as an important regulatory mechanism, where the loss of this interaction leads to channel dysfunction. In the heart, phosphorylation of RyR has been suggested to disrupt the RyR-FKBP interaction promoting altered Ca2+ signaling, heart failure and arrhythmias. However, the functional result of FKBP interaction with RyR and how this interaction is regulated remains highly controversial. Recently, high resolution structures of RyR have provided novel aspects to the ongoing debate. This review will discuss the most recent functional data in light of these new structures.
Collapse
Affiliation(s)
- Luis A Gonano
- a Department of Physiology , School of Biomedical Sciences and HeartOtago, University of Otago , Dunedin, Otago , New Zealand
| | - Peter P Jones
- a Department of Physiology , School of Biomedical Sciences and HeartOtago, University of Otago , Dunedin, Otago , New Zealand
| |
Collapse
|
21
|
Abstract
There has been a significant progress in our understanding of the molecular mechanisms by which calcium (Ca2+) ions mediate various types of cardiac arrhythmias. A growing list of inherited gene defects can cause potentially lethal cardiac arrhythmia syndromes, including catecholaminergic polymorphic ventricular tachycardia, congenital long QT syndrome, and hypertrophic cardiomyopathy. In addition, acquired deficits of multiple Ca2+-handling proteins can contribute to the pathogenesis of arrhythmias in patients with various types of heart disease. In this review article, we will first review the key role of Ca2+ in normal cardiac function-in particular, excitation-contraction coupling and normal electric rhythms. The functional involvement of Ca2+ in distinct arrhythmia mechanisms will be discussed, followed by various inherited arrhythmia syndromes caused by mutations in Ca2+-handling proteins. Finally, we will discuss how changes in the expression of regulation of Ca2+ channels and transporters can cause acquired arrhythmias, and how these mechanisms might be targeted for therapeutic purposes.
Collapse
Affiliation(s)
- Andrew P Landstrom
- From the Section of Cardiology, Department of Pediatrics (A.P.L.), Cardiovascular Research Institute (A.P.L., X.H.T.W.), and Departments of Molecular Physiology and Biophysics, Medicine (Cardiology), Center for Space Medicine (X.H.T.W.), Baylor College of Medicine, Houston, TX; and Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.)
| | - Dobromir Dobrev
- From the Section of Cardiology, Department of Pediatrics (A.P.L.), Cardiovascular Research Institute (A.P.L., X.H.T.W.), and Departments of Molecular Physiology and Biophysics, Medicine (Cardiology), Center for Space Medicine (X.H.T.W.), Baylor College of Medicine, Houston, TX; and Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.)
| | - Xander H T Wehrens
- From the Section of Cardiology, Department of Pediatrics (A.P.L.), Cardiovascular Research Institute (A.P.L., X.H.T.W.), and Departments of Molecular Physiology and Biophysics, Medicine (Cardiology), Center for Space Medicine (X.H.T.W.), Baylor College of Medicine, Houston, TX; and Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.).
| |
Collapse
|
22
|
Pereira L, Bare DJ, Galice S, Shannon TR, Bers DM. β-Adrenergic induced SR Ca 2+ leak is mediated by an Epac-NOS pathway. J Mol Cell Cardiol 2017; 108:8-16. [PMID: 28476660 DOI: 10.1016/j.yjmcc.2017.04.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/26/2017] [Accepted: 04/27/2017] [Indexed: 02/08/2023]
Abstract
Cardiac β-adrenergic receptors (β-AR) and Ca2+-Calmodulin dependent protein kinase (CaMKII) regulate both physiological and pathophysiological Ca2+ signaling. Elevated diastolic Ca2+ leak from the sarcoplasmic reticulum (SR) contributes to contractile dysfunction in heart failure and to arrhythmogenesis. β-AR activation is known to increase SR Ca2+ leak via CaMKII-dependent phosphorylation of the ryanodine receptor. Two independent and reportedly parallel pathways have been implicated in this β-AR-CaMKII cascade, one involving exchange protein directly activated by cAMP (Epac2) and another involving nitric oxide synthase 1 (NOS1). Here we tested whether Epac and NOS function in a single series pathway to increase β-AR induced and CaMKII-dependent SR Ca2+ leak. Leak was measured as both Ca2+ spark frequency and tetracaine-induced shifts in SR Ca2+, in mouse and rabbit ventricular myocytes. Direct Epac activation by 8-CPT (8-(4-chlorophenylthio)-2'-O-methyl-cAMP) mimicked β-AR-induced SR Ca2+ leak, and both were blocked by NOS inhibition. The same was true for myocyte CaMKII activation (assessed via a FRET-based reporter) and ryanodine receptor phosphorylation. Inhibitor and phosphorylation studies also implicated phosphoinositide 3-kinase (PI3K) and protein kinase B (Akt) downstream of Epac and above NOS activation in this pathway. We conclude that these two independently characterized parallel pathways function mainly via a single series arrangement (β-AR-cAMP-Epac-PI3K-Akt-NOS1-CaMKII) to mediate increased SR Ca2+ leak. Thus, for β-AR activation the cAMP-PKA branch effects inotropy and lusitropy (by effects on Ca2+ current and SR Ca2+-ATPase), this cAMP-Epac-NOS pathway increases pathological diastolic SR Ca2+leak. This pathway distinction may allow novel SR Ca2+ leak therapeutic targeting in treatment of arrhythmias in heart failure that spare the inotropic and lusitropic effects of the PKA branch.
Collapse
Affiliation(s)
- Laëtitia Pereira
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, United States
| | - Dan J Bare
- Department of Molecular Biophysics and Physiology, Rush University, Chicago, IL 60612, United States
| | - Samuel Galice
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, United States
| | - Thomas R Shannon
- Department of Molecular Biophysics and Physiology, Rush University, Chicago, IL 60612, United States.
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, United States.
| |
Collapse
|
23
|
Tykocki NR, Boerman EM, Jackson WF. Smooth Muscle Ion Channels and Regulation of Vascular Tone in Resistance Arteries and Arterioles. Compr Physiol 2017; 7:485-581. [PMID: 28333380 DOI: 10.1002/cphy.c160011] [Citation(s) in RCA: 241] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Vascular tone of resistance arteries and arterioles determines peripheral vascular resistance, contributing to the regulation of blood pressure and blood flow to, and within the body's tissues and organs. Ion channels in the plasma membrane and endoplasmic reticulum of vascular smooth muscle cells (SMCs) in these blood vessels importantly contribute to the regulation of intracellular Ca2+ concentration, the primary determinant of SMC contractile activity and vascular tone. Ion channels provide the main source of activator Ca2+ that determines vascular tone, and strongly contribute to setting and regulating membrane potential, which, in turn, regulates the open-state-probability of voltage gated Ca2+ channels (VGCCs), the primary source of Ca2+ in resistance artery and arteriolar SMCs. Ion channel function is also modulated by vasoconstrictors and vasodilators, contributing to all aspects of the regulation of vascular tone. This review will focus on the physiology of VGCCs, voltage-gated K+ (KV) channels, large-conductance Ca2+-activated K+ (BKCa) channels, strong-inward-rectifier K+ (KIR) channels, ATP-sensitive K+ (KATP) channels, ryanodine receptors (RyRs), inositol 1,4,5-trisphosphate receptors (IP3Rs), and a variety of transient receptor potential (TRP) channels that contribute to pressure-induced myogenic tone in resistance arteries and arterioles, the modulation of the function of these ion channels by vasoconstrictors and vasodilators, their role in the functional regulation of tissue blood flow and their dysfunction in diseases such as hypertension, obesity, and diabetes. © 2017 American Physiological Society. Compr Physiol 7:485-581, 2017.
Collapse
Affiliation(s)
- Nathan R Tykocki
- Department of Pharmacology, University of Vermont, Burlington, Vermont, USA
| | - Erika M Boerman
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
| | - William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
24
|
The effect of PKA-mediated phosphorylation of ryanodine receptor on SR Ca 2+ leak in ventricular myocytes. J Mol Cell Cardiol 2017; 104:9-16. [PMID: 28131630 DOI: 10.1016/j.yjmcc.2017.01.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 11/22/2016] [Accepted: 01/24/2017] [Indexed: 01/08/2023]
Abstract
Functional impact of cardiac ryanodine receptor (type 2 RyR or RyR2) phosphorylation by protein kinase A (PKA) remains highly controversial. In this study, we characterized a functional link between PKA-mediated RyR2 phosphorylation level and sarcoplasmic reticulum (SR) Ca2+ release and leak in permeabilized rabbit ventricular myocytes. Changes in cytosolic [Ca2+] and intra-SR [Ca2+]SR were measured with Fluo-4 and Fluo-5N, respectively. Changes in RyR2 phosphorylation at two PKA sites, serine-2031 and -2809, were measured with phospho-specific antibodies. cAMP (10μM) increased Ca2+ spark frequency approximately two-fold. This effect was associated with an increase in SR Ca2+ load from 0.84 to 1.24mM. PKA inhibitory peptide (PKI; 10μM) abolished the cAMP-dependent increase of SR Ca2+ load and spark frequency. When SERCA was completely blocked by thapsigargin, cAMP did not affect RyR2-mediated Ca2+ leak. The lack of a cAMP effect on RyR2 function can be explained by almost maximal phosphorylation of RyR2 at serine-2809 after sarcolemma permeabilization. This high RyR2 phosphorylation level is likely the consequence of a balance shift between protein kinase and phosphatase activity after permeabilization. When RyR2 phosphorylation at serine-2809 was reduced to its "basal" level (i.e. RyR2 phosphorylation level in intact myocytes) using kinase inhibitor staurosporine, SR Ca2+ leak was significantly reduced. Surprisingly, further dephosphorylation of RyR2 with protein phosphatase 1 (PP1) markedly increased SR Ca2+ leak. At the same time, phosphorylation of RyR2 at serine 2031 did not significantly change under identical experimental conditions. These results suggest that RyR2 phosphorylation by PKA has a complex effect on SR Ca2+ leak in ventricular myocytes. At an intermediate level of RyR2 phosphorylation SR Ca2+ leak is minimal. However, complete dephosphorylation and maximal phosphorylation of RyR2 increases SR Ca2+ leak.
Collapse
|
25
|
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
|
26
|
Crocini C, Coppini R, Ferrantini C, Yan P, Loew LM, Poggesi C, Cerbai E, Pavone FS, Sacconi L. T-Tubular Electrical Defects Contribute to Blunted β-Adrenergic Response in Heart Failure. Int J Mol Sci 2016; 17:ijms17091471. [PMID: 27598150 PMCID: PMC5037749 DOI: 10.3390/ijms17091471] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 08/17/2016] [Accepted: 08/30/2016] [Indexed: 11/18/2022] Open
Abstract
Alterations of the β-adrenergic signalling, structural remodelling, and electrical failure of T-tubules are hallmarks of heart failure (HF). Here, we assess the effect of β-adrenoceptor activation on local Ca2+ release in electrically coupled and uncoupled T-tubules in ventricular myocytes from HF rats. We employ an ultrafast random access multi-photon (RAMP) microscope to simultaneously record action potentials and Ca2+ transients from multiple T-tubules in ventricular cardiomyocytes from a HF rat model of coronary ligation compared to sham-operated rats as a control. We confirmed that β-adrenergic stimulation increases the frequency of Ca2+ sparks, reduces Ca2+ transient variability, and hastens the decay of Ca2+ transients: all these effects are similarly exerted by β-adrenergic stimulation in control and HF cardiomyocytes. Conversely, β-adrenergic stimulation in HF cells accelerates a Ca2+ rise exclusively in the proximity of T-tubules that regularly conduct the action potential. The delayed Ca2+ rise found at T-tubules that fail to conduct the action potential is instead not affected by β-adrenergic signalling. Taken together, these findings indicate that HF cells globally respond to β-adrenergic stimulation, except at T-tubules that fail to conduct action potentials, where the blunted effect of the β-adrenergic signalling may be directly caused by the lack of electrical activity.
Collapse
Affiliation(s)
- Claudia Crocini
- European Laboratory for Non-Linear Spectroscopy, Florence 50019, Italy.
- National Institute of Optics, National Research Council, Florence 50125, Italy.
| | - Raffaele Coppini
- Division of Pharmacology, Department "NeuroFarBa", University of Florence, Florence 50139, Italy.
| | - Cecilia Ferrantini
- Division of Physiology, Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy.
| | - Ping Yan
- R. D. Berlin Center for Cell Analysis and Modeling, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | - Leslie M Loew
- R. D. Berlin Center for Cell Analysis and Modeling, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | - Corrado Poggesi
- Division of Physiology, Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy.
| | - Elisabetta Cerbai
- Division of Pharmacology, Department "NeuroFarBa", University of Florence, Florence 50139, Italy.
| | - Francesco S Pavone
- European Laboratory for Non-Linear Spectroscopy, Florence 50019, Italy.
- Department of Physics and Astronomy, University of Florence, Sesto Fiorentino 50019, Italy.
| | - Leonardo Sacconi
- European Laboratory for Non-Linear Spectroscopy, Florence 50019, Italy.
- National Institute of Optics, National Research Council, Florence 50125, Italy.
| |
Collapse
|
27
|
Salazar-Cantú A, Pérez-Treviño P, Montalvo-Parra D, Balderas-Villalobos J, Gómez-Víquez NL, García N, Altamirano J. Role of SERCA and the sarcoplasmic reticulum calcium content on calcium waves propagation in rat ventricular myocytes. Arch Biochem Biophys 2016; 604:11-9. [DOI: 10.1016/j.abb.2016.05.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 05/14/2016] [Accepted: 05/26/2016] [Indexed: 11/25/2022]
|
28
|
CaMKII-dependent phosphorylation of RyR2 promotes targetable pathological RyR2 conformational shift. J Mol Cell Cardiol 2016; 98:62-72. [PMID: 27318036 DOI: 10.1016/j.yjmcc.2016.06.007] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 05/27/2016] [Accepted: 06/14/2016] [Indexed: 01/27/2023]
Abstract
Diastolic calcium (Ca) leak via cardiac ryanodine receptors (RyR2) can cause arrhythmias and heart failure (HF). Ca/calmodulin (CaM)-dependent kinase II (CaMKII) is upregulated and more active in HF, promoting RyR2-mediated Ca leak by RyR2-Ser2814 phosphorylation. Here, we tested a mechanistic hypothesis that RyR2 phosphorylation by CaMKII increases Ca leak by promoting a pathological RyR2 conformation with reduced CaM affinity. Acute CaMKII activation in wild-type RyR2, and phosphomimetic RyR2-S2814D (vs. non-phosphorylatable RyR2-S2814A) knock-in mouse myocytes increased SR Ca leak, reduced CaM-RyR2 affinity, and caused a pathological shift in RyR2 conformation (detected via increased access of the RyR2 structural peptide DPc10). This same trio of effects was seen in myocytes from rabbits with pressure/volume-overload induced HF. Excess CaM quieted leak and restored control conformation, consistent with negative allosteric coupling between CaM affinity and DPc10 accessible conformation. Dantrolene (DAN) also restored CaM affinity, reduced DPc10 access, and suppressed RyR2-mediated Ca leak and ventricular tachycardia in RyR2-S2814D mice. We propose that a common pathological RyR2 conformational state (low CaM affinity, high DPc10 access, and elevated leak) may be caused by CaMKII-dependent phosphorylation, oxidation, and HF. Moreover, DAN (or excess CaM) can shift this pathological gating state back to the normal physiological conformation, a potentially important therapeutic approach.
Collapse
|
29
|
Zhang Y, Chen A, Song L, Li M, Luo Z, Zhang W, Chen Y, He B. Low-Level Vagus Nerve Stimulation Reverses Cardiac Dysfunction and Subcellular Calcium Handling in Rats With Post-Myocardial Infarction Heart Failure. Int Heart J 2016; 57:350-5. [PMID: 27181040 DOI: 10.1536/ihj.15-516] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Vagus nerve stimulation (VNS), targeting the imbalanced autonomic nervous system, is a promising therapeutic approach for chronic heart failure (HF). Moreover, calcium cycling is an important part of cardiac excitation-contraction coupling (ECC), which also participates in the antiarrhythmic effects of VNS. We hypothesized that low-level VNS (LL-VNS) could improve cardiac function by regulation of intracellular calcium handling properties. The experimental HF model was established by ligation of the left anterior descending coronary artery (LAD). Thirty-two male Sprague-Dawley rats were divided into 3 groups as follows; control group (sham operated without coronary ligation, n = 10), HF-VNS group (HF rats with VNS, n = 12), and HF-SS group (HF rats with sham nerve stimulation, n = 10). After 8 weeks of treatment, LL-VNS significantly improved left ventricular ejection fraction (LVEF) and attenuated myocardial interstitial fibrosis in the HF-VNS group compared with the HF-SS group. Elevated plasma norepinephrine and dopamine, but not epinephrine, were partially reduced by LL-VNS. Additionally, LL-VNS restored the protein and mRNA levels of sarcoplasmic reticulum Ca(2+) ATPase (SERCA2a), Na(+)-Ca(2+) exchanger 1 (NCX1), and phospholamban (PLB) whereas the expression of ryanodine receptor 2 (RyR2) as well as mRNA level was unaffected. Thus, our study results suggest that the improvement of cardiac performance by LL-VNS is accompanied by the reversal of dysfunctional calcium handling properties including SERCA2a, NCX1, and PLB which may be a potential molecular mechanism of VNS for HF.
Collapse
Affiliation(s)
- Yunhe Zhang
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Mazzocchi G, Sommese L, Palomeque J, Felice JI, Di Carlo MN, Fainstein D, Gonzalez P, Contreras P, Skapura D, McCauley MD, Lascano EC, Negroni JA, Kranias EG, Wehrens XHT, Valverde CA, Mattiazzi A. Phospholamban ablation rescues the enhanced propensity to arrhythmias of mice with CaMKII-constitutive phosphorylation of RyR2 at site S2814. J Physiol 2016; 594:3005-30. [PMID: 26695843 DOI: 10.1113/jp271622] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 12/14/2015] [Indexed: 01/27/2023] Open
Abstract
KEY POINTS Mice with Ca(2+) -calmodulin-dependent protein kinase (CaMKII) constitutive pseudo-phosphorylation of the ryanodine receptor RyR2 at Ser2814 (S2814D(+/+) mice) exhibit a higher open probability of RyR2, higher sarcoplasmic reticulum (SR) Ca(2+) leak in diastole and increased propensity to arrhythmias under stress conditions. We generated phospholamban (PLN)-deficient S2814D(+/+) knock-in mice by crossing two colonies, S2814D(+/+) and PLNKO mice, to test the hypothesis that PLN ablation can prevent the propensity to arrhythmias of S2814D(+/+) mice. PLN ablation partially rescues the altered intracellular Ca(2+) dynamics of S2814D(+/+) hearts and myocytes, but enhances SR Ca(2+) sparks and leak on confocal microscopy. PLN ablation diminishes ventricular arrhythmias promoted by CaMKII phosphorylation of S2814 on RyR2. PLN ablation aborts the arrhythmogenic SR Ca(2+) waves of S2814D(+/+) and transforms them into non-propagating events. A mathematical human myocyte model replicates these results and predicts the increase in SR Ca(2+) uptake required to prevent the arrhythmias induced by a CaMKII-dependent leaky RyR2. ABSTRACT Mice with constitutive pseudo-phosphorylation at Ser2814-RyR2 (S2814D(+/+) ) have increased propensity to arrhythmias under β-adrenergic stress conditions. Although abnormal Ca(2+) release from the sarcoplasmic reticulum (SR) has been linked to arrhythmogenesis, the role played by SR Ca(2+) uptake remains controversial. We tested the hypothesis that an increase in SR Ca(2+) uptake is able to rescue the increased arrhythmia propensity of S2814D(+/+) mice. We generated phospholamban (PLN)-deficient/S2814D(+/+) knock-in mice by crossing two colonies, S2814D(+/+) and PLNKO mice (SD(+/+) /KO). SD(+/+) /KO myocytes exhibited both increased SR Ca(2+) uptake seen in PLN knock-out (PLNKO) myocytes and diminished SR Ca(2+) load (relative to PLNKO), a characteristic of S2814D(+/+) myocytes. Ventricular arrhythmias evoked by catecholaminergic challenge (caffeine/adrenaline) in S2814D(+/+) mice in vivo or programmed electric stimulation and high extracellular Ca(2+) in S2814D(+) /(-) hearts ex vivo were significantly diminished by PLN ablation. At the myocyte level, PLN ablation converted the arrhythmogenic Ca(2+) waves evoked by high extracellular Ca(2+) provocation in S2814D(+/+) mice into non-propagated Ca(2+) mini-waves on confocal microscopy. Myocyte Ca(2+) waves, typical of S2814D(+/+) mice, could be evoked in SD(+/+) /KO cells by partially inhibiting SERCA2a. A mathematical human myocyte model replicated these results and allowed for predicting the increase in SR Ca(2+) uptake required to prevent the arrhythmias induced by a Ca(2+) -calmodulin-dependent protein kinase (CaMKII)-dependent leaky RyR2. Our results demonstrate that increasing SR Ca(2+) uptake by PLN ablation can prevent the arrhythmic events triggered by SR Ca(2+) leak due to CaMKII-dependent phosphorylation of the RyR2-S2814 site and underscore the benefits of increasing SERCA2a activity on SR Ca(2+) -triggered arrhythmias.
Collapse
Affiliation(s)
- G Mazzocchi
- Centro de Investigaciones Cardiovasculares, CCT-La Plata-CONICET, Facultad de Cs Médicas, UNLP, La Plata, Argentina
| | - L Sommese
- Centro de Investigaciones Cardiovasculares, CCT-La Plata-CONICET, Facultad de Cs Médicas, UNLP, La Plata, Argentina
| | - J Palomeque
- Centro de Investigaciones Cardiovasculares, CCT-La Plata-CONICET, Facultad de Cs Médicas, UNLP, La Plata, Argentina
| | - J I Felice
- Centro de Investigaciones Cardiovasculares, CCT-La Plata-CONICET, Facultad de Cs Médicas, UNLP, La Plata, Argentina
| | - M N Di Carlo
- Centro de Investigaciones Cardiovasculares, CCT-La Plata-CONICET, Facultad de Cs Médicas, UNLP, La Plata, Argentina
| | - D Fainstein
- Centro de Investigaciones Cardiovasculares, CCT-La Plata-CONICET, Facultad de Cs Médicas, UNLP, La Plata, Argentina
| | - P Gonzalez
- Cátedra de Patología, Facultad de Cs Médicas, UNLP, La Plata, Argentina
| | - P Contreras
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - D Skapura
- Departments of Molecular Physiology and Biophysics, Medicine (in Cardiology), and Pediatrics, Baylor College of Medicine, Cardiovascular Research Institute, Houston, TX, 77030, USA
| | - M D McCauley
- Departments of Molecular Physiology and Biophysics, Medicine (in Cardiology), and Pediatrics, Baylor College of Medicine, Cardiovascular Research Institute, Houston, TX, 77030, USA
| | - E C Lascano
- Departamento de Biología Comparada, Celular y Molecular, Universidad Favaloro, Ciudad Autónoma de Buenos Aires, Argentina
| | - J A Negroni
- Departamento de Biología Comparada, Celular y Molecular, Universidad Favaloro, Ciudad Autónoma de Buenos Aires, Argentina
| | - E G Kranias
- Department of Pharmacology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267
| | - X H T Wehrens
- Departments of Molecular Physiology and Biophysics, Medicine (in Cardiology), and Pediatrics, Baylor College of Medicine, Cardiovascular Research Institute, Houston, TX, 77030, USA
| | - C A Valverde
- Centro de Investigaciones Cardiovasculares, CCT-La Plata-CONICET, Facultad de Cs Médicas, UNLP, La Plata, Argentina
| | - A Mattiazzi
- Centro de Investigaciones Cardiovasculares, CCT-La Plata-CONICET, Facultad de Cs Médicas, UNLP, La Plata, Argentina
| |
Collapse
|
31
|
Mesubi OO, Anderson ME. Atrial remodelling in atrial fibrillation: CaMKII as a nodal proarrhythmic signal. Cardiovasc Res 2016; 109:542-57. [PMID: 26762270 DOI: 10.1093/cvr/cvw002] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/05/2016] [Indexed: 01/10/2023] Open
Abstract
CaMKII is a serine-threonine protein kinase that is abundant in myocardium. Emergent evidence suggests that CaMKII may play an important role in promoting atrial fibrillation (AF) by targeting a diverse array of proteins involved in membrane excitability, cell survival, calcium homeostasis, matrix remodelling, inflammation, and metabolism. Furthermore, CaMKII inhibition appears to protect against AF in animal models and correct proarrhythmic, defective intracellular Ca(2+) homeostasis in fibrillating human atrial cells. This review considers current concepts and evidence from animal and human studies on the role of CaMKII in AF.
Collapse
Affiliation(s)
- Olurotimi O Mesubi
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA Department of Medicine, The Johns Hopkins University School of Medicine, 1830 E. Monument Street, Suite 9026, Baltimore, MD 21287, USA
| | - Mark E Anderson
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA Department of Medicine, The Johns Hopkins University School of Medicine, 1830 E. Monument Street, Suite 9026, Baltimore, MD 21287, USA Department of Physiology and the Program in Cellular and Molecular Medicine, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
32
|
Roe AT, Frisk M, Louch WE. Targeting cardiomyocyte Ca2+ homeostasis in heart failure. Curr Pharm Des 2015; 21:431-48. [PMID: 25483944 PMCID: PMC4475738 DOI: 10.2174/138161282104141204124129] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 08/06/2014] [Indexed: 12/19/2022]
Abstract
Improved treatments for heart failure patients will require the development of novel therapeutic strategies that target basal disease
mechanisms. Disrupted cardiomyocyte Ca2+ homeostasis is recognized as a major contributor to the heart failure phenotype, as it
plays a key role in systolic and diastolic dysfunction, arrhythmogenesis, and hypertrophy and apoptosis signaling. In this review, we outline
existing knowledge of the involvement of Ca2+ homeostasis in these deficits, and identify four promising targets for therapeutic intervention:
the sarcoplasmic reticulum Ca2+ ATPase, the Na+-Ca2+ exchanger, the ryanodine receptor, and t-tubule structure. We discuss
experimental data indicating the applicability of these targets that has led to recent and ongoing clinical trials, and suggest future therapeutic
approaches.
Collapse
Affiliation(s)
| | | | - William E Louch
- Institute for Experimental Medical Research, Kirkeveien 166, 4.etg. Bygg 7, Oslo University Hospital Ullevål, 0407 Oslo, Norway.
| |
Collapse
|
33
|
Erickson JR, Nichols CB, Uchinoumi H, Stein ML, Bossuyt J, Bers DM. S-Nitrosylation Induces Both Autonomous Activation and Inhibition of Calcium/Calmodulin-dependent Protein Kinase II δ. J Biol Chem 2015; 290:25646-56. [PMID: 26316536 DOI: 10.1074/jbc.m115.650234] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Indexed: 01/15/2023] Open
Abstract
NO is known to modulate calcium handling and cellular signaling in the myocardium, but key targets for NO in the heart remain unidentified. Recent reports have implied that NO can activate calcium/calmodulin (Ca(2+)/CaM)-dependent protein kinase II (CaMKII) in neurons and the heart. Here we use our novel sensor of CaMKII activation, Camui, to monitor changes in the conformation and activation of cardiac CaMKII (CaMKIIδ) activity after treatment with the NO donor S-nitrosoglutathione (GSNO). We demonstrate that exposure to NO after Ca(2+)/CaM binding to CaMKIIδ results in autonomous kinase activation, which is abolished by mutation of the Cys-290 site. However, exposure of CaMKIIδ to GSNO prior to Ca(2+)/CaM exposure strongly suppresses kinase activation and conformational change by Ca(2+)/CaM. This NO-induced inhibition was ablated by mutation of the Cys-273 site. We found parallel effects of GSNO on CaM/CaMKIIδ binding and CaMKIIδ-dependent ryanodine receptor activation in adult cardiac myocytes. We conclude that NO can play a dual role in regulating cardiac CaMKIIδ activity.
Collapse
Affiliation(s)
- Jeffrey R Erickson
- From the Department of Physiology, University of Otago, Dunedin 9016, New Zealand and
| | - C Blake Nichols
- the Department of Pharmacology, University of California, Davis, CA 95616
| | - Hitoshi Uchinoumi
- the Department of Pharmacology, University of California, Davis, CA 95616
| | - Matthew L Stein
- the Department of Pharmacology, University of California, Davis, CA 95616
| | - Julie Bossuyt
- the Department of Pharmacology, University of California, Davis, CA 95616
| | - Donald M Bers
- the Department of Pharmacology, University of California, Davis, CA 95616
| |
Collapse
|
34
|
Abstract
Optimal cardiac function depends on proper timing of excitation and contraction in various regions of the heart, as well as on appropriate heart rate. This is accomplished via specialized electrical properties of various components of the system, including the sinoatrial node, atria, atrioventricular node, His-Purkinje system, and ventricles. Here we review the major regionally determined electrical properties of these cardiac regions and present the available data regarding the molecular and ionic bases of regional cardiac function and dysfunction. Understanding these differences is of fundamental importance for the investigation of arrhythmia mechanisms and pharmacotherapy.
Collapse
Affiliation(s)
- Daniel C Bartos
- Department of Pharmacology, University of California Davis, Davis, California, USA
| | - Eleonora Grandi
- Department of Pharmacology, University of California Davis, Davis, California, USA
| | - Crystal M Ripplinger
- Department of Pharmacology, University of California Davis, Davis, California, USA
| |
Collapse
|
35
|
Oxidation of ryanodine receptor (RyR) and calmodulin enhance Ca release and pathologically alter, RyR structure and calmodulin affinity. J Mol Cell Cardiol 2015; 85:240-8. [PMID: 26092277 DOI: 10.1016/j.yjmcc.2015.06.009] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Revised: 05/12/2015] [Accepted: 06/12/2015] [Indexed: 11/21/2022]
Abstract
Oxidative stress may contribute to cardiac ryanodine receptor (RyR2) dysfunction in heart failure (HF) and arrhythmias. Altered RyR2 domain-domain interaction (domain unzipping) and calmodulin (CaM) binding affinity are allosterically coupled indices of RyR2 conformation. In HF RyR2 exhibits reduced CaM binding, increased domain unzipping and greater SR Ca leak, and dantrolene can reverse these changes. However, effects of oxidative stress on RyR2 conformation and leak in myocytes are poorly understood. We used fluorescent CaM, FKBP12.6, and domain-peptide biosensor (F-DPc10) to measure, directly in cardiac myocytes, (1) RyR2 activation by hydrogen peroxide (H2O2)-induced oxidation, (2) RyR2 conformation change caused by oxidation, (3) CaM-RyR2 and FK506-binding protein (FKBP12.6)-RyR2 interaction upon oxidation, and (4) whether dantrolene affects 1-3. H2O2 was used to mimic oxidative stress. H2O2 significantly increased the frequency of Ca(2+) sparks and spontaneous Ca(2+) waves, and dantrolene almost completely blocked these effects. H2O2 pretreatment significantly reduced CaM-RyR2 binding, but had no effect on FKBP12.6-RyR2 binding. Dantrolene restored CaM-RyR2 binding but had no effect on intracellular and RyR2 oxidation levels. H2O2 also accelerated F-DPc10-RyR2 association while dantrolene slowed it. Thus, H2O2 causes conformational changes (sensed by CaM and DPc10 binding) associated with Ca leak, and dantrolene reverses these RyR2 effects. In conclusion, in cardiomyocytes, H2O2 treatment markedly reduces the CaM-RyR2 affinity, has no effect on FKBP12.6-RyR2 affinity, and causes domain unzipping. Dantrolene can correct domain unzipping, restore CaM-RyR2 affinity, and quiet pathological RyR2 channel gating. F-DPc10 and CaM are useful biosensors of a pathophysiological RyR2 state.
Collapse
|
36
|
Walweel K, Laver DR. Mechanisms of SR calcium release in healthy and failing human hearts. Biophys Rev 2015; 7:33-41. [PMID: 28509976 PMCID: PMC5425750 DOI: 10.1007/s12551-014-0152-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 11/25/2014] [Indexed: 01/08/2023] Open
Abstract
Normal heart contraction and rhythm relies on the proper flow of calcium ions (Ca2+) into cardiac cells and between their intracellular organelles, and any disruption can lead to arrhythmia and sudden cardiac death. Electrical excitation of the surface membrane activates voltage-dependent L-type Ca2+ channels to open and allow Ca2+ to enter the cytoplasm. The subsequent increase in cytoplasmic Ca2+ concentration activates calcium release channels (RyR2) located at specialised Ca2+ release sites in the sarcoplasmic reticulum (SR), which serves as an intracellular Ca2+ store. Animal models have provided valuable insights into how intracellular Ca2+ transport mechanisms are altered in human heart failure. The aim of this review is to examine how Ca2+ release sites are remodelled in heart failure and how this affects intracellular Ca2+ transport with an emphasis on Ca2+ release mechanisms in the SR. Current knowledge on how heart failure alters the regulation of RyR2 by Ca2+ and Mg2+ and how these mechanisms control the activity of RyR2 in the confines of the Ca2+ release sites is reviewed.
Collapse
Affiliation(s)
- K Walweel
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW, 2308, Australia
| | - D R Laver
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW, 2308, Australia.
| |
Collapse
|
37
|
Negroni JA, Morotti S, Lascano EC, Gomes AV, Grandi E, Puglisi JL, Bers DM. β-adrenergic effects on cardiac myofilaments and contraction in an integrated rabbit ventricular myocyte model. J Mol Cell Cardiol 2015; 81:162-75. [PMID: 25724724 DOI: 10.1016/j.yjmcc.2015.02.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 01/10/2015] [Accepted: 02/17/2015] [Indexed: 12/21/2022]
Abstract
A five-state model of myofilament contraction was integrated into a well-established rabbit ventricular myocyte model of ion channels, Ca(2+) transporters and kinase signaling to analyze the relative contribution of different phosphorylation targets to the overall mechanical response driven by β-adrenergic stimulation (β-AS). β-AS effect on sarcoplasmic reticulum Ca(2+) handling, Ca(2+), K(+) and Cl(-) currents, and Na(+)/K(+)-ATPase properties was included based on experimental data. The inotropic effect on the myofilaments was represented as reduced myofilament Ca(2+) sensitivity (XBCa) and titin stiffness, and increased cross-bridge (XB) cycling rate (XBcy). Assuming independent roles of XBCa and XBcy, the model reproduced experimental β-AS responses on action potentials and Ca(2+) transient amplitude and kinetics. It also replicated the behavior of force-Ca(2+), release-restretch, length-step, stiffness-frequency and force-velocity relationships, and increased force and shortening in isometric and isotonic twitch contractions. The β-AS effect was then switched off from individual targets to analyze their relative impact on contractility. Preventing β-AS effects on L-type Ca(2+) channels or phospholamban limited Ca(2+) transients and contractile responses in parallel, while blocking phospholemman and K(+) channel (IKs) effects enhanced Ca(2+) and inotropy. Removal of β-AS effects from XBCa enhanced contractile force while decreasing peak Ca(2+) (due to greater Ca(2+) buffering), but had less effect on shortening. Conversely, preventing β-AS effects on XBcy preserved Ca(2+) transient effects, but blunted inotropy (both isometric force and especially shortening). Removal of titin effects had little impact on contraction. Finally, exclusion of β-AS from XBCa and XBcy while preserving effects on other targets resulted in preserved peak isometric force response (with slower kinetics) but nearly abolished enhanced shortening. β-AS effects on XBCa and XBcy have greater impact on isometric and isotonic contraction, respectively.
Collapse
Affiliation(s)
- Jorge A Negroni
- Department of Comparative, Cellular and Molecular Biology, Universidad Favaloro, Buenos Aires, Argentina.
| | - Stefano Morotti
- Department of Pharmacology, University of California Davis, CA, USA
| | - Elena C Lascano
- Department of Comparative, Cellular and Molecular Biology, Universidad Favaloro, Buenos Aires, Argentina
| | - Aldrin V Gomes
- Department of Neurobiology, Physiology and Behavior, University of California Davis, CA, USA
| | - Eleonora Grandi
- Department of Pharmacology, University of California Davis, CA, USA
| | - José L Puglisi
- Department of Pharmacology, University of California Davis, CA, USA
| | - Donald M Bers
- Department of Pharmacology, University of California Davis, CA, USA.
| |
Collapse
|
38
|
Sato D, Bartos DC, Ginsburg KS, Bers DM. Depolarization of cardiac membrane potential synchronizes calcium sparks and waves in tissue. Biophys J 2015; 107:1313-7. [PMID: 25229139 DOI: 10.1016/j.bpj.2014.07.053] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 03/05/2014] [Accepted: 07/15/2014] [Indexed: 10/24/2022] Open
Abstract
The diastolic membrane potential (Vm) can be hyperpolarized or depolarized by various factors such as hyperkalemia or hypokalemia in the long term, or by delayed afterdepolarizations in the short term. In this study, we investigate how Vm affects Ca sparks and waves. We use a physiologically detailed mathematical model to investigate individual factors that affect Ca spark generation and wave propagation. We focus on the voltage range of -90 ∼ -70 mV, which is just below the Vm for sodium channel activation. We find that Vm depolarization promotes Ca wave propagation and hyperpolarization prevents it. This finding is directly validated in voltage clamp experiments with Ca waves using isolated rat ventricular myocytes. Ca transport by the sodium-calcium exchanger (NCX) is determined by Vm as well as Na and Ca concentrations. Depolarized Vm reduces NCX-mediated efflux, elevating [Ca]i, and thus promoting Ca wave propagation. Moreover, depolarized Vm promotes spontaneous Ca releases that can cause initiation of multiple Ca waves. This indicates that during delayed afterdepolarizations, Ca release units (CRUs) interact with not just the immediately adjacent CRUs via Ca diffusion, but also further CRUs via fast (∼0.1 ms) changes in Vm mediated by the voltage and Ca-sensitive NCX. This may contribute significantly to synchronization of Ca waves among multiple cells in tissue.
Collapse
Affiliation(s)
- Daisuke Sato
- Department of Pharmacology, University of California, Davis, Davis, California.
| | - Daniel C Bartos
- Department of Pharmacology, University of California, Davis, Davis, California
| | - Kenneth S Ginsburg
- Department of Pharmacology, University of California, Davis, Davis, California
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, Davis, California.
| |
Collapse
|
39
|
Chan YH, Tsai WC, Song Z, Ko CY, Qu Z, Weiss JN, Lin SF, Chen PS, Jones LR, Chen Z. Acute reversal of phospholamban inhibition facilitates the rhythmic whole-cell propagating calcium waves in isolated ventricular myocytes. J Mol Cell Cardiol 2015; 80:126-35. [PMID: 25596331 DOI: 10.1016/j.yjmcc.2014.12.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 12/08/2014] [Accepted: 12/30/2014] [Indexed: 02/01/2023]
Abstract
Phospholamban (PLB) inhibits the activity of cardiac sarcoplasmic reticulum (SR) Ca(2+)-ATPase (SERCA2a). Phosphorylation of PLB during sympathetic activation reverses SERCA2a inhibition, increasing SR Ca(2+) uptake. However, sympathetic activation also modulates multiple other intracellular targets in ventricular myocytes (VMs), making it impossible to determine the specific effects of the reversal of PLB inhibition on the spontaneous SR Ca(2+) release. Therefore, it remains unclear how PLB regulates rhythmic activity in VMs. Here, we used the Fab fragment of 2D12, a monoclonal anti-PLB antibody, to test how acute reversal of PLB inhibition affects the spontaneous SR Ca(2+) release in normal VMs. Ca(2+) sparks and spontaneous Ca(2+) waves (SCWs) were recorded in the line-scan mode of confocal microscopy using the Ca(2+) fluorescent dye Fluo-4 in isolated permeabilized mouse VMs. Fab, which reverses PLB inhibition, significantly increased the frequency, amplitude, and spatial/temporal spread of Ca(2+) sparks in VMs exposed to 50 nM free [Ca(2+)]. At physiological diastolic free [Ca(2+)] (100-200 nM), Fab facilitated the formation of whole-cell propagating SCWs. At higher free [Ca(2+)], Fab increased the frequency and velocity, but decreased the decay time of the SCWs. cAMP had little additional effect on the frequency or morphology of Ca(2+) sparks or SCWs after Fab addition. These findings were complemented by computer simulations. In conclusion, acute reversal of PLB inhibition alone significantly increased the spontaneous SR Ca(2+) release, leading to the facilitation and organization of whole-cell propagating SCWs in normal VMs. PLB thus plays a key role in subcellular Ca(2+) dynamics and rhythmic activity of VMs.
Collapse
Affiliation(s)
- Yi-Hsin Chan
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA; Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Linkou, Taoyuan, Taiwan
| | - Wei-Chung Tsai
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA; Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung University College of Medicine, Kaohsiung, Taiwan
| | - Zhen Song
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, CA, USA
| | - Christopher Y Ko
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, CA, USA
| | - Zhilin Qu
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, CA, USA
| | - James N Weiss
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, CA, USA
| | - Shien-Fong Lin
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA; Institute of Biomedical Engineering, National Chiao-Tung University, Hsin-Chu, Taiwan
| | - Peng-Sheng Chen
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Larry R Jones
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Zhenhui Chen
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
40
|
Dobrev D, Wehrens XHT. Role of RyR2 phosphorylation in heart failure and arrhythmias: Controversies around ryanodine receptor phosphorylation in cardiac disease. Circ Res 2014; 114:1311-9; discussion 1319. [PMID: 24723656 DOI: 10.1161/circresaha.114.300568] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cardiac ryanodine receptor type 2 plays a key role in excitation-contraction coupling. The ryanodine receptor type 2 channel protein is modulated by various post-translational modifications, including phosphorylation by protein kinase A and Ca(2+)/calmodulin protein kinase II. Despite extensive research in this area, the functional effects of ryanodine receptor type 2 phosphorylation remain disputed. In particular, the potential involvement of increased ryanodine receptor type 2 phosphorylation in the pathogenesis of heart failure and arrhythmias remains a controversial area, which is discussed in this review article.
Collapse
Affiliation(s)
- Dobromir Dobrev
- From the Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany (D.D.); and Cardiovascular Research Institute, Departments of Molecular Physiology and Biophysics, and Medicine-Cardiology, Baylor College of Medicine, Houston, TX (X.H.T.W.)
| | | |
Collapse
|
41
|
Pereira L, Ruiz-Hurtado G, Rueda A, Mercadier JJ, Benitah JP, Gómez AM. Calcium signaling in diabetic cardiomyocytes. Cell Calcium 2014; 56:372-80. [PMID: 25205537 DOI: 10.1016/j.ceca.2014.08.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 07/24/2014] [Accepted: 08/07/2014] [Indexed: 12/18/2022]
Abstract
Diabetes mellitus is one of the most common medical conditions. It is associated to medical complications in numerous organs and tissues, of which the heart is one of the most important and most prevalent organs affected by this disease. In fact, cardiovascular complications are the most common cause of death among diabetic patients. At the end of the 19th century, the weakness of the heart in diabetes was noted as part of the general muscular weakness that exists in that disease. However, it was only in the eighties that diabetic cardiomyopathy was recognized, which comprises structural and functional abnormalities in the myocardium in diabetic patients even in the absence of coronary artery disease or hypertension. This disorder has been associated with both type 1 and type 2 diabetes, and is characterized by early-onset diastolic dysfunction and late-onset systolic dysfunction, in which alteration in Ca(2+) signaling is of major importance, since it controls not only contraction, but also excitability (and therefore is involved in rhythmic disorder), enzymatic activity, and gene transcription. Here we attempt to give a brief overview of Ca(2+) fluxes alteration reported on diabetes, and provide some new data on differential modulation of Ca(2+) handling alteration in males and females type 2 diabetic mice to promote further research. Due to space limitations, we apologize for those authors whose important work is not cited.
Collapse
Affiliation(s)
- Laetitia Pereira
- Department of Pharmacology, University of California Davis, Davis, CA 95616, USA
| | - Gema Ruiz-Hurtado
- Unidad de Hipertensión, Instituto de Investigación i+12, Hospital Universitario 12 de Octubre, Madrid, Spain; Instituto Pluridisciplinar, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Angélica Rueda
- Departamento de Bioquímica, Cinvestav-IPN, México, DF, Mexico
| | - Jean-Jacques Mercadier
- Inserm, UMR S769, Faculté de Pharmacie, Université Paris Sud, Labex LERMIT, DHU TORINO, Châtenay-Malabry, France; Université Paris Diderot - Sorbonne Paris Cité, Assistance Publique - Hôpitaux de Paris (AP-HP), France
| | - Jean-Pierre Benitah
- Inserm, UMR S769, Faculté de Pharmacie, Université Paris Sud, Labex LERMIT, DHU TORINO, Châtenay-Malabry, France
| | - Ana María Gómez
- Inserm, UMR S769, Faculté de Pharmacie, Université Paris Sud, Labex LERMIT, DHU TORINO, Châtenay-Malabry, France.
| |
Collapse
|
42
|
Rueda A, de Alba-Aguayo DR, Valdivia HH. [Ryanodine receptor, calcium leak and arrhythmias]. ARCHIVOS DE CARDIOLOGIA DE MEXICO 2014; 84:191-201. [PMID: 25103920 DOI: 10.1016/j.acmx.2013.12.008] [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: 07/30/2013] [Revised: 10/30/2013] [Accepted: 12/02/2013] [Indexed: 11/28/2022] Open
Abstract
The participation of the ionic Ca(2+) release channel/ryanodine receptor in cardiac excitation-contraction coupling is well known since the late '80s, when various seminal papers communicated its purification for the first time and its identity with the "foot" structures located at the terminal cisternae of the sarcoplasmic reticulum. In addition to its main role as the Ca(2+) channel responsible for the transient Ca(2+) increase that activates the contractile machinery of the cardiomyocytes, the ryanodine receptor releases Ca(2+) during the relaxation phase of the cardiac cycle, giving rise to a diastolic Ca(2+) leak. In normal physiological conditions, diastolic Ca(2+) leak regulates the proper level of luminal Ca(2+), but in pathological conditions it participates in the generation of both, acquired and hereditary arrhythmias. Very recently, several groups have focused their efforts into the development of pharmacological tools to control the altered diastolic Ca(2+) leak via ryanodine receptors. In this review, we focus our interest on describing the participation of cardiac ryanodine receptor in the diastolic Ca(2+) leak under physiological or pathological conditions and also on the therapeutic approaches to control its undesired exacerbated activity during diastole.
Collapse
Affiliation(s)
- Angélica Rueda
- Departamento de Bioquímica, Centro de Investigación y de Estudios Avanzados del IPN, Unidad Zacatenco, México D.F., México.
| | - David R de Alba-Aguayo
- Departamento de Bioquímica, Centro de Investigación y de Estudios Avanzados del IPN, Unidad Zacatenco, México D.F., México
| | - Héctor H Valdivia
- Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI, Estados Unidos
| |
Collapse
|
43
|
Sex differences in SR Ca(2+) release in murine ventricular myocytes are regulated by the cAMP/PKA pathway. J Mol Cell Cardiol 2014; 75:162-73. [PMID: 25066697 DOI: 10.1016/j.yjmcc.2014.07.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 06/26/2014] [Accepted: 07/15/2014] [Indexed: 11/22/2022]
Abstract
Previous studies have shown that ventricular myocytes from female rats have smaller contractions and Ca(2+) transients than males. As cardiac contraction is regulated by the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) pathway, we hypothesized that sex differences in cAMP contribute to differences in Ca(2+) handling. Ca(2+) transients (fura-2) and ionic currents were measured simultaneously (37°C, 2Hz) in ventricular myocytes from adult male and female C57BL/6 mice. Under basal conditions, diastolic Ca(2+), sarcoplasmic reticulum (SR) Ca(2+) stores, and L-type Ca(2+) current did not differ between the sexes. However, female myocytes had smaller Ca(2+) transients (26% smaller), Ca(2+) sparks (6% smaller), and excitation-contraction coupling gain in comparison to males (23% smaller). Interestingly, basal levels of intracellular cAMP were lower in female myocytes (0.7±0.1 vs. 1.7±0.2fmol/μg protein; p<0.001). Importantly, PKA inhibition (2μM H-89) eliminated male-female differences in Ca(2+) transients and gain, as well as Ca(2+) spark amplitude. Western blots showed that PKA inhibition also reduced the ratio of phospho:total RyR2 in male hearts, but not in female hearts. Stimulation of cAMP production with 10μM forskolin abolished sex differences in cAMP levels, as well as differences in Ca(2+) transients, sparks, and gain. To determine if the breakdown of cAMP differed between the sexes, phosphodiesterase (PDE) mRNA levels were measured. PDE3 expression was similar in males and females, but PDE4B expression was higher in female ventricles. The inhibition of cAMP breakdown by PDE4 (10μM rolipram) abolished differences in Ca(2+) transients and gain. These findings suggest that female myocytes have lower levels of basal cAMP due, in part, to higher expression of PDE4B. Lower cAMP levels in females may attenuate PKA phosphorylation of Ca(2+) handling proteins in females, and may limit positive inotropic responses to stimulation of the cAMP/PKA pathway in female hearts.
Collapse
|
44
|
Camors E, Valdivia HH. CaMKII regulation of cardiac ryanodine receptors and inositol triphosphate receptors. Front Pharmacol 2014; 5:101. [PMID: 24847270 PMCID: PMC4021131 DOI: 10.3389/fphar.2014.00101] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 04/17/2014] [Indexed: 01/08/2023] Open
Abstract
Ryanodine receptors (RyRs) and inositol triphosphate receptors (InsP3Rs) are structurally related intracellular calcium release channels that participate in multiple primary or secondary amplified Ca(2+) signals, triggering muscle contraction and oscillatory Ca(2+) waves, or activating transcription factors. In the heart, RyRs play an indisputable role in the process of excitation-contraction coupling as the main pathway for Ca(2+) release from sarcoplasmic reticulum (SR), and a less prominent role in the process of excitation-transcription coupling. Conversely, InsP3Rs are believed to contribute in subtle ways, only, to contraction of the heart, and in more important ways to regulation of transcription factors. Because uncontrolled activity of either RyRs or InsP3Rs may elicit life-threatening arrhythmogenic and/or remodeling Ca(2+) signals, regulation of their activity is of paramount importance for normal cardiac function. Due to their structural similarity, many regulatory factors, accessory proteins, and post-translational processes are equivalent for RyRs and InsP3Rs. Here we discuss regulation of RyRs and InsP3Rs by CaMKII phosphorylation, but touch on other kinases whenever appropriate. CaMKII is emerging as a powerful modulator of RyR and InsP3R activity but interestingly, some of the complexities and controversies surrounding phosphorylation of RyRs also apply to InsP3Rs, and a clear-cut effect of CaMKII on either channel eludes investigators for now. Nevertheless, some effects of CaMKII on global cellular activity, such as SR Ca(2+) leak or force-frequency potentiation, appear clear now, and this constrains the limits of the controversies and permits a more tractable approach to elucidate the effects of phosphorylation at the single channel level.
Collapse
Affiliation(s)
- Emmanuel Camors
- Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor MI, USA
| | - Héctor H Valdivia
- Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor MI, USA
| |
Collapse
|
45
|
Abstract
It has been persuasively shown in the last two decades that the development of heart failure is closely linked to distinct alterations in Ca(2+) cycling. A crucial point in this respect is an increased spontaneous release of Ca(2+) out of the sarcoplasmic reticulum during diastole via ryanodine receptors type 2 (RyR2). The consequence is a compromised sarcoplasmic reticulum Ca(2+) storage capacity, which impairs systolic contractility and possibly diastolic cardiac function due to Ca(2+) overload. Additionally, leaky RyR2 are more and more regarded to potently induce proarrhythmic triggers. Elimination of spontaneously released Ca(2+) via RyR2 in diastole can cause a transient sarcolemmal inward current and hence delayed after depolarisations as substrate for cardiac arrhythmias. In this article, the pathological role and consequences of the SR Ca(2+)-leak and its regulation are reviewed with a main focus on protein kinase A and Ca(2+)-calmodulin-dependent kinase II. We summarise clinical consequences of "leaky RyR2" as well as possible therapeutic strategies in order to correct RyR2 dysfunction and discuss the significance of the available data.
Collapse
|
46
|
Mustroph J, Maier LS, Wagner S. CaMKII regulation of cardiac K channels. Front Pharmacol 2014; 5:20. [PMID: 24600393 PMCID: PMC3930912 DOI: 10.3389/fphar.2014.00020] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 01/31/2014] [Indexed: 11/23/2022] Open
Abstract
Cardiac K channels are critical determinants of cardiac excitability. In hypertrophied and failing myocardium, alterations in the expression and activity of voltage-gated K channels are frequently observed and contribute to the increased propensity for life-threatening arrhythmias. Thus, understanding the mechanisms of disturbed K channel regulation in heart failure (HF) is of critical importance. Amongst others, Ca/calmodulin-dependent protein kinase II (CaMKII) has been identified as an important regulator of K channel activity. In human HF but also various animal models, increased CaMKII expression and activity has been linked to deteriorated contractile function and arrhythmias. This review will discuss the current knowledge about CaMKII regulation of several K channels, its influence on action potential properties, dispersion of repolarization, and arrhythmias with special focus on HF.
Collapse
Affiliation(s)
- Julian Mustroph
- Department of Cardiology, University Medical Center Göttingen Göttingen, Germany
| | - Lars S Maier
- Department of Cardiology, University Medical Center Göttingen Göttingen, Germany
| | - Stefan Wagner
- Department of Cardiology, University Medical Center Göttingen Göttingen, Germany
| |
Collapse
|
47
|
Liu B, Ho HT, Velez-Cortes F, Lou Q, Valdivia CR, Knollmann BC, Valdivia HH, Gyorke S. Genetic ablation of ryanodine receptor 2 phosphorylation at Ser-2808 aggravates Ca(2+)-dependent cardiomyopathy by exacerbating diastolic Ca2+ release. J Physiol 2014; 592:1957-73. [PMID: 24445321 DOI: 10.1113/jphysiol.2013.264689] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Phosphorylation of the cardiac ryanodine receptor (RyR2) by protein kinase A (PKA) at Ser-2808 is suggested to mediate the physiological 'fight or flight' response and contribute to heart failure by rendering the sarcoplasmic reticulum (SR) leaky for Ca(2+). In the present study, we examined the potential role of RyR2 phosphorylation at Ser-2808 in the progression of Ca(2+)-dependent cardiomyopathy (CCM) by using mice genetically modified to feature elevated SR Ca(2+) leak while expressing RyR2s that cannot be phosphorylated at this site (S2808A). Surprisingly, rather than alleviating the disease phenotype, constitutive dephosphorylation of Ser-2808 aggravated CCM as manifested by shortened survival, deteriorated in vivo cardiac function, exacerbated SR Ca(2+) leak and mitochondrial injury. Notably, the deteriorations of cardiac function, myocyte Ca(2+) handling, and mitochondria integrity were consistently worse in mice with heterozygous ablation of Ser-2808 than in mice with complete ablation. Wild-type (WT) and CCM myocytes expressing unmutated RyR2s exhibited a high level of baseline phosphorylation at Ser-2808. Exposure of these CCM cells to protein phosphatase 1 caused a transitory increase in Ca(2+) leak attributable to partial dephosphorylation of RyR2 tetramers at Ser-2808 from more fully phosphorylated state. Thus, exacerbated Ca(2+) leak through partially dephosphorylated RyR2s accounts for the prevalence of the disease phenotype in the heterozygous S2808A CCM mice. These results do not support the importance of RyR2 hyperphosphorylation in Ca(2+)-dependent heart disease, and rather suggest roles for the opposite process, the RyR2 dephosphorylation at this residue in physiological and pathophysiological Ca(2+) signalling.
Collapse
Affiliation(s)
- Bin Liu
- Department of Physiology and Cell Biology, 507 Davis Heart & Lung Research Institute (office), 473 W. 12th Avenue, Columbus, OH 43210, USA.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Merchant FM, Sayadi O, Puppala D, Moazzami K, Heller V, Armoundas AA. A translational approach to probe the proarrhythmic potential of cardiac alternans: a reversible overture to arrhythmogenesis? Am J Physiol Heart Circ Physiol 2013; 306:H465-74. [PMID: 24322612 DOI: 10.1152/ajpheart.00639.2013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Electrocardiographic alternans, a phenomenon of beat-to-beat alternation in cardiac electrical waveforms, has been implicated in the pathogenesis of ventricular arrhythmias and sudden cardiac death (SCD). In the clinical setting, a positive microvolt T-wave alternans test has been associated with a heightened risk of arrhythmic mortality and SCD during medium- and long-term follow-up. However, rather than merely being associated with an increased risk for SCD, several lines of preclinical and clinical evidence suggest that cardiac alternans may play a causative role in generating the acute electrophysiological substrate necessary for the onset of ventricular arrhythmias. Deficiencies in Ca(2+) transport processes have been implicated in the genesis of alternans at the subcellular and cellular level and are hypothesized to contribute to the conditions necessary for dispersion of refractoriness, wave break, reentry, and onset of arrhythmia. As such, detecting acute surges in alternans may provide a mechanism for predicting the impending onset of arrhythmia and opens the door to delivering upstream antiarrhythmic therapies. In this review, we discuss the preclinical and clinical evidence to support a causative association between alternans and acute arrhythmogenesis and outline the potential clinical implications of such an association.
Collapse
Affiliation(s)
- Faisal M Merchant
- Cardiology Division, Emory University School of Medicine, Atlanta, Georgia; and
| | | | | | | | | | | |
Collapse
|
49
|
Guerrero-Hernández A, Ávila G, Rueda A. Ryanodine receptors as leak channels. Eur J Pharmacol 2013; 739:26-38. [PMID: 24291096 DOI: 10.1016/j.ejphar.2013.11.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 11/21/2013] [Indexed: 01/18/2023]
Abstract
Ryanodine receptors are Ca(2+) release channels of internal stores. This review focuses on those situations and conditions that transform RyRs from a finely regulated ion channel to an unregulated Ca(2+) leak channel and the pathological consequences of this alteration. In skeletal muscle, mutations in either CaV1.1 channel or RyR1 results in a leaky behavior of the latter. In heart cells, RyR2 functions normally as a Ca(2+) leak channel during diastole within certain limits, the enhancement of this activity leads to arrhythmogenic situations that are tackled with different pharmacological strategies. In smooth muscle, RyRs are involved more in reducing excitability than in stimulating contraction so the leak activity of RyRs in the form of Ca(2+) sparks, locally activates Ca(2+)-dependent potassium channels to reduce excitability. In neurons the enhanced activity of RyRs is associated with the development of different neurodegenerative disorders such as Alzheimer and Huntington diseases. It appears then that the activity of RyRs as leak channels can have both physiological and pathological consequences depending on the cell type and the metabolic condition.
Collapse
Affiliation(s)
| | | | - Angélica Rueda
- Departamento de Bioquímica, Cinvestav, Mexico city, México
| |
Collapse
|
50
|
Abstract
Synchronized SR calcium (Ca) release is critical to normal cardiac myocyte excitation-contraction coupling, and ideally this release shuts off completely between heartbeats. However, other SR Ca release events are referred to collectively as SR Ca leak (which includes Ca sparks and waves as well as smaller events not detectable as Ca sparks). Much, but not all, of the SR Ca leak occurs via ryanodine receptors and can be exacerbated in pathological states such as heart failure. The extent of SR Ca leak is important because it can (a) reduce SR Ca available for release, causing systolic dysfunction; (b) elevate diastolic [Ca]i, contributing to diastolic dysfunction; (c) cause triggered arrhythmias; and (d) be energetically costly because of extra ATP used to repump Ca. This review addresses quantitative aspects and manifestations of SR Ca leak and its measurement, and how leak is modulated by Ca, associated proteins, and posttranslational modifications in health and disease.
Collapse
Affiliation(s)
- Donald M Bers
- Department of Pharmacology, University of California, Davis, California 95616;
| |
Collapse
|