1
|
Bézard M, Vartanian-Grimaldi JS, Henri J, Calin D, Zaroui A, Kharoubi M, Damy T, Agbulut O, Kordeli E. Amyloidogenic immunoglobulin light chains disturb contractile function and calcium transients in a human cardiac spheroid model of light chain (AL) amyloidosis. Sci Rep 2025; 15:4292. [PMID: 39905088 PMCID: PMC11794598 DOI: 10.1038/s41598-024-82442-3] [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: 07/27/2024] [Accepted: 12/05/2024] [Indexed: 02/06/2025] Open
Abstract
Light chain (AL) amyloidosis is a serious systemic disease caused by the deposition of free misfolded immunoglobulin light chains (LCs) in the form of amyloid fibrils within tissues. Cardiac involvement determines prognosis and mortality. An important cytotoxic impact of amyloidogenic prefibrillar LC oligomers on cardiomyocytes is by now established in isolated rodent cardiomyocytes, simple animal models, or cardiomyocyte-like cell lines. However, the response of human cardiomyocytes to this pathogenic condition is currently unknown. In this work, we have set up a human cellular disease model of AL cardiac amyloidosis (AL-CA) in the form of cardiac spheroids, to study the cytotoxic effects of amyloidogenic LCs with regard to contractile function and calcium handling. To mimic the disease in a reconstituted system, soluble amyloidogenic LCs purified from urine of AL-CA patients were added to a mixture of induced pluripotent stem cell-issued human cardiomyocytes (hiPSC-CM) and human primary cardiac fibroblasts, which resulted in formation of spheroids within 7 days. This procedure ensured a uniform pericellular LC distribution within spheroids. LC-treated hiPSC-CM cultures and LC-containing spheroids presented structural and functional defects including: (1) decreased levels and subcellular disorganization of sarcomeric protein alpha-actinin; (2) abnormal accumulation of calcium handling SERCA2a protein; (3) impaired contractility of spheroids and altered calcium transients. Three independent patient-derived LCs had similar effects, albeit to varying degrees, highlighting the patient-specific properties of this type of amyloids. Taken together, these results indicate that the present cardiac spheroid disease model could be appropriate to the study of cardiac cytotoxicity caused by different amyloidogenic LCs in AL-CA patients, contributing to a better understanding and therapeutic handling of the disease.
Collapse
Affiliation(s)
- Mélanie Bézard
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8263, INSERM U1345, Development, Adaptation and Ageing, Paris, France
- Referral Centre for Cardiac Amyloidosis, GRC Amyloid Research Institute, Réseau amylose Mondor and Cardiology Department, University Hospital Henri Mondor, Assistance Publique des Hôpitaux de Paris, and Clinical Epidemiology and Ageing (CEpiA) at Henri Mondor University Hospital and Institut National de la Santé et de la Recherche Médicale (INSERM) U955, Institut Henri Mondor University Hospital, Créteil, France
| | - Jean-Sébastien Vartanian-Grimaldi
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8263, INSERM U1345, Development, Adaptation and Ageing, Paris, France
| | - Julien Henri
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 7238, Computational and Quantitative Biology, Paris, France
| | - Denisa Calin
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8263, INSERM U1345, Development, Adaptation and Ageing, Paris, France
| | - Amira Zaroui
- Referral Centre for Cardiac Amyloidosis, GRC Amyloid Research Institute, Réseau amylose Mondor and Cardiology Department, University Hospital Henri Mondor, Assistance Publique des Hôpitaux de Paris, and Clinical Epidemiology and Ageing (CEpiA) at Henri Mondor University Hospital and Institut National de la Santé et de la Recherche Médicale (INSERM) U955, Institut Henri Mondor University Hospital, Créteil, France
| | - Mounira Kharoubi
- Referral Centre for Cardiac Amyloidosis, GRC Amyloid Research Institute, Réseau amylose Mondor and Cardiology Department, University Hospital Henri Mondor, Assistance Publique des Hôpitaux de Paris, and Clinical Epidemiology and Ageing (CEpiA) at Henri Mondor University Hospital and Institut National de la Santé et de la Recherche Médicale (INSERM) U955, Institut Henri Mondor University Hospital, Créteil, France
| | - Thibaud Damy
- Referral Centre for Cardiac Amyloidosis, GRC Amyloid Research Institute, Réseau amylose Mondor and Cardiology Department, University Hospital Henri Mondor, Assistance Publique des Hôpitaux de Paris, and Clinical Epidemiology and Ageing (CEpiA) at Henri Mondor University Hospital and Institut National de la Santé et de la Recherche Médicale (INSERM) U955, Institut Henri Mondor University Hospital, Créteil, France
| | - Onnik Agbulut
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8263, INSERM U1345, Development, Adaptation and Ageing, Paris, France.
| | - Ekaterini Kordeli
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8263, INSERM U1345, Development, Adaptation and Ageing, Paris, France.
| |
Collapse
|
2
|
Young AM, Miller JA, Ednie AR, Bennett ES. Cardiomyocyte Reduction of Hybrid/Complex N-Glycosylation in the Adult Causes Heart Failure With Reduced Ejection Fraction in the Absence of Cellular Remodeling. J Am Heart Assoc 2024; 13:e036626. [PMID: 39392134 PMCID: PMC11935583 DOI: 10.1161/jaha.124.036626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Heart failure (HF) presents a massive burden to health care with a complex pathophysiology that results in HF with reduced left ventricle ejection fraction (EF) or HF with preserved EF. It has been shown that relatively modest changes in protein glycosylation, an essential posttranslational modification, are associated with clinical presentations of HF. We and others previously showed that such aberrant protein glycosylation in animal models can lead to HF. METHODS AND RESULTS We develop and characterize a novel, tamoxifen-inducible, cardiomyocyte Mgat1 knockout mouse strain, achieved through deletion of Mgat1, alpha-1,3-mannosyl-glycoproten 2-beta-N-acetlyglucosaminyltransferase, which encodes N-acetylglucosaminyltransferase I. We investigate the role of hybrid/complex N-glycosylation in adult HFrEF pathogenesis at the ion channel, cardiomyocyte, tissue, and gross cardiac level. The data demonstrate successful reduction of N-acetylglucosaminyltransferase I activity and confirm that hybrid/complex N-glycans modulate gating of cardiomyocyte voltage-gated calcium channels. A longitudinal study shows that the tamoxifen-inducible, cardiomyocyte Mgat1 knockout mice present with significantly reduced systolic function by 28 days post induction that progresses into HFrEF by 8 weeks post induction, without significant ventricular dilation or hypertrophy. Further, there was minimal, if any, physiologic or pathophysiologic cardiomyocyte electromechanical remodeling or fibrosis observed before (10-21 days post induction) or after (90-130 days post induction) HFrEF development. CONCLUSIONS The tamoxifen-inducible, cardiomyocyte Mgat1 knockout mouse strain created and characterized here provides a model to describe novel mechanisms and causes responsible for HFrEF onset in the adult, likely occurring primarily through tissue-level reductions in electromechanical activity in the absence of (or at least before) cardiomyocyte remodeling and fibrosis.
Collapse
Affiliation(s)
- Anthony M. Young
- Department of Neuroscience, Cell Biology & PhysiologyBoonshoft School of Medicine and College of Science and Mathematics, Wright State UniversityDaytonOH
| | - John A. Miller
- Department of Neuroscience, Cell Biology & PhysiologyBoonshoft School of Medicine and College of Science and Mathematics, Wright State UniversityDaytonOH
| | - Andrew R. Ednie
- Department of Neuroscience, Cell Biology & PhysiologyBoonshoft School of Medicine and College of Science and Mathematics, Wright State UniversityDaytonOH
| | - Eric S. Bennett
- Department of Neuroscience, Cell Biology & PhysiologyBoonshoft School of Medicine and College of Science and Mathematics, Wright State UniversityDaytonOH
| |
Collapse
|
3
|
Wu XY, Lee YK, Lau YM, Au KW, Tse YL, Ng KM, Wong CK, Tse HF. The Pathogenic Mechanisms of and Novel Therapies for Lamin A/C-Related Dilated Cardiomyopathy Based on Patient-Specific Pluripotent Stem Cell Platforms and Animal Models. Pharmaceuticals (Basel) 2024; 17:1030. [PMID: 39204134 PMCID: PMC11357512 DOI: 10.3390/ph17081030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 09/03/2024] Open
Abstract
Variants (pathogenic) of the LMNA gene are a common cause of familial dilated cardiomyopathy (DCM), which is characterised by early-onset atrioventricular (AV) block, atrial fibrillation and ventricular tachyarrhythmias (VTs), and progressive heart failure. The unstable internal nuclear lamina observed in LMNA-related DCM is a consequence of the disassembly of lamins A and C. This suggests that LMNA variants produce truncated or alternative forms of protein that alter the nuclear structure and the signalling pathway related to cardiac muscle diseases. To date, the pathogenic mechanisms and phenotypes of LMNA-related DCM have been studied using different platforms, such as patient-specific induced pluripotent stem-cell-derived cardiomyocytes (iPSC-CMs) and transgenic mice. In this review, point variants in the LMNA gene that cause autosomal dominantly inherited forms of LMNA-related DCM are summarised. In addition, potential therapeutic targets based on preclinical studies of LMNA variants using transgenic mice and human iPSC-CMs are discussed. They include mitochondria deficiency, variants in nuclear deformation, chromatin remodelling, altered platelet-derived growth factor and ERK1/2-related pathways, and abnormal calcium handling.
Collapse
Affiliation(s)
- Xin-Yi Wu
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
| | - Yee-Ki Lee
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
| | - Yee-Man Lau
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
| | - Ka-Wing Au
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
| | - Yiu-Lam Tse
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
| | - Kwong-Man Ng
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
- Centre for Stem Cell Translational Biology, Hong Kong SAR, China
| | - Chun-Ka Wong
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
- Centre for Stem Cell Translational Biology, Hong Kong SAR, China
- Cardiac and Vascular Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Hong Kong-Guangdong Stem Cell and Regenerative Medicine Research Centre, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Hong Kong SAR, China
- Advanced Biomedical Instrumentation Centre, Hong Kong SAR, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
| |
Collapse
|
4
|
Pásek M, Bébarová M, Šimurdová M, Šimurda J. Functional consequences of changes in the distribution of Ca 2+ extrusion pathways between t-tubular and surface membranes in a model of human ventricular cardiomyocyte. J Mol Cell Cardiol 2024; 193:113-124. [PMID: 38960316 DOI: 10.1016/j.yjmcc.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 06/10/2024] [Accepted: 06/29/2024] [Indexed: 07/05/2024]
Abstract
The sarcolemmal Ca2+ efflux pathways, Na+-Ca2+-exchanger (NCX) and Ca2+-ATPase (PMCA), play a crucial role in the regulation of intracellular Ca2+ load and Ca2+ transient in cardiomyocytes. The distribution of these pathways between the t-tubular and surface membrane of ventricular cardiomyocytes varies between species and is not clear in human. Moreover, several studies suggest that this distribution changes during the development and heart diseases. However, the consequences of NCX and PMCA redistribution in human ventricular cardiomyocytes have not yet been elucidated. In this study, we aimed to address this point by using a mathematical model of the human ventricular myocyte incorporating t-tubules, dyadic spaces, and subsarcolemmal spaces. Effects of various combinations of t-tubular fractions of NCX and PMCA were explored, using values between 0.2 and 1 as reported in animal experiments under normal and pathological conditions. Small variations in the action potential duration (≤ 2%), but significant changes in the peak value of cytosolic Ca2+ transient (up to 17%) were observed at stimulation frequencies corresponding to the human heart rate at rest and during activity. The analysis of model results revealed that the changes in Ca2+ transient induced by redistribution of NCX and PMCA were mainly caused by alterations in Ca2+ concentrations in the subsarcolemmal spaces and cytosol during the diastolic phase of the stimulation cycle. The results suggest that redistribution of both transporters between the t-tubular and surface membranes contributes to changes in contractility in human ventricular cardiomyocytes during their development and heart disease and may promote arrhythmogenesis.
Collapse
Affiliation(s)
- Michal Pásek
- Institute of Thermomechanics, Czech Academy of Sciences, Prague, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| | - Markéta Bébarová
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Department of Internal Medicine and Cardiology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Milena Šimurdová
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jiří Šimurda
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
5
|
Bovo E, Seflova J, Robia SL, Zima AV. Protein carbonylation causes sarcoplasmic reticulum Ca 2+ overload by increasing intracellular Na + level in ventricular myocytes. Pflugers Arch 2024; 476:1077-1086. [PMID: 38769127 DOI: 10.1007/s00424-024-02972-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/22/2024] [Accepted: 05/15/2024] [Indexed: 05/22/2024]
Abstract
Diabetes is commonly associated with an elevated level of reactive carbonyl species due to alteration of glucose and fatty acid metabolism. These metabolic changes cause an abnormality in cardiac Ca2+ regulation that can lead to cardiomyopathies. In this study, we explored how the reactive α-dicarbonyl methylglyoxal (MGO) affects Ca2+ regulation in mouse ventricular myocytes. Analysis of intracellular Ca2+ dynamics revealed that MGO (200 μM) increases action potential (AP)-induced Ca2+ transients and sarcoplasmic reticulum (SR) Ca2+ load, with a limited effect on L-type Ca2+ channel-mediated Ca2+ transients and SERCA-mediated Ca2+ uptake. At the same time, MGO significantly slowed down cytosolic Ca2+ extrusion by Na+/Ca2+ exchanger (NCX). MGO also increased the frequency of Ca2+ waves during rest and these Ca2+ release events were abolished by an external solution with zero [Na+] and [Ca2+]. Adrenergic receptor activation with isoproterenol (10 nM) increased Ca2+ transients and SR Ca2+ load, but it also triggered spontaneous Ca2+ waves in 27% of studied cells. Pretreatment of myocytes with MGO increased the fraction of cells with Ca2+ waves during adrenergic receptor stimulation by 163%. Measurements of intracellular [Na+] revealed that MGO increases cytosolic [Na+] by 57% from the maximal effect produced by the Na+-K+ ATPase inhibitor ouabain (20 μM). This increase in cytosolic [Na+] was a result of activation of a tetrodotoxin-sensitive Na+ influx, but not an inhibition of Na+-K+ ATPase. An increase in cytosolic [Na+] after treating cells with ouabain produced similar effects on Ca2+ regulation as MGO. These results suggest that protein carbonylation can affect cardiac Ca2+ regulation by increasing cytosolic [Na+] via a tetrodotoxin-sensitive pathway. This, in turn, reduces Ca2+ extrusion by NCX, causing SR Ca2+ overload and spontaneous Ca2+ waves.
Collapse
Affiliation(s)
- Elisa Bovo
- Department of Cell & Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, 2160 South First Avenue, Maywood, IL, 60153, USA
| | - Jaroslava Seflova
- Department of Cell & Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, 2160 South First Avenue, Maywood, IL, 60153, USA
| | - Seth L Robia
- Department of Cell & Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, 2160 South First Avenue, Maywood, IL, 60153, USA
| | - Aleksey V Zima
- Department of Cell & Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, 2160 South First Avenue, Maywood, IL, 60153, USA.
| |
Collapse
|
6
|
Kepreotis SV, Oh JG, Park M, Yoo J, Lee C, Mercola M, Hajjar RJ, Jeong D. Inhibition of miR-25 ameliorates cardiac and skeletal muscle dysfunction in aged mdx/utrn haploinsufficient (+/-) mice. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102174. [PMID: 38584818 PMCID: PMC10998245 DOI: 10.1016/j.omtn.2024.102174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 03/14/2024] [Indexed: 04/09/2024]
Abstract
Dystrophic cardiomyopathy is a significant feature of Duchenne muscular dystrophy (DMD). Increased cardiomyocyte cytosolic calcium (Ca2+) and interstitial fibrosis are major pathophysiological hallmarks that ultimately result in cardiac dysfunction. MicroRNA-25 (miR-25) has been identified as a suppressor of both sarcoplasmic reticulum calcium ATPase 2a (SERCA2a) and mothers against decapentaplegic homolog-7 (Smad7) proteins. In this study, we created a gene transfer using an miR-25 tough decoy (TuD) RNA inhibitor delivered via recombinant adeno-associated virus serotype 9 (AAV9) to evaluate the effect of miR-25 inhibition on cardiac and skeletal muscle function in aged dystrophin/utrophin haploinsufficient mice mdx/utrn (+/-), a validated transgenic murine model of DMD. We found that the intravenous delivery of AAV9 miR-25 TuD resulted in strong and stable inhibition of cardiac miR-25 levels, together with the restoration of SERCA2a and Smad7 expression. This was associated with the amelioration of cardiomyocyte interstitial fibrosis as well as recovered cardiac function. Furthermore, the direct quadricep intramuscular injection of AAV9 miR-25 TuD significantly restored skeletal muscle Smad7 expression, reduced tissue fibrosis, and enhanced skeletal muscle performance in mdx/utrn (+/-) mice. These results imply that miR-25 TuD gene transfer may be a novel therapeutic approach to restore cardiomyocyte Ca2+ homeostasis and abrogate tissue fibrosis in DMD.
Collapse
Affiliation(s)
- Sacha V. Kepreotis
- Cardiovascular Research Institute, Icahn School of Medicine, Mount Sinai, NY, USA
| | - Jae Gyun Oh
- Cardiovascular Research Institute, Icahn School of Medicine, Mount Sinai, NY, USA
| | - Mina Park
- Department of Medicinal and Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan, South Korea
| | - Jimeen Yoo
- Cardiovascular Research Institute, Icahn School of Medicine, Mount Sinai, NY, USA
| | - Cholong Lee
- Department of Medicinal and Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan, South Korea
| | - Mark Mercola
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Roger J. Hajjar
- Mass General Brigham Gene and Cell Therapy Institute, Boston, MA, USA
| | - Dongtak Jeong
- Department of Medicinal and Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan, South Korea
- Cardiovascular Research Institute, Icahn School of Medicine, Mount Sinai, NY, USA
| |
Collapse
|
7
|
Bovo E, Seflova J, Robia SL, Zima AV. Protein carbonylation causes sarcoplasmic reticulum Ca2+ overload by increasing intracellular Na+ level in ventricular myocytes. RESEARCH SQUARE 2024:rs.3.rs-3991887. [PMID: 38464201 PMCID: PMC10925417 DOI: 10.21203/rs.3.rs-3991887/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Diabetes is commonly associated with an elevated level of reactive carbonyl species due to alteration of glucose and fatty acid metabolism. These metabolic changes cause an abnormality in cardiac Ca2+ regulation that can lead to cardiomyopathies. In this study, we explored how the reactive α-dicarbonyl methylglyoxal (MGO) affects Ca2+ regulation in mouse ventricular myocytes. Analysis of intracellular Ca2+ dynamics revealed that MGO (200 μM) increases action potential (AP)-induced Ca2+ transients and sarcoplasmic reticulum (SR) Ca2+ load, with a limited effect on L-type Ca2+ channel-mediated Ca2+ transients and SERCA-mediated Ca2+ uptake. At the same time, MGO significantly slowed down cytosolic Ca2+ extrusion by Na+/Ca2+ exchanger (NCX). MGO also increased the frequency of Ca2+ waves during rest and these Ca2+ release events were abolished by an external solution with zero [Na+] and [Ca2+]. Adrenergic receptor activation with isoproterenol (10 nM) increased Ca2+ transients and SR Ca2+ load, but it also triggered spontaneous Ca2+ waves in 27% of studied cells. Pretreatment of myocytes with MGO increased the fraction of cells with Ca2+ waves during adrenergic receptor stimulation by 163%. Measurements of intracellular [Na+] revealed that MGO increases cytosolic [Na+] by 57% from the maximal effect produced by the Na+-K+ ATPase inhibitor ouabain (20 μM). This increase in cytosolic [Na+] was a result of activation of a tetrodotoxin-sensitive Na+ influx, but not an inhibition of Na+-K+ ATPase. An increase in cytosolic [Na+] after treating cells with ouabain produced similar effects on Ca2+ regulation as MGO. These results suggest that protein carbonylation can affect cardiac Ca2+ regulation by increasing cytosolic [Na+] via a tetrodotoxin-sensitive pathway. This, in turn, reduces Ca2+ extrusion by NCX, causing SR Ca2+ overload and spontaneous Ca2+ waves.
Collapse
Affiliation(s)
- Elisa Bovo
- Loyola University Chicago, Stritch School of Medicine
| | | | - Seth L Robia
- Loyola University Chicago, Stritch School of Medicine
| | | |
Collapse
|
8
|
Saad NS, Mashali MA, Repas SJ, Janssen PML. Altering Calcium Sensitivity in Heart Failure: A Crossroads of Disease Etiology and Therapeutic Innovation. Int J Mol Sci 2023; 24:17577. [PMID: 38139404 PMCID: PMC10744146 DOI: 10.3390/ijms242417577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
Heart failure (HF) presents a significant clinical challenge, with current treatments mainly easing symptoms without stopping disease progression. The targeting of calcium (Ca2+) regulation is emerging as a key area for innovative HF treatments that could significantly alter disease outcomes and enhance cardiac function. In this review, we aim to explore the implications of altered Ca2+ sensitivity, a key determinant of cardiac muscle force, in HF, including its roles during systole and diastole and its association with different HF types-HF with preserved and reduced ejection fraction (HFpEF and HFrEF, respectively). We further highlight the role of the two rate constants kon (Ca2+ binding to Troponin C) and koff (its dissociation) to fully comprehend how changes in Ca2+ sensitivity impact heart function. Additionally, we examine how increased Ca2+ sensitivity, while boosting systolic function, also presents diastolic risks, potentially leading to arrhythmias and sudden cardiac death. This suggests that strategies aimed at moderating myofilament Ca2+ sensitivity could revolutionize anti-arrhythmic approaches, reshaping the HF treatment landscape. In conclusion, we emphasize the need for precision in therapeutic approaches targeting Ca2+ sensitivity and call for comprehensive research into the complex interactions between Ca2+ regulation, myofilament sensitivity, and their clinical manifestations in HF.
Collapse
Affiliation(s)
- Nancy S. Saad
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA;
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt
| | - Mohammed A. Mashali
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA;
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
- Department of Surgery, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22514, Egypt
| | - Steven J. Repas
- Department of Emergency Medicine, Wright State University Boonshoft School of Medicine, Dayton, OH 45324, USA;
| | - Paul M. L. Janssen
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA;
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
9
|
Ahmed F, Kahlon T, Mohamed TMA, Ghafghazi S, Settles D. Literature Review: Pathophysiology of Heart Failure with Preserved Ejection Fraction. Curr Probl Cardiol 2023; 48:101745. [PMID: 37087081 DOI: 10.1016/j.cpcardiol.2023.101745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 04/14/2023] [Indexed: 04/24/2023]
Abstract
Heart failure with preserved ejection fraction is a growing public health concern, a disease with poor health outcomes, and is showing increased prevalence globally. This review paper explores the literature with a focus on the pathophysiology and microbiology of preserved ejection fraction heart failure while drawing connections between preserved and reduced ejection fraction states. The discussion teases out the cellular level changes that affect the overall dysfunction of the cardiac tissue, including the clinical manifestations, microbiological changes (endothelial cells, fibroblasts, cardiomyocytes, and excitation-contraction coupling), and the burden of structural diastolic dysfunction. The goal of this review is to summarize the pathophysiological disease state of heart failure with preserved ejection fraction to enhance understanding, knowledge, current treatment models of this pathology.
Collapse
Affiliation(s)
- Faizan Ahmed
- Department of Anesthesiology, University of Louisville School of Medicine, Louisville, Kentucky, USA.
| | - Tani Kahlon
- Department of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Tamer M A Mohamed
- Department of Cardiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Shahab Ghafghazi
- Department of Cardiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Dana Settles
- Department of Cardiothoracic Anesthesia, University of Louisville School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
10
|
Karatsai O, Lehka L, Wojton D, Grabowska AI, Duda MK, Lenartowski R, Redowicz MJ. Unconventional myosin VI in the heart: Involvement in cardiac dysfunction progressing with age. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166748. [PMID: 37169038 DOI: 10.1016/j.bbadis.2023.166748] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/13/2023]
Abstract
Hypertrophic cardiomyopathy is the most common cardiovascular disease, which is characterized by structural and functional myocardial abnormalities. It is caused predominantly by autosomal dominant mutations, mainly in genes encoding cardiac sarcomeric proteins, resulting in diverse phenotypical patterns and a heterogenic clinical course. Unconventional myosin VI (MVI) is one of the proteins important for heart function, as it was shown that a point mutation within MYO6 is associated with left ventricular hypertrophy. Previously, we showed that MVI is expressed in the cardiac muscle, where it localizes to the sarcoplasmic reticulum and intercalated discs. Here, we addressed the mechanisms of its involvement in cardiac dysfunction in Snell's waltzer mice (natural MVI knockouts) during heart development. We showed that heart enlargement was already seen in the E14.5 embryos and newborn animals (P0), and was maintained throughout the examined lifespan (up to 12 months). The higher levels of MVI were observed in the hearts of E14.5 embryos and P0 of control heterozygous mice. A search for the mechanisms behind the observed phenotype revealed several changes, accumulation of which resulted in age-progressing heart dysfunction. The main changes that mostly contribute to this functional impairment are the increase in cardiomyocyte proliferation in newborns, disorganization of intercalated discs, and overexpression of SERCA2 in hearts isolated from 12-month-old mice, indicative of functional alterations of sarcoplasmic reticulum. Also, possible aberrations in the heart vascularization, observed in 12-month-old animals could be additional factors responsible for MVI-associated heart dysfunction.
Collapse
Affiliation(s)
- Olena Karatsai
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland.
| | - Lilya Lehka
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland.
| | - Dominika Wojton
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland.
| | - Anna Izabela Grabowska
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland.
| | - Monika Katarzyna Duda
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, 99/103 Marymoncka St., 01-813 Warsaw, Poland.
| | - Robert Lenartowski
- Faculty of Biological and Veterinary Sciences, The Nicolaus Copernicus University in Torun, 1 Lwowska St., 87-100 Torun, Poland.
| | - Maria Jolanta Redowicz
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland.
| |
Collapse
|
11
|
Hamilton S, Terentyev D. ER stress and calcium-dependent arrhythmias. Front Physiol 2022; 13:1041940. [PMID: 36425292 PMCID: PMC9679650 DOI: 10.3389/fphys.2022.1041940] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022] Open
Abstract
The sarcoplasmic reticulum (SR) plays the key role in cardiac function as the major source of Ca2+ that activates cardiomyocyte contractile machinery. Disturbances in finely-tuned SR Ca2+ release by SR Ca2+ channel ryanodine receptor (RyR2) and SR Ca2+ reuptake by SR Ca2+-ATPase (SERCa2a) not only impair contraction, but also contribute to cardiac arrhythmia trigger and reentry. Besides being the main Ca2+ storage organelle, SR in cardiomyocytes performs all the functions of endoplasmic reticulum (ER) in other cell types including protein synthesis, folding and degradation. In recent years ER stress has become recognized as an important contributing factor in many cardiac pathologies, including deadly ventricular arrhythmias. This brief review will therefore focus on ER stress mechanisms in the heart and how these changes can lead to pro-arrhythmic defects in SR Ca2+ handling machinery.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, United States,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States,*Correspondence: Shanna Hamilton,
| | - Dmitry Terentyev
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, United States,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
12
|
Bhullar S, Shah A, Dhalla N. Mechanisms for the development of heart failure and improvement of cardiac function by angiotensin-converting enzyme inhibitors. SCRIPTA MEDICA 2022. [DOI: 10.5937/scriptamed53-36256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Angiotensin-converting enzyme (ACE) inhibitors, which prevent the conversion of angiotensin I to angiotensin II, are well-known for the treatments of cardiovascular diseases, such as heart failure, hypertension and acute coronary syndrome. Several of these inhibitors including captopril, enalapril, ramipril, zofenopril and imidapril attenuate vasoconstriction, cardiac hypertrophy and adverse cardiac remodeling, improve clinical outcomes in patients with cardiac dysfunction and decrease mortality. Extensive experimental and clinical research over the past 35 years has revealed that the beneficial effects of ACE inhibitors in heart failure are associated with full or partial prevention of adverse cardiac remodeling. Since cardiac function is mainly determined by coordinated activities of different subcellular organelles, including sarcolemma, sarcoplasmic reticulum, mitochondria and myofibrils, for regulating the intracellular concentration of Ca2+ and myocardial metabolism, there is ample evidence to suggest that adverse cardiac remodelling and cardiac dysfunction in the failing heart are the consequence of subcellular defects. In fact, the improvement of cardiac function by different ACE inhibitors has been demonstrated to be related to the attenuation of abnormalities in subcellular organelles for Ca2+-handling, metabolic alterations, signal transduction defects and gene expression changes in failing cardiomyocytes. Various ACE inhibitors have also been shown to delay the progression of heart failure by reducing the formation of angiotensin II, the development of oxidative stress, the level of inflammatory cytokines and the occurrence of subcellular defects. These observations support the view that ACE inhibitors improve cardiac function in the failing heart by multiple mechanisms including the reduction of oxidative stress, myocardial inflammation and Ca2+-handling abnormalities in cardiomyocytes.
Collapse
|
13
|
Rebrova TY, Afanasiev SA, Kondratieva DS, Popov SV. Ontogenetic Features of Changes in the Calcium-Accumulating Ability of the Sarcoplasmic Reticulum of the Myocardium in Rats with Postinfarction Cardiosclerosis. ADVANCES IN GERONTOLOGY 2021. [DOI: 10.1134/s2079057021040111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
14
|
Fernandes ADF, Fernandes GC, Ternes CMP, Cardoso R, Chaparro SV, Goldberger JJ. Sacubitril/valsartan versus angiotensin inhibitors and arrhythmia endpoints in heart failure with reduced ejection fraction. Heart Rhythm O2 2021; 2:724-732. [PMID: 34988523 PMCID: PMC8710618 DOI: 10.1016/j.hroo.2021.09.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Angiotensin receptor–neprilysin inhibitor (ARNI) therapy has been associated with improved survival for patients with symptomatic heart failure and reduced ejection fraction (HFrEF). Objectives We performed a meta-analysis of arrhythmia endpoints from studies comparing ARNI with angiotensin-converting enzyme inhibitors (ACEIs) or angiotensin receptor blockers (ARBs) for patients with HFrEF to assess for incremental benefit. Methods We searched PubMed, Embase, and ClinicalTrials.gov. Baseline study characteristics were collected and outcomes were sustained ventricular arrhythmias, atrial arrhythmias, appropriate implantable cardioverter-defibrillator (ICD) therapy, sudden cardiac death (SCD), and biventricular (BiV) pacing rate. Results We included 9 studies, 4 randomized trials, and 5 observational studies (5589 patients on ARNI vs 5615 on ACEIs/ARBs). Follow-up ranged from 2 to 51 months. The mean age was 65.4 ± 9.8 years, with 77.3% male patients and a mean ejection fraction of 29.0% ± 7.6%. Ischemic cardiomyopathy was present in 62% of patients. In the ARNI group, there were less SCD (odds ratio [OR] 0.78, 95% confidence interval [CI] 0.63–0.96; P = .02), ventricular arrhythmias (OR 0.45, 95% CI 0.25–0.79; P = .005), and appropriate ICD therapy (OR 0.39, 95% CI 0.21–0.74; P = .004). Higher rates of BiV pacing were seen (mean difference 3.13, 95% CI 2.58–3.68; P < .00001) when compared with ACEIs/ARBs. No difference in atrial arrhythmias was seen. Conclusion ARNI therapy provides incremental benefit with respect to ventricular tachyarrhythmias/SCD, which may, in part, explain improved outcomes in patients with HFrEF compared to ACEIs/ARBs. There was increased BiV pacing and decreased ICD therapy in the ARNI group.
Collapse
Affiliation(s)
- Amanda D F Fernandes
- Department of Internal Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Gilson C Fernandes
- Division of Cardiology, University of Miami Miller School of Medicine, Miami, Florida
| | - Caique M P Ternes
- Cardiac Arrhythmia Service, SOS Cardio Hospital, Florianopolis, Brazil
| | - Rhanderson Cardoso
- Division of Cardiology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Sandra V Chaparro
- Miami Cardiac and Vascular Institute, Baptist Health South Florida, Miami, Florida
| | - Jeffrey J Goldberger
- Division of Cardiology, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
15
|
Cardiac ryanodine receptor N-terminal region biosensors identify novel inhibitors via FRET-based high-throughput screening. J Biol Chem 2021; 298:101412. [PMID: 34793835 PMCID: PMC8689225 DOI: 10.1016/j.jbc.2021.101412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 12/15/2022] Open
Abstract
The N-terminal region (NTR) of ryanodine receptor (RyR) channels is critical for the regulation of Ca2+ release during excitation–contraction (EC) coupling in muscle. The NTR hosts numerous mutations linked to skeletal (RyR1) and cardiac (RyR2) myopathies, highlighting its potential as a therapeutic target. Here, we constructed two biosensors by labeling the mouse RyR2 NTR at domains A, B, and C with FRET pairs. Using fluorescence lifetime (FLT) detection of intramolecular FRET signal, we developed high-throughput screening (HTS) assays with these biosensors to identify small-molecule RyR modulators. We then screened a small validation library and identified several hits. Hits with saturable FRET dose–response profiles and previously unreported effects on RyR were further tested using [3H]ryanodine binding to isolated sarcoplasmic reticulum vesicles to determine effects on intact RyR opening in its natural membrane. We identified three novel inhibitors of both RyR1 and RyR2 and two RyR1-selective inhibitors effective at nanomolar Ca2+. Two of these hits activated RyR1 only at micromolar Ca2+, highlighting them as potential enhancers of excitation–contraction coupling. To determine whether such hits can inhibit RyR leak in muscle, we further focused on one, an FDA-approved natural antibiotic, fusidic acid (FA). In skinned skeletal myofibers and permeabilized cardiomyocytes, FA inhibited RyR leak with no detrimental effect on skeletal myofiber excitation–contraction coupling. However, in intact cardiomyocytes, FA induced arrhythmogenic Ca2+ transients, a cautionary observation for a compound with an otherwise solid safety record. These results indicate that HTS campaigns using the NTR biosensor can identify compounds with therapeutic potential.
Collapse
|
16
|
Distributed synthesis of sarcolemmal and sarcoplasmic reticulum membrane proteins in cardiac myocytes. Basic Res Cardiol 2021; 116:63. [PMID: 34713358 PMCID: PMC8553722 DOI: 10.1007/s00395-021-00895-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/30/2021] [Accepted: 09/11/2021] [Indexed: 02/05/2023]
Abstract
It is widely assumed that synthesis of membrane proteins, particularly in the heart, follows the classical secretory pathway with mRNA translation occurring in perinuclear regions followed by protein trafficking to sites of deployment. However, this view is based on studies conducted in less-specialized cells, and has not been experimentally addressed in cardiac myocytes. Therefore, we undertook direct experimental investigation of protein synthesis in cardiac tissue and isolated myocytes using single-molecule visualization techniques and a novel proximity-ligated in situ hybridization approach for visualizing ribosome-associated mRNA molecules for a specific protein species, indicative of translation sites. We identify here, for the first time, that the molecular machinery for membrane protein synthesis occurs throughout the cardiac myocyte, and enables distributed synthesis of membrane proteins within sub-cellular niches where the synthesized protein functions using local mRNA pools trafficked, in part, by microtubules. We also observed cell-wide distribution of membrane protein mRNA in myocardial tissue from both non-failing and hypertrophied (failing) human hearts, demonstrating an evolutionarily conserved distributed mechanism from mouse to human. Our results identify previously unanticipated aspects of local control of cardiac myocyte biology and highlight local protein synthesis in cardiac myocytes as an important potential determinant of the heart’s biology in health and disease.
Collapse
|
17
|
Lookin O, Kuznetsov D, Protsenko Y. Omecamtiv mecarbil attenuates length-tension relationship in healthy rat myocardium and preserves it in monocrotaline-induced pulmonary heart failure. Clin Exp Pharmacol Physiol 2021; 49:84-93. [PMID: 34459025 DOI: 10.1111/1440-1681.13584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 01/10/2023]
Abstract
The cardiac-specific myosin activator, omecamtiv mecarbil (OM), is an effective inotrope for treating heart failure but its effects on active force and Ca2+ kinetics in healthy and diseased myocardium remain poorly studied. We tested the effect of two concentrations of OM (0.2 and 1 µmol/L in saline) on isometric contraction and Ca-transient (CaT) in right ventricular trabeculae of healthy rats (CONT, n = 8) and rats with monocrotaline-induced pulmonary heart failure (MCT, n = 8). The contractions were obtained under preload of 75%-100% of optimal length (tension-length relationship). The 0.2 µmol/L OM did not affect the diastolic level, amplitude, or kinetics of isometric contraction and CaT, irrespective of the group of rats or preload. The 1 µmol/L OM significantly suppressed active tension-length relationships in CONT but not in MCT, while leading in both groups to a significantly prolonged relaxation. CaT time-to-peak was unaffected in CONT and MCT, but CaT decay was slightly accelerated in its early phase and considerably prolonged in its late phase to a similar extent in both groups. We conclude that the substantial prolongation of CaT decay is due to enhanced Ca2+ utilisation by troponin C mediated by the direct effect of OM on the cooperative activation of myofilaments. The lack of beneficial effect of OM in the healthy rat myocardium may be due to a relatively high level of activating Ca2+ in cells with normal Ca2+ handling, whereas the preservation of the tension-length relationship in the failing heart may relate to the diminished Ca2+ levels of sarcoplasmic reticulum.
Collapse
Affiliation(s)
- Oleg Lookin
- Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, Yekaterinburg, Russian Federation
| | - Daniil Kuznetsov
- Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, Yekaterinburg, Russian Federation
| | - Yuri Protsenko
- Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, Yekaterinburg, Russian Federation
| |
Collapse
|
18
|
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
|
19
|
Li J, Richmond B, Hong T. Cardiac T-Tubule cBIN1-Microdomain, a Diagnostic Marker and Therapeutic Target of Heart Failure. Int J Mol Sci 2021; 22:ijms22052299. [PMID: 33669042 PMCID: PMC7956774 DOI: 10.3390/ijms22052299] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 12/23/2022] Open
Abstract
Since its first identification as a cardiac transverse tubule (t-tubule) protein, followed by the cloning of the cardiac isoform responsible for t-tubule membrane microdomain formation, cardiac bridging integrator 1 (cBIN1) and its organized microdomains have emerged as a key mechanism in maintaining normal beat-to-beat heart contraction and relaxation. The abnormal remodeling of cBIN1-microdomains occurs in stressed and diseased cardiomyocytes, contributing to the pathophysiology of heart failure. Due to the homeostatic turnover of t-tubule cBIN1-microdomains via microvesicle release into the peripheral circulation, plasma cBIN1 can be assayed as a liquid biopsy of cardiomyocyte health. A new blood test cBIN1 score (CS) has been developed as a dimensionless inverse index derived from plasma cBIN1 concentration with a diagnostic and prognostic power for clinical outcomes in stable ambulatory patients with heart failure with reduced or preserved ejection fraction (HFrEF or HFpEF). Recent evidence further indicates that exogenous cBIN1 introduced by adeno-associated virus 9-based gene therapy can rescue cardiac contraction and relaxation in failing hearts. The therapeutic potential of cBIN1 gene therapy is enormous given its ability to rescue cardiac inotropy and provide lusitropic protection in the meantime. These unprecedented capabilities of cBIN1 gene therapy are shifting the current paradigm of therapy development for heart failure, particularly HFpEF.
Collapse
Affiliation(s)
- Jing Li
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA; (J.L.); (B.R.)
| | - Bradley Richmond
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA; (J.L.); (B.R.)
| | - TingTing Hong
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA; (J.L.); (B.R.)
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT 84112, USA
- Correspondence: ; Tel.: +1-801-581-3090
| |
Collapse
|
20
|
Stress-driven cardiac calcium mishandling via a kinase-to-kinase crosstalk. Pflugers Arch 2021; 473:363-375. [PMID: 33590296 PMCID: PMC7940337 DOI: 10.1007/s00424-021-02533-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/19/2021] [Accepted: 02/02/2021] [Indexed: 01/25/2023]
Abstract
Calcium homeostasis in the cardiomyocyte is critical to the regulation of normal cardiac function. Abnormal calcium dynamics such as altered uptake by the sarcoplasmic reticulum (SR) Ca2+-ATPase and increased diastolic SR calcium leak are involved in the development of maladaptive cardiac remodeling under pathological conditions. Ca2+/calmodulin-dependent protein kinase II-δ (CaMKIIδ) is a well-recognized key molecule in calcium dysregulation in cardiomyocytes. Elevated cellular stress is known as a common feature during pathological remodeling, and c-jun N-terminal kinase (JNK) is an important stress kinase that is activated in response to intrinsic and extrinsic stress stimuli. Our lab recently identified specific actions of JNK isoform 2 (JNK2) in CaMKIIδ expression, activation, and CaMKIIδ-dependent SR Ca2+ mishandling in the stressed heart. This review focuses on the current understanding of cardiac SR calcium handling under physiological and pathological conditions as well as the newly identified contribution of the stress kinase JNK2 in CaMKIIδ-dependent SR Ca2+ abnormal mishandling. The new findings identifying dual roles of JNK2 in CaMKIIδ expression and activation are also discussed in this review.
Collapse
|
21
|
Hamilton S, Veress R, Belevych A, Terentyev D. The role of calcium homeostasis remodeling in inherited cardiac arrhythmia syndromes. Pflugers Arch 2021; 473:377-387. [PMID: 33404893 PMCID: PMC7940310 DOI: 10.1007/s00424-020-02505-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023]
Abstract
Sudden cardiac death due to malignant ventricular arrhythmias remains the major cause of mortality in the postindustrial world. Defective intracellular Ca2+ homeostasis has been well established as a key contributing factor to the enhanced propensity for arrhythmia in acquired cardiac disease, such as heart failure or diabetic cardiomyopathy. More recent advances provide a strong basis to the emerging view that hereditary cardiac arrhythmia syndromes are accompanied by maladaptive remodeling of Ca2+ homeostasis which substantially increases arrhythmic risk. This brief review will focus on functional changes in elements of Ca2+ handling machinery in cardiomyocytes that occur secondary to genetic mutations associated with catecholaminergic polymorphic ventricular tachycardia, and long QT syndrome.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Roland Veress
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Andriy Belevych
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Dmitry Terentyev
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
22
|
Celestino-Montes A, Pérez-Treviño P, Sandoval-Herrera MD, Gómez-Víquez NL, Altamirano J. Relative role of T-tubules disruption and decreased SERCA2 on contractile dynamics of isolated rat ventricular myocytes. Life Sci 2021; 264:118700. [PMID: 33130073 DOI: 10.1016/j.lfs.2020.118700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/25/2020] [Accepted: 10/28/2020] [Indexed: 10/23/2022]
Abstract
AIMS Ventricular myocytes (VM) depolarization activates L-type Ca2+ channels (LCC) allowing Ca2+ influx (ICa) to synchronize sarcoplasmic reticulum (SR) Ca2+ release, via Ca2+-release channels (RyR2). The resulting whole-cell Ca2+ transient triggers contraction, while cytosolic Ca2+ removal by SR Ca2+ pump (SERCA2) and sarcolemmal Na+/Ca2+ exchanger (NCX) allows relaxation. In diseased hearts, extensive VM remodeling causes heterogeneous, blunted and slow Ca2+ transients. Among remodeling changes are: A) T-tubules disorganization. B) Diminished SERCA2 and low SR Ca2+. However, those often overlap, hindering their relative contribution to contractile dysfunction (CD). Furthermore, few studies have assessed their specific impact on the spatiotemporal Ca2+ transient properties and contractile dynamics simultaneously. Therefore, we sought to perform a quantitative comparison of how heterogeneous and slow Ca2+ transients, with different underlying determinants, affect contractile performance. METHODS We used two experimental models: A) formamide-induced acute "detubulation", where VM retain functional RyR2 and SERCA2, but lack T-tubules-associated LCC and NCX. B) Intact VM from hypothyroid rats, presenting decreased SERCA2 and SR Ca2+, but maintained T-tubules. By confocal imaging of Fluo-4-loaded VM, under field-stimulation, simultaneously acquired Ca2+ transients and shortening, allowing direct correlations. KEY FINDINGS We found near-linear correlations among key parameters of altered Ca2+ transients, caused independently by T-tubules disruption or decreased SR Ca2+, and shortening and relaxation, SIGNIFICANCE: Unrelated structural and molecular alterations converge in similarly abnormal Ca2+ transients and CD, highlighting the importance of independently reproduce disease-specific alterations, to quantitatively assess their impact on Ca2+ signaling and contractility, which would be valuable to determine potential disease-specific therapeutic targets.
Collapse
Affiliation(s)
- Antonio Celestino-Montes
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Av. Morones Prieto No. 3000 Pte., Monterrey, N.L. 64710, Mexico
| | - Perla Pérez-Treviño
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Av. Morones Prieto No. 3000 Pte., Monterrey, N.L. 64710, Mexico
| | - Maya D Sandoval-Herrera
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Av. Morones Prieto No. 3000 Pte., Monterrey, N.L. 64710, Mexico
| | - Norma L Gómez-Víquez
- Departamento de Farmacobiologia, CINVESTAV-IPN sede Sur, Mexico, D.F. 14330, Mexico
| | - Julio Altamirano
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Av. Morones Prieto No. 3000 Pte., Monterrey, N.L. 64710, Mexico.
| |
Collapse
|
23
|
Kilfoil PJ, Lotteau S, Zhang R, Yue X, Aynaszyan S, Solymani RE, Cingolani E, Marbán E, Goldhaber JI. Distinct features of calcium handling and β-adrenergic sensitivity in heart failure with preserved versus reduced ejection fraction. J Physiol 2020; 598:5091-5108. [PMID: 32829489 PMCID: PMC7693093 DOI: 10.1113/jp280425] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 08/19/2020] [Indexed: 12/21/2022] Open
Abstract
Key points Heart failure (HF), the leading cause of death in developed countries, occurs in the setting of reduced (HFrEF) or preserved (HFpEF) ejection fraction. Unlike HFrEF, there are no effective treatments for HFpEF, which accounts for ∼50% of heart failure. Abnormal intracellular calcium dynamics in cardiomyocytes have major implications for contractility and rhythm, but compared to HFrEF, very little is known about calcium cycling in HFpEF. We used rat models of HFpEF and HFrEF to reveal distinct differences in intracellular calcium regulation and excitation‐contraction (EC) coupling. While HFrEF is characterized by defective EC coupling at baseline, HFpEF exhibits enhanced coupling fidelity, further aggravated by a reduction in β‐adrenergic sensitivity. These differences in EC coupling and β‐adrenergic sensitivity may help explain why therapies that work in HFrEF are ineffective in HFpEF.
Abstract Heart failure with reduced or preserved ejection fraction (respectively, HFrEF and HFpEF) is the leading cause of death in developed countries. Although numerous therapies improve outcomes in HFrEF, there are no effective treatments for HFpEF. We studied phenotypically verified rat models of HFrEF and HFpEF to compare excitation‐contraction (EC) coupling and protein expression in these two forms of heart failure. Dahl salt‐sensitive rats were fed a high‐salt diet (8% NaCl) from 7 weeks of age to induce HFpEF. Impaired diastolic relaxation and preserved ejection fraction were confirmed in each animal echocardiographically, and clinical signs of heart failure were documented. To generate HFrEF, Sprague‐Dawley (SD) rats underwent permanent left anterior descending coronary artery ligation which, 8–10 weeks later, led to systolic dysfunction (verified echocardiographically) and clinical signs of heart failure. Calcium (Ca2+) transients were measured in isolated cardiomyocytes under field stimulation or patch clamp. Ultra‐high‐speed laser scanning confocal imaging captured Ca2+ sparks evoked by voltage steps. Western blotting and PCR were used to assay changes in EC coupling protein and RNA expression. Cardiomyocytes from rats with HFrEF exhibited impaired EC coupling, including decreased Ca2+ transient (CaT) amplitude and defective couplon recruitment, associated with transverse (t)‐tubule disruption. In stark contrast, HFpEF cardiomyocytes showed saturated EC coupling (increased ICa, high probability of couplon recruitment with greater Ca2+ release synchrony, increased CaT) and preserved t‐tubule integrity. β‐Adrenergic stimulation of HFpEF myocytes with isoprenaline (isoproterenol) failed to elicit robust increases in ICa or CaT and relaxation kinetics. Fundamental differences in EC coupling distinguish HFrEF from HFpEF. Heart failure (HF), the leading cause of death in developed countries, occurs in the setting of reduced (HFrEF) or preserved (HFpEF) ejection fraction. Unlike HFrEF, there are no effective treatments for HFpEF, which accounts for ∼50% of heart failure. Abnormal intracellular calcium dynamics in cardiomyocytes have major implications for contractility and rhythm, but compared to HFrEF, very little is known about calcium cycling in HFpEF. We used rat models of HFpEF and HFrEF to reveal distinct differences in intracellular calcium regulation and excitation‐contraction (EC) coupling. While HFrEF is characterized by defective EC coupling at baseline, HFpEF exhibits enhanced coupling fidelity, further aggravated by a reduction in β‐adrenergic sensitivity. These differences in EC coupling and β‐adrenergic sensitivity may help explain why therapies that work in HFrEF are ineffective in HFpEF.
Collapse
Affiliation(s)
- Peter J Kilfoil
- Cedars-Sinai Medical Center, Smidt Heart Institute, Los Angeles, CA, USA
| | - Sabine Lotteau
- Cedars-Sinai Medical Center, Smidt Heart Institute, Los Angeles, CA, USA
| | - Rui Zhang
- Cedars-Sinai Medical Center, Smidt Heart Institute, Los Angeles, CA, USA
| | - Xin Yue
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Stephan Aynaszyan
- Cedars-Sinai Medical Center, Smidt Heart Institute, Los Angeles, CA, USA
| | - Ryan E Solymani
- Cedars-Sinai Medical Center, Smidt Heart Institute, Los Angeles, CA, USA
| | - Eugenio Cingolani
- Cedars-Sinai Medical Center, Smidt Heart Institute, Los Angeles, CA, USA
| | - Eduardo Marbán
- Cedars-Sinai Medical Center, Smidt Heart Institute, Los Angeles, CA, USA
| | - Joshua I Goldhaber
- Cedars-Sinai Medical Center, Smidt Heart Institute, Los Angeles, CA, USA
| |
Collapse
|
24
|
Liu CM, Lin FZ, Chen YC, Lin YK, Lu YY, Wu CI, Higa S, Chen SA, Chen YJ. Concurrent increases in post-pacing action potential duration and contractility predict occurrence of ventricular arrhythmia. Pflugers Arch 2020; 472:1783-1791. [PMID: 32794054 DOI: 10.1007/s00424-020-02445-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/29/2020] [Accepted: 07/31/2020] [Indexed: 12/14/2022]
Abstract
Excitation-contraction coupling from the integration of action potential duration (APD) and muscle contractility plays an important role in arrhythmogenesis. We aimed to determine whether distinctive excitation-contraction coupling contributes to the genesis of ventricular tachycardias (VTs). Action potential (AP) and mechanical activity were simultaneously recorded under electrical pacing (cycle lengths from 1000 to 100 ms) in the tissue model created from isolated rabbit right ventricular outflow tracts treated with NS 5806 (10 μM, transient outward potassium current enhancer), pinacidil (2 μM, ATP-sensitive potassium channel opener), and pilsicainide (5 μM, sodium channel blocker). There were 15 (9.9%) inducible VT episodes (group 1) and 136 (90.1%) non-inducible VT episodes (group 2) in our tissue model. Group 1 had greater post-pacing increases of the first occurrence of AP at 90% repolarization (ΔAPD90, p < 0.001) and contractility (ΔContractility, p = 0.003) compared with group 2. Triggered VT episodes were common (72.7%) in cases with a ΔAPD90 > 15% and a ΔContractility > 270%, but were undetectable in those with a ΔAPD90 < 15% and a ΔContractility < 270%. In those with pacing-induced VTs, KB-R7943 (10 μM, a Na+-Ca2+ exchanger inhibitor, NCX inhibitor) significantly reduced the occurrence of VTs from 100.0 to 20.0% (15/15 to 3/15 episodes, p < 0.001). Concurrent increases in both post-pacing APD and contractility resulted in the occurrence of ventricular arrhythmias. NCX inhibition may be a potential therapeutic strategy for ventricular arrhythmias.
Collapse
Affiliation(s)
- Chih-Min Liu
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Feng-Zhi Lin
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Yung-Kuo Lin
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, 111 Hsin-Lung Road, Sec. 3, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yen-Yu Lu
- Division of Cardiology, Sijhih Cathay General Hospital, New Taipei City, Taiwan
| | - Cheng-I Wu
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Satoshi Higa
- Cardiac Electrophysiology and Pacing Laboratory, Division of Cardiovascular Medicine, Makiminato Central Hospital, Okinawa, Japan
| | - Shih-Ann Chen
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Jen Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, 111 Hsin-Lung Road, Sec. 3, Taipei, Taiwan.
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- Cardiovacular Research Center, Wan Fang Hospital, Taipei Medical University , Taipei, Taiwan.
| |
Collapse
|
25
|
Sarcoplasmic reticulum calcium mishandling: central tenet in heart failure? Biophys Rev 2020; 12:865-878. [PMID: 32696300 DOI: 10.1007/s12551-020-00736-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 07/08/2020] [Indexed: 12/17/2022] Open
Abstract
Excitation-contraction coupling links excitation of the sarcolemmal surface membrane to mechanical contraction. In the heart this link is established via a Ca2+-induced Ca2+ release process, which, following sarcolemmal depolarisation, prompts Ca2+ release from the sarcoplasmic reticulum (SR) though the ryanodine receptor (RyR2). This substantially raises the cytoplasmic Ca2+ concentration to trigger systole. In diastole, Ca2+ is removed from the cytoplasm, primarily via the sarcoplasmic-endoplasmic reticulum Ca2+-dependent ATPase (SERCA) pump on the SR membrane, returning Ca2+ to the SR store. Ca2+ movement across the SR is thus fundamental to the systole/diastole cycle and plays an essential role in maintaining cardiac contractile function. Altered SR Ca2+ homeostasis (due to disrupted Ca2+ release, storage, and reuptake pathways) is a central tenet of heart failure and contributes to depressed contractility, impaired relaxation, and propensity to arrhythmia. This review will focus on the molecular mechanisms that underlie asynchronous Ca2+ cycling around the SR in the failing heart. Further, this review will illustrate that the combined effects of expression changes and disruptions to RyR2 and SERCA2a regulatory pathways are critical to the pathogenesis of heart failure.
Collapse
|
26
|
Spinozzi S, Liu C, Chen Z, Feng W, Zhang L, Ouyang K, Evans SM, Chen J. Nexilin Is Necessary for Maintaining the Transverse-Axial Tubular System in Adult Cardiomyocytes. Circ Heart Fail 2020; 13:e006935. [PMID: 32635769 PMCID: PMC7583668 DOI: 10.1161/circheartfailure.120.006935] [Citation(s) in RCA: 15] [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: 01/24/2020] [Accepted: 03/31/2020] [Indexed: 01/28/2023]
Abstract
BACKGROUND NEXN (nexilin) is a protein of the junctional membrane complex required for development of cardiac T-tubules. Global and cardiomyocyte-specific loss of Nexn in mice leads to a rapidly progressive dilated cardiomyopathy and premature death. Therefore, little is known as to the role of NEXN in adult cardiomyocytes. Transverse-axial tubular system remodeling are well-known features in heart failure. Although NEXN is required during development for T-tubule formation, its role, if any, in mature T-tubules remains to be addressed. METHODS Nexn inducible adult cardiomyocyte-specific KO mice were generated. Comprehensive morphological and functional analyses were performed. Heart samples (n>3) were analyzed by molecular, biochemical, and electron microscopy analyses. Isolated single adult cardiomyocytes were analyzed by confocal microscopy, and myocyte shortening/re-lengthening and Ca2+ transient studies were conducted. RESULTS Inducible cardiomyocyte-specific loss of Nexn in adult mice resulted in a dilated cardiomyopathy with reduced cardiac function (13% reduction in percentage fractional shortening; P<0.05). In vivo and in vitro analyses of adult mouse heart samples revealed that NEXN was essential for optimal contraction and calcium handling and was required for maintenance of T-tubule network organization (transverse tubular component in Nexn inducible adult cardiomyocyte-specific KO mice reduced by 40% with respect to controls, P<0.05). CONCLUSIONS Results here reported reveal NEXN to be a pivotal component of adult junctional membrane complexes required for maintenance of transverse-axial tubular architecture. These results demonstrate that NEXN plays an essential role in the adult cardiomyocyte and give further understanding of pathological mechanisms responsible for cardiomyopathy in patients carrying mutations in the NEXN gene.
Collapse
MESH Headings
- Age Factors
- Animals
- Calcium/metabolism
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/physiopathology
- Disease Models, Animal
- Mice
- Mice, Knockout
- Microfilament Proteins/genetics
- Microfilament Proteins/metabolism
- Microfilament Proteins/physiology
- Microtubules/metabolism
- Microtubules/physiology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/physiology
- Ventricular Dysfunction, Left/genetics
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/physiopathology
Collapse
Affiliation(s)
- Simone Spinozzi
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Canzhao Liu
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Ze’e Chen
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Wei Feng
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Lunfeng Zhang
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Kunfu Ouyang
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Sylvia M. Evans
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Ju Chen
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
27
|
Alford RF, Smolin N, Young HS, Gray JJ, Robia SL. Protein docking and steered molecular dynamics suggest alternative phospholamban-binding sites on the SERCA calcium transporter. J Biol Chem 2020; 295:11262-11274. [PMID: 32554805 DOI: 10.1074/jbc.ra120.012948] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 06/16/2020] [Indexed: 01/27/2023] Open
Abstract
The transport activity of the sarco(endo)plasmic reticulum calcium ATPase (SERCA) in cardiac myocytes is modulated by an inhibitory interaction with a transmembrane peptide, phospholamban (PLB). Previous biochemical studies have revealed that PLB interacts with a specific inhibitory site on SERCA, and low-resolution structural evidence suggests that PLB interacts with distinct alternative sites on SERCA. High-resolution details of the structural determinants of SERCA regulation have been elusive because of the dynamic nature of the regulatory complex. In this study, we used computational approaches to develop a structural model of SERCA-PLB interactions to gain a mechanistic understanding of PLB-mediated SERCA transport regulation. We combined steered molecular dynamics and membrane protein-protein docking experiments to achieve both a global search and all-atom force calculations to determine the relative affinities of PLB for candidate sites on SERCA. We modeled the binding of PLB to several SERCA conformations, representing different enzymatic states sampled during the calcium transport catalytic cycle. The results of the steered molecular dynamics and docking experiments indicated that the canonical PLB-binding site (comprising transmembrane helices M2, M4, and M9) is the preferred site. This preference was even more stringent for a superinhibitory PLB variant. Interestingly, PLB-binding specificity became more ambivalent for other SERCA conformers. These results provide evidence for polymorphic PLB interactions with novel sites on M3 and with the outside of the SERCA helix M9. Our findings are compatible with previous physical measurements that suggest that PLB interacts with multiple binding sites, conferring dynamic responsiveness to changing physiological conditions.
Collapse
Affiliation(s)
- Rebecca F Alford
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Nikolai Smolin
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Cardiovascular Research Institute, Loyola University Chicago, Maywood, Illinois, USA
| | - Howard S Young
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Jeffrey J Gray
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Seth L Robia
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Cardiovascular Research Institute, Loyola University Chicago, Maywood, Illinois, USA
| |
Collapse
|
28
|
Lookin O, Butova X, Protsenko Y. The role of pacing rate in the modulation of mechano-induced immediate and delayed changes in the force and Ca-transient of cardiac muscle. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2020; 159:34-45. [PMID: 32450183 DOI: 10.1016/j.pbiomolbio.2020.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 03/11/2020] [Accepted: 05/11/2020] [Indexed: 12/21/2022]
Abstract
Myocardial function is tuned by dynamic changes in length and load via mechano-calcium feedback. This regulation may be significantly affected by heart rhythm. We evaluated the mechano-induced modulation of contractility and Ca-transient (CaT) in the rat myocardium subjected to twitch-by-twitch shortening-re-lengthening (↓-↑) trains of different lengths (N = 1 … 720 cycles) at low (1 Hz) and near-physiological (3.5 Hz) pacing rates. Force/CaT characteristics were evaluated in the first post-train isometric twitch (immediate effect) and during slow changes (delayed maximal elevation/decrease) and compared with those of the pre-train twitch. The immediate inotropic effect was positive for N = 30 … 720 and negative for N = 1 … 20, while the delayed effect was always positive. The immediate and delayed inotropic effects were significantly higher at 3.5-Hz vs 1-Hz (P < 0.05). The prominent inotropism was accompanied by much smaller changes in the CaT diastolic level/amplitude. The shortening-re-lengthening train induced oscillations of the slow change in force at 3.5-Hz (always) and at 1-Hz (∼50% of muscles), which were dependent of the train length and independent of the pacing rate. We suggest that twitch-by-twitch shortening-re-lengthening of cardiac muscle decreases Ca2+ buffering by troponin C and elevates Ca2+ loading of the sarcoplasmic reticulum (SR); the latter cumulatively depends on the train length. A high pacing rate intensifies the cumulative transient shift in the SR Ca2+ loading, augmenting the post-train inotropic response and prolonging its recovery to the pre-train level. The pacing-dependent mechano-induced inotropic effects remain to be elucidated in the myocardium with impaired Ca handling.
Collapse
Affiliation(s)
- Oleg Lookin
- Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, 620049, 106 Pervomayskaya St., Yekaterinburg, Russia; Center for Fundamental Biotechnology and Bioengineering, Institute of Natural Sciences and Mathematics, Ural Federal University, 620002, 19 Mira St., Yekaterinburg, Russia.
| | - Xenia Butova
- Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, 620049, 106 Pervomayskaya St., Yekaterinburg, Russia; Center for Fundamental Biotechnology and Bioengineering, Institute of Natural Sciences and Mathematics, Ural Federal University, 620002, 19 Mira St., Yekaterinburg, Russia
| | - Yuri Protsenko
- Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, 620049, 106 Pervomayskaya St., Yekaterinburg, Russia
| |
Collapse
|
29
|
Morales Rodriguez B, Domínguez-Rodríguez A, Benitah JP, Lefebvre F, Marais T, Mougenot N, Beauverger P, Bonne G, Briand V, Gómez AM, Muchir A. Activation of sarcolipin expression and altered calcium cycling in LMNA cardiomyopathy. Biochem Biophys Rep 2020; 22:100767. [PMID: 32490213 PMCID: PMC7261707 DOI: 10.1016/j.bbrep.2020.100767] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/12/2020] [Accepted: 04/13/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiomyopathy caused by A-type lamins gene (LMNA) mutations (LMNA cardiomyopathy) is associated with dysfunction of the heart, often leading to heart failure. LMNA cardiomyopathy is highly penetrant with bad prognosis with no specific therapy available. Searching for alternative ways to halt the progression of LMNA cardiomyopathy, we studied the role of calcium homeostasis in the evolution of this disease. We showed that sarcolipin, an inhibitor of the sarco/endoplasmic reticulum (SR) Ca2+ ATPase (SERCA) was abnormally elevated in the ventricular cardiomyocytes of mutated mice compared with wild type mice, leading to an alteration of calcium handling. This occurs early in the progression of the disease, when the left ventricular function was not altered. We further demonstrated that down regulation of sarcolipin using adeno-associated virus (AAV) 9-mediated RNA interference delays cardiac dysfunction in mouse model of LMNA cardiomyopathy. These results showed a novel role for sarcolipin on calcium homeostasis in heart and open perspectives for future therapeutic interventions to LMNA cardiomyopathy. Sarcolipin, an inhibitor of the sarco/endoplasmic reticulum (SR) Ca2+ ATPase (SERCA) was abnormally elevated in the cardiac muscle of a mouse model of cardiomyopathy caused by LMNA mutations. The elevation of sarcolipin expression leads to an alteration of calcium handling. Down regulation of sarcolipin using adeno-associated virus (AAV) 9-mediated RNA interference delays cardiac dysfunction in mouse model of cardiomyopathy caused by LMNA mutations.
Collapse
Affiliation(s)
| | - Alejandro Domínguez-Rodríguez
- Inserm, Univ. Paris-Sud, Université Paris-Saclay, UMR-S 1180, “Signaling and Cardiovascular Pathophysiology”, Châtenay-Malabry, France
| | - Jean-Pierre Benitah
- Inserm, Univ. Paris-Sud, Université Paris-Saclay, UMR-S 1180, “Signaling and Cardiovascular Pathophysiology”, Châtenay-Malabry, France
| | - Florence Lefebvre
- Inserm, Univ. Paris-Sud, Université Paris-Saclay, UMR-S 1180, “Signaling and Cardiovascular Pathophysiology”, Châtenay-Malabry, France
| | | | - Nathalie Mougenot
- Sorbonne Université, INSERM, UMS28 Phénotypage du Petit animal, Paris, F-75013, France
| | | | - Gisèle Bonne
- Sorbonne Université, INSERM UMRS974, Paris, France
| | | | - Ana-María Gómez
- Inserm, Univ. Paris-Sud, Université Paris-Saclay, UMR-S 1180, “Signaling and Cardiovascular Pathophysiology”, Châtenay-Malabry, France
| | - Antoine Muchir
- Sorbonne Université, INSERM UMRS974, Paris, France
- Corresponding author.
| |
Collapse
|
30
|
TRPV2 channel as a possible drug target for the treatment of heart failure. J Transl Med 2020; 100:207-217. [PMID: 31857697 DOI: 10.1038/s41374-019-0349-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/09/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023] Open
Abstract
Heart transplantation is currently the only viable option available for the treatment of severe heart failure conditions such as dilated cardiomyopathy. Hence, novel drugs for treating such conditions need to be developed urgently. Recent studies suggest that Ca2+ overload is involved in the onset and progression of dilated cardiomyopathy, and thus heart failure. The expression and activation of the Ca2+ permeable channel, transient receptor potential vanilloid 2 (TRPV2) channel have been found to play an essential role in sustained intracellular Ca2+ concentration increase, leading to heart failure. However, since there have been no TRPV2-specific inhibitors available until recently, the effect of TRPV2 inhibition on the pathology has not been clearly elucidated. Recent reports show that inhibiting TRPV2 activity effectively improves cardiac function, suppressing myocardial fibrosis and ameliorating the prognosis in animal models of cardiomyopathy with heart failure. In addition to that, inflammation is reported to be involved in the development of heart failure. Here, we review the recent findings on TRPV2 in cardiomyocytes and immune cells involved in the development of heart failure and discuss the current progress of drug development for the treatment of heart failure via targeting TRPV2.
Collapse
|
31
|
Pérez-Treviño P, Sepúlveda-Leal J, Altamirano J. Simultaneous assessment of calcium handling and contractility dynamics in isolated ventricular myocytes of a rat model of post-acute isoproterenol-induced cardiomyopathy. Cell Calcium 2019; 86:102138. [PMID: 31838436 DOI: 10.1016/j.ceca.2019.102138] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 12/02/2019] [Accepted: 12/02/2019] [Indexed: 12/21/2022]
Abstract
Stress-induced cardiomyopathy (SIC) results from a profound catecholaminergic surge during strong emotional or physical stress. SIC is characterized by acute left ventricular apex hypokinesia, in the absence of coronary arteries occlusion, and can lead to arrhythmias and acute heart failure. Although, most SIC patients recover, the process could be slow, and recurrence or death may occur. Despite that the SIC common denominator is a large catecholamine discharge, the pathophysiological mechanism is incompletely understood. It is thought that catecholamines have direct cytotoxicity on apical ventricular myocytes (VM), which have the highest β-adrenergic receptors density, and whose overstimulation might cause acute Ca2+ overload and oxidative stress, causing death in some VM and stunning others. Rodents receiving acute isoproterenol (ISO) overdose (OV) mimic SIC development, however, they have not been used to simultaneously assess Ca2+ handling and contractility status in isolated VM, which might explain ventricular hypokinesia. Therefore, treating rats with a single ISO-OV (67 mg/kg body weight), we sought out to characterize, with confocal imaging, Ca2+ and shortening dynamics in Fluo-4-loaded VM, during the early (1-5 days) and late post-acute phases (15 days). We found that ISO-OV VM showed contractile dysfunction; blunted shortening with slower force development and relaxation. These correlated with Ca2+ mishandling; blunted Ca2+ transient, with slower time to peak and SR Ca2+ recovery. SR Ca2+ content was low, nevertheless, diastolic Ca2+ sparks were more frequent, and their duration increased. Contractility and Ca2+ dysfunction aggravated or remained altered over time, explaining slow recovery. We conclude that diminished VM contractility is the main determinant of ISO-OV hypokinesia and is mostly related to Ca2+ mishandling.
Collapse
Affiliation(s)
- Perla Pérez-Treviño
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Av. Morones Prieto No. 3000 Pte., Monterrey, N.L., 64710, Mexico
| | - José Sepúlveda-Leal
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Av. Morones Prieto No. 3000 Pte., Monterrey, N.L., 64710, Mexico
| | - Julio Altamirano
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Av. Morones Prieto No. 3000 Pte., Monterrey, N.L., 64710, Mexico.
| |
Collapse
|
32
|
Zhang D, Zhang X, Li F, Li C, La Y, Mo F, Li G, Zhang Y, Li X, Song Q, Zhao Y, Wang W. Transcriptome Analysis Identifies Candidate Genes and Pathways Associated With Feed Efficiency in Hu Sheep. Front Genet 2019; 10:1183. [PMID: 31798641 PMCID: PMC6878960 DOI: 10.3389/fgene.2019.01183] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/24/2019] [Indexed: 12/20/2022] Open
Abstract
In the genetic improvement of livestock and poultry, residual feed intake (RFI) is an important economic trait. However, in sheep, the genetic regulatory mechanisms of RFI are unclear. In the present study, we measured the feed efficiency (FE)-related phenotypes of 137 male Hu lambs, and selected six lambs with very high (n = 3) and very low (n = 3) RFI values and analyzed their liver transcriptomes. A total of 101 differentially expressed genes were identified, of which 40 were upregulated and 61 were downregulated in the low-RFI group compared with that in the high-RFI group. The downregulated genes were mainly concentrated in immune function pathways, while the upregulated genes were mainly involved in energy metabolism pathways. Two differentially expressed genes, ADRA2A (encoding adrenoceptor alpha 2A) and RYR2 (ryanodine receptor 2), were selected as candidate genes for FE and subjected to single nucleotide polymorphism scanning and association analysis. Two synonymous mutations, ADRA2A g.1429 C > A and RYR2 g.1117 A > C, were detected, which were both significantly associated with the feed conversion rate. These findings provide a deeper understanding of the molecular mechanisms regulating FE, and reveal key genes and genetic variants that could be used to genetically improve FE in sheep.
Collapse
Affiliation(s)
- Deyin Zhang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Xiaoxue Zhang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China.,Engineering Laboratory of Sheep Breeding and Reproduction Biotechnology in Gansu Province, Minqin Zhongtian Sheep Industry Co. Ltd., Minqin, China
| | - Fadi Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China.,Engineering Laboratory of Sheep Breeding and Reproduction Biotechnology in Gansu Province, Minqin Zhongtian Sheep Industry Co. Ltd., Minqin, China.,The State Key Laboratory of Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Chong Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Yongfu La
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Futao Mo
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Guoze Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Yukun Zhang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Xiaolong Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Qizhi Song
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Yuan Zhao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Weimin Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
33
|
Murphy KR, Baggett B, Cooper LL, Lu Y, O-Uchi J, Sedivy JM, Terentyev D, Koren G. Enhancing Autophagy Diminishes Aberrant Ca 2+ Homeostasis and Arrhythmogenesis in Aging Rabbit Hearts. Front Physiol 2019; 10:1277. [PMID: 31636573 PMCID: PMC6787934 DOI: 10.3389/fphys.2019.01277] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 09/20/2019] [Indexed: 12/19/2022] Open
Abstract
Aim Aging in humans is associated with a 10–40-fold greater incidence of sudden cardiac death from malignant tachyarrhythmia. We have reported that thiol oxidation of ryanodine receptors (RyR2s) by mitochondria-derived reactive oxygen species (mito-ROS) contributes to defective Ca2+ homeostasis in cardiomyocytes (CMs) from aging rabbit hearts. However, mechanisms responsible for the increase in mito-ROS in the aging heart remain poorly understood. Here we test the hypothesis that age-associated decrease in autophagy is a major contributor to enhanced mito-ROS production and thereby pro-arrhythmic disturbances in Ca2+ homeostasis. Methods and Results Ventricular tissues from aged rabbits displayed significant downregulation of proteins involved in mitochondrial autophagy compared with tissues from young controls. Blocking autophagy with chloroquine increased total ROS production in primary rabbit CMs and mito-ROS production in HL-1 CMs. Furthermore, chloroquine treatment of HL-1 cells depolarized mitochondrial membrane potential (Δψm) to 50% that of controls. Blocking autophagy significantly increased oxidation of RyR2, resulting in enhanced propensity to pro-arrhythmic spontaneous Ca2+ release under β-adrenergic stimulation. Aberrant Ca2+ release was abolished by treatment with the mito-ROS scavenger mito-TEMPO. Importantly, the autophagy enhancer Torin1 and ATG7 overexpression reduced the rate of mito-ROS production and restored both Δψm and defective Ca2+ handling in CMs derived from aged rabbit hearts. Conclusion Decreased autophagy is a major cause of increased mito-ROS production in the aging heart. Our data suggest that promoting autophagy may reduce pathologic mito-ROS during normal aging and reduce pro-arrhythmic spontaneous Ca2+ release via oxidized RyR2s.
Collapse
Affiliation(s)
- Kevin R Murphy
- Cardiovascular Research Center at the Cardiovascular Institute, Division of Cardiology, Warren Alpert Medical School of Brown University, Providence, RI, United States.,Department of Molecular Biology, Cellular Biology and Biochemistry, Brown University, Providence, RI, United States
| | - Brett Baggett
- Cardiovascular Research Center at the Cardiovascular Institute, Division of Cardiology, Warren Alpert Medical School of Brown University, Providence, RI, United States.,Department of Molecular Biology, Cellular Biology and Biochemistry, Brown University, Providence, RI, United States
| | - Leroy L Cooper
- Cardiovascular Research Center at the Cardiovascular Institute, Division of Cardiology, Warren Alpert Medical School of Brown University, Providence, RI, United States.,Department of Biology, Vassar College, Poughkeepsie, NY, United States
| | - Yichun Lu
- Cardiovascular Research Center at the Cardiovascular Institute, Division of Cardiology, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Jin O-Uchi
- Cardiovascular Research Center at the Cardiovascular Institute, Division of Cardiology, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - John M Sedivy
- Department of Molecular Biology, Cellular Biology and Biochemistry, Brown University, Providence, RI, United States
| | - Dmitry Terentyev
- Cardiovascular Research Center at the Cardiovascular Institute, Division of Cardiology, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Gideon Koren
- Cardiovascular Research Center at the Cardiovascular Institute, Division of Cardiology, Warren Alpert Medical School of Brown University, Providence, RI, United States
| |
Collapse
|
34
|
Kitmitto A, Baudoin F, Cartwright EJ. Cardiomyocyte damage control in heart failure and the role of the sarcolemma. J Muscle Res Cell Motil 2019; 40:319-333. [PMID: 31520263 PMCID: PMC6831538 DOI: 10.1007/s10974-019-09539-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/03/2019] [Indexed: 01/07/2023]
Abstract
The cardiomyocyte plasma membrane, termed the sarcolemma, is fundamental for regulating a myriad of cellular processes. For example, the structural integrity of the cardiomyocyte sarcolemma is essential for mediating cardiac contraction by forming microdomains such as the t-tubular network, caveolae and the intercalated disc. Significantly, remodelling of these sarcolemma microdomains is a key feature in the development and progression of heart failure (HF). However, despite extensive characterisation of the associated molecular and ultrastructural events there is a lack of clarity surrounding the mechanisms driving adverse morphological rearrangements. The sarcolemma also provides protection, and is the cell's first line of defence, against external stresses such as oxygen and nutrient deprivation, inflammation and oxidative stress with a loss of sarcolemma viability shown to be a key step in cell death via necrosis. Significantly, cumulative cell death is also a feature of HF, and is linked to disease progression and loss of cardiac function. Herein, we will review the link between structural and molecular remodelling of the sarcolemma associated with the progression of HF, specifically considering the evidence for: (i) Whether intrinsic, evolutionary conserved, plasma membrane injury-repair mechanisms are in operation in the heart, and (ii) if deficits in key 'wound-healing' proteins (annexins, dysferlin, EHD2 and MG53) may play a yet to be fully appreciated role in triggering sarcolemma microdomain remodelling and/or necrosis. Cardiomyocytes are terminally differentiated with very limited regenerative capability and therefore preserving cell viability and cardiac function is crucially important. This review presents a novel perspective on sarcolemma remodelling by considering whether targeting proteins that regulate sarcolemma injury-repair may hold promise for developing new strategies to attenuate HF progression.
Collapse
Affiliation(s)
- Ashraf Kitmitto
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, AV Hill, Dover Street, Manchester, M13 9PL, UK.
| | - Florence Baudoin
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, AV Hill, Dover Street, Manchester, M13 9PL, UK
| | - Elizabeth J Cartwright
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, AV Hill, Dover Street, Manchester, M13 9PL, UK
| |
Collapse
|
35
|
Lomivorotov VV, Leonova EA, Belletti A, Shmyrev VA, Landoni G. Calcium Administration During Weaning From Cardiopulmonary Bypass: A Narrative Literature Review. J Cardiothorac Vasc Anesth 2019; 34:235-244. [PMID: 31350149 DOI: 10.1053/j.jvca.2019.06.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 06/07/2019] [Accepted: 06/10/2019] [Indexed: 02/08/2023]
Abstract
The search for safe and effective patient management strategies during weaning from cardiopulmonary bypass is ongoing; intravenous calcium is occasionally used as a first-line drug. The physiologic role of calcium suggests that it can support the function of the cardiovascular system during this critical period. Patients may be mildly hypocalcemic after cardiopulmonary bypass; however, this degree of hypocalcemia does not significantly impair the cardiovascular system. The transient beneficial effects of calcium administration (increase in arterial blood pressure, systemic vascular resistance, cardiac index, stroke volume, and coronary perfusion pressure) might be helpful in cases of moderate contractility reduction or vasoplegia. Nonetheless, effects on clinically relevant endpoints are unknown, and possible systemic side effects, such as transient reduction in internal mammary artery graft flow, attenuation of the effects of β-sympathomimetics, "stone heart" phenomenon, and pancreatic cellular injury, may limit the use of calcium salts. Further studies are needed to expand the understanding of the effects of calcium administration on patient outcomes.
Collapse
Affiliation(s)
- Vladimir V Lomivorotov
- Department of Anaesthesiology and Intensive Care, E. Meshalkin National Medical Research Center, Novosibirsk, Russia; Novosibirsk State University, Novosibirsk, Russia
| | - Elizaveta A Leonova
- Department of Anaesthesiology and Intensive Care, E. Meshalkin National Medical Research Center, Novosibirsk, Russia
| | - Alessandro Belletti
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vladimir A Shmyrev
- Department of Anaesthesiology and Intensive Care, E. Meshalkin National Medical Research Center, Novosibirsk, Russia
| | - Giovanni Landoni
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
36
|
Mundiña-Weilenmann CB, Mattiazzi A. Tracking nitroxyl-derived posttranslational modifications of phospholamban in cardiac myocytes. J Gen Physiol 2019; 151:718-721. [PMID: 31010809 PMCID: PMC6571997 DOI: 10.1085/jgp.201912342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Mundiña-Weilenmann and Mattiazzi examine new work revealing the mechanism by which nitroxide modifies uptake of Ca2+ into the SR.
Collapse
Affiliation(s)
- Cecilia Beatriz Mundiña-Weilenmann
- Centro de Investigaciones Cardiovasculares, CCT-CONICET La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Alicia Mattiazzi
- Centro de Investigaciones Cardiovasculares, CCT-CONICET La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| |
Collapse
|
37
|
Bovo E, Nikolaienko R, Bhayani S, Kahn D, Cao Q, Martin JL, Kuo IY, Robia SL, Zima AV. Novel approach for quantification of endoplasmic reticulum Ca 2+ transport. Am J Physiol Heart Circ Physiol 2019; 316:H1323-H1331. [PMID: 30901276 PMCID: PMC6620677 DOI: 10.1152/ajpheart.00031.2019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/01/2019] [Accepted: 03/04/2019] [Indexed: 01/14/2023]
Abstract
The type 2a sarco-/endoplasmic reticulum Ca2+-ATPase (SERCA2a) plays a key role in Ca2+ regulation in the heart. However, available techniques to study SERCA function are either cell destructive or lack sensitivity. The goal of this study was to develop an approach to selectively measure SERCA2a function in the cellular environment. The genetically encoded Ca2+ sensor R-CEPIA1er was used to measure the concentration of Ca2+ in the lumen of the endoplasmic reticulum (ER) ([Ca2+]ER) in HEK293 cells expressing human SERCA2a. Coexpression of the ER Ca2+ release channel ryanodine receptor (RyR2) created a Ca2+ release/reuptake system that mimicked aspects of cardiac myocyte Ca2+ handling. SERCA2a function was quantified from the rate of [Ca2+]ER refilling after ER Ca2+ depletion; then, ER Ca2+ leak was measured after SERCA inhibition. ER Ca2+ uptake and leak were analyzed as a function of [Ca2+]ER to determine maximum ER Ca2+ uptake rate and maximum ER Ca2+ load. The sensitivity of this assay was validated by analyzing effects of SERCA inhibitors, [ATP]/[ADP], oxidative stress, phospholamban, and a loss-of-function SERCA2a mutation. In addition, the feasibility of using R-CEPIA1er to study SERCA2a in a native system was evaluated by using in vivo gene delivery to express R-CEPIA1er in mouse hearts. After ventricular myocyte isolation, the same methodology used in HEK293 cells was applied to study endogenous SERCA2a. In conclusion, this new approach can be used as a sensitive screening tool to study the effect of different drugs, posttranslational modifications, and mutations on SERCA function. NEW & NOTEWORTHY The aim of this study was to develop a sensitive approach to selectively measure sarco-/endoplasmic reticulum Ca2+-ATPase (SERCA) function in the cellular environment. The newly developed Ca2+ sensor R-CEPIA1er was used to successfully analyze Ca2+ uptake mediated by recombinant and native cardiac SERCA. These results demonstrate that this new approach can be used as a powerful tool to study new mechanisms of Ca2+ pump regulation.
Collapse
Affiliation(s)
- Elisa Bovo
- Department of Cell and Molecular Physiology, Loyola University Chicago , Chicago, Illinois
| | - Roman Nikolaienko
- Department of Cell and Molecular Physiology, Loyola University Chicago , Chicago, Illinois
| | - Siddharth Bhayani
- Department of Cell and Molecular Physiology, Loyola University Chicago , Chicago, Illinois
| | - Daniel Kahn
- Department of Cell and Molecular Physiology, Loyola University Chicago , Chicago, Illinois
| | - Quan Cao
- Department of Cell and Molecular Physiology, Loyola University Chicago , Chicago, Illinois
| | - Jody L Martin
- Department of Physiology and Biophysics, University of Illinois at Chicago , Chicago, Illinois
| | - Ivana Y Kuo
- Department of Cell and Molecular Physiology, Loyola University Chicago , Chicago, Illinois
| | - Seth L Robia
- Department of Cell and Molecular Physiology, Loyola University Chicago , Chicago, Illinois
| | - Aleksey V Zima
- Department of Cell and Molecular Physiology, Loyola University Chicago , Chicago, Illinois
| |
Collapse
|
38
|
Hamilton S, Terentyev D. Altered Intracellular Calcium Homeostasis and Arrhythmogenesis in the Aged Heart. Int J Mol Sci 2019; 20:ijms20102386. [PMID: 31091723 PMCID: PMC6566636 DOI: 10.3390/ijms20102386] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 04/29/2019] [Accepted: 05/08/2019] [Indexed: 12/17/2022] Open
Abstract
Aging of the heart is associated with a blunted response to sympathetic stimulation, reduced contractility, and increased propensity for arrhythmias, with the risk of sudden cardiac death significantly increased in the elderly population. The altered cardiac structural and functional phenotype, as well as age-associated prevalent comorbidities including hypertension and atherosclerosis, predispose the heart to atrial fibrillation, heart failure, and ventricular tachyarrhythmias. At the cellular level, perturbations in mitochondrial function, excitation-contraction coupling, and calcium homeostasis contribute to this electrical and contractile dysfunction. Major determinants of cardiac contractility are the intracellular release of Ca2+ from the sarcoplasmic reticulum by the ryanodine receptors (RyR2), and the following sequestration of Ca2+ by the sarco/endoplasmic Ca2+-ATPase (SERCa2a). Activity of RyR2 and SERCa2a in myocytes is not only dependent on expression levels and interacting accessory proteins, but on fine-tuned regulation via post-translational modifications. In this paper, we review how aberrant changes in intracellular Ca2+ cycling via these proteins contributes to arrhythmogenesis in the aged heart.
Collapse
Affiliation(s)
- Shanna Hamilton
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Dmitry Terentyev
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
39
|
Afanasiev SA, Kondratieva DS, Egorova MV, Akhmedov SD, Budnikova OV, Popov SV. Features the interaction of functional and metabolic remodeling of myocardium in comorbid course of ischemic heart disease and 2 type diabetes mellitus. DIABETES MELLITUS 2019. [DOI: 10.14341/dm9735] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background: Metabolic and structural changes in cardiomyocytes in diabetes mellitus lead to aggravation of contractile myocardial dysfunction in coronary heart disease (CHD). The contractility dysfunction of cardiomyocytes is determined by a change in the levels of sarcoplasmic reticulum (SR) Ca2+-ATPase and energetic supply of the cardiomyocytes.
Aims: To study the features of functional remodeling of the heart muscle in coronary heart disease with and without type 2 diabetes mellitus (DM2) depend on the level of Ca2+-ATPase and the activity of enzymes involved in energy metabolism.
Materials and methods: The work was performed on the heart biopsy of patients with CHD and patients with CHD combined with DM2. The inotropic reaction of myocardial strips on rest periods was assessed. The expression level of Ca2+-ATPase, the activity of enzymes succinate dehydrogenase (SDH) and lactate dehydrogenase (LDH) and the intensity of oxidative phosphorylation processes were determined.
Results: The interval-force relationship in patients with CHD with and without DM2 had both negative and positive dynamics. The positive dynamics corresponds to the "high content" of the Ca2+-ATPase and the negative dynamics corresponds to the "low content" were found. At the combined pathology the positive inotropic dynamics is more pronounced and corresponds to a higher protein level. In the patients myocardium with CHD the activity of SDH and LDH was higher, while the oxygen uptake rate by mitochondria was higher in the myocardium with combined pathology.
Conclusions: The potentiation of inotropic response of patient myocardium with CHD with and without DM2 corresponds to the "high level" of Ca2+-ATPase. In the combined pathology the inotropic capabilities of the myocardium are more expressed. In CHD the synthesis of ATP in cardiomyocytes is realized mainly due to glycolytic processes and Krebs cycle. In combined pathology the ATP synthesis is realized to a greater extent due to the oxidative phosphorylation.
Collapse
|
40
|
Savoji H, Mohammadi MH, Rafatian N, Toroghi MK, Wang EY, Zhao Y, Korolj A, Ahadian S, Radisic M. Cardiovascular disease models: A game changing paradigm in drug discovery and screening. Biomaterials 2019; 198:3-26. [PMID: 30343824 PMCID: PMC6397087 DOI: 10.1016/j.biomaterials.2018.09.036] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/11/2018] [Accepted: 09/22/2018] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease is the leading cause of death worldwide. Although investment in drug discovery and development has been sky-rocketing, the number of approved drugs has been declining. Cardiovascular toxicity due to therapeutic drug use claims the highest incidence and severity of adverse drug reactions in late-stage clinical development. Therefore, to address this issue, new, additional, replacement and combinatorial approaches are needed to fill the gap in effective drug discovery and screening. The motivation for developing accurate, predictive models is twofold: first, to study and discover new treatments for cardiac pathologies which are leading in worldwide morbidity and mortality rates; and second, to screen for adverse drug reactions on the heart, a primary risk in drug development. In addition to in vivo animal models, in vitro and in silico models have been recently proposed to mimic the physiological conditions of heart and vasculature. Here, we describe current in vitro, in vivo, and in silico platforms for modelling healthy and pathological cardiac tissues and their advantages and disadvantages for drug screening and discovery applications. We review the pathophysiology and the underlying pathways of different cardiac diseases, as well as the new tools being developed to facilitate their study. We finally suggest a roadmap for employing these non-animal platforms in assessing drug cardiotoxicity and safety.
Collapse
Affiliation(s)
- Houman Savoji
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada
| | - Mohammad Hossein Mohammadi
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada; Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada
| | - Naimeh Rafatian
- Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada
| | - Masood Khaksar Toroghi
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada
| | - Erika Yan Wang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada
| | - Yimu Zhao
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada
| | - Anastasia Korolj
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada
| | - Samad Ahadian
- Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada; Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada.
| |
Collapse
|
41
|
Hamilton S, Polina I, Terentyeva R, Bronk P, Kim TY, Roder K, Clements RT, Koren G, Choi BR, Terentyev D. PKA phosphorylation underlies functional recruitment of sarcolemmal SK2 channels in ventricular myocytes from hypertrophic hearts. J Physiol 2019; 598:2847-2873. [PMID: 30771223 PMCID: PMC7496687 DOI: 10.1113/jp277618] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 02/08/2019] [Indexed: 12/11/2022] Open
Abstract
KEY POINTS Small-conductance Ca2+ -activated K+ (SK) channels expressed in ventricular myocytes are dormant in health, yet become functional in cardiac disease. SK channels are voltage independent and their gating is controlled by intracellular [Ca2+ ] in a biphasic manner. Submicromolar [Ca2+ ] activates the channel via constitutively-bound calmodulin, whereas higher [Ca2+ ] exerts inhibitory effect during depolarization. Using a rat model of cardiac hypertrophy induced by thoracic aortic banding, we found that functional upregulation of SK2 channels in hypertrophic rat ventricular cardiomyocytes is driven by protein kinase A (PKA) phosphorylation. Using site-directed mutagenesis, we identified serine-465 as the site conferring PKA-dependent effects on SK2 channel function. PKA phosphorylation attenuates ISK rectification by reducing the Ca2+ /voltage-dependent inhibition of SK channels without changing their sensitivity to activating submicromolar [Ca2+ ]i . This mechanism underlies the functional recruitment of SK channels not only in cardiac disease, but also in normal physiology, contributing to repolarization under conditions of enhanced adrenergic drive. ABSTRACT Small-conductance Ca2+ -activated K+ (SK) channels expressed in ventricular myocytes (VMs) are dormant in health, yet become functional in cardiac disease. We aimed to test the hypothesis that post-translational modification of SK channels under conditions accompanied by enhanced adrenergic drive plays a central role in disease-related activation of the channels. We investigated this phenomenon using a rat model of hypertrophy induced by thoracic aortic banding (TAB). Western blot analysis using anti-pan-serine/threonine antibodies demonstrated enhanced phosphorylation of immunoprecipitated SK2 channels in VMs from TAB rats vs. Shams, which was reversible by incubation of the VMs with PKA inhibitor H89 (1 μmol L-1 ). Patch clamped VMs under basal conditions from TABs but not Shams exhibited outward current sensitive to the specific SK inhibitor apamin (100 nmol L-1 ), which was eliminated by inhibition of PKA (1 μmol L-1 ). Beta-adrenergic stimulation (isoproterenol, 100 nmol L-1 ) evoked ISK in VMs from Shams, resulting in shortening of action potentials in VMs and ex vivo optically mapped Sham hearts. Using adenoviral gene transfer, wild-type and mutant SK2 channels were overexpressed in adult rat VMs, revealing serine-465 as the site that elicits PKA-dependent phosphorylation effects on SK2 channel function. Concurrent confocal Ca2+ imaging experiments established that PKA phosphorylation lessens rectification of ISK via reduction Ca2+ /voltage-dependent inhibition of the channels at high [Ca2+ ] without affecting their sensitivity to activation by Ca2+ in the submicromolar range. In conclusion, upregulation of SK channels in diseased VMs is mediated by hyperadrenergic drive in cardiac hypertrophy, with functional effects on the channel conferred by PKA-dependent phosphorylation at serine-465.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Iuliia Polina
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA.,Medical University of South Carolina, Department of Medicine, Division of Nephrology, Charleston, SC, USA
| | - Radmila Terentyeva
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Peter Bronk
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA
| | - Tae Yun Kim
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA
| | - Karim Roder
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA
| | - Richard T Clements
- Department of Surgery, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA.,Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Gideon Koren
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA
| | - Bum-Rak Choi
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA
| | - Dmitry Terentyev
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
42
|
Cao JL, Adaniya SM, Cypress MW, Suzuki Y, Kusakari Y, Jhun BS, O-Uchi J. Role of mitochondrial Ca 2+ homeostasis in cardiac muscles. Arch Biochem Biophys 2019; 663:276-287. [PMID: 30684463 PMCID: PMC6469710 DOI: 10.1016/j.abb.2019.01.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/10/2019] [Accepted: 01/22/2019] [Indexed: 12/22/2022]
Abstract
Recent discoveries of the molecular identity of mitochondrial Ca2+ influx/efflux mechanisms have placed mitochondrial Ca2+ transport at center stage in views of cellular regulation in various cell-types/tissues. Indeed, mitochondria in cardiac muscles also possess the molecular components for efficient uptake and extraction of Ca2+. Over the last several years, multiple groups have taken advantage of newly available molecular information about these proteins and applied genetic tools to delineate the precise mechanisms for mitochondrial Ca2+ handling in cardiomyocytes and its contribution to excitation-contraction/metabolism coupling in the heart. Though mitochondrial Ca2+ has been proposed as one of the most crucial secondary messengers in controlling a cardiomyocyte's life and death, the detailed mechanisms of how mitochondrial Ca2+ regulates physiological mitochondrial and cellular functions in cardiac muscles, and how disorders of this mechanism lead to cardiac diseases remain unclear. In this review, we summarize the current controversies and discrepancies regarding cardiac mitochondrial Ca2+ signaling that remain in the field to provide a platform for future discussions and experiments to help close this gap.
Collapse
Affiliation(s)
- Jessica L Cao
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, USA; Department of Medicine, Division of Cardiology, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Stephanie M Adaniya
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, USA; Department of Medicine, Division of Cardiology, The Warren Alpert Medical School of Brown University, Providence, RI, USA; Lillehei Heart Institute, Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA
| | - Michael W Cypress
- Lillehei Heart Institute, Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA
| | - Yuta Suzuki
- Lillehei Heart Institute, Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA
| | - Yoichiro Kusakari
- Department of Cell Physiology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Bong Sook Jhun
- Lillehei Heart Institute, Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA
| | - Jin O-Uchi
- Lillehei Heart Institute, Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
43
|
Hamilton S, Terentyeva R, Kim TY, Bronk P, Clements RT, O-Uchi J, Csordás G, Choi BR, Terentyev D. Pharmacological Modulation of Mitochondrial Ca 2+ Content Regulates Sarcoplasmic Reticulum Ca 2+ Release via Oxidation of the Ryanodine Receptor by Mitochondria-Derived Reactive Oxygen Species. Front Physiol 2018; 9:1831. [PMID: 30622478 PMCID: PMC6308295 DOI: 10.3389/fphys.2018.01831] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 12/06/2018] [Indexed: 01/09/2023] Open
Abstract
In a physiological setting, mitochondria increase oxidative phosphorylation during periods of stress to meet increased metabolic demand. This in part is mediated via enhanced mitochondrial Ca2+ uptake, an important regulator of cellular ATP homeostasis. In a pathophysiological setting pharmacological modulation of mitochondrial Ca2+ uptake or retention has been suggested as a therapeutic strategy to improve metabolic homeostasis or attenuate Ca2+-dependent arrhythmias in cardiac disease states. To explore the consequences of mitochondrial Ca2+ accumulation, we tested the effects of kaempferol, an activator of mitochondrial Ca2+ uniporter (MCU), CGP-37157, an inhibitor of mitochondrial Na+/Ca2+ exchanger, and MCU inhibitor Ru360 in rat ventricular myocytes (VMs) from control rats and rats with hypertrophy induced by thoracic aortic banding (TAB). In periodically paced VMs under β-adrenergic stimulation, treatment with kaempferol (10 μmol/L) or CGP-37157 (1 μmol/L) enhanced mitochondrial Ca2+ accumulation monitored by mitochondrial-targeted Ca2+ biosensor mtRCamp1h. Experiments with mitochondrial membrane potential-sensitive dye TMRM revealed this was accompanied by depolarization of the mitochondrial matrix. Using redox-sensitive OMM-HyPer and ERroGFP_iE biosensors, we found treatment with kaempferol or CGP-37157 increased the levels of reactive oxygen species (ROS) in mitochondria and the sarcoplasmic reticulum (SR), respectively. Confocal Ca2+ imaging showed that accelerated Ca2+ accumulation reduced Ca2+ transient amplitude and promoted generation of spontaneous Ca2+ waves in VMs paced under ISO, suggestive of abnormally high activity of the SR Ca2+ release channel ryanodine receptor (RyR). Western blot analyses showed increased RyR oxidation after treatment with kaempferol or CGP-37157 vs. controls. Furthermore, in freshly isolated TAB VMs, confocal Ca2+ imaging demonstrated that enhancement of mitochondrial Ca2+ accumulation further perturbed global Ca2+ handling, increasing the number of cells exhibiting spontaneous Ca2+ waves, shortening RyR refractoriness and decreasing SR Ca2+ content. In ex vivo optically mapped TAB hearts, kaempferol exacerbated proarrhythmic phenotype. On the contrary, incubation of cells with MCU inhibitor Ru360 (2 μmol/L, 30 min) normalized RyR oxidation state, improved intracellular Ca2+ homeostasis and reduced triggered activity in ex vivo TAB hearts. These findings suggest facilitation of mitochondrial Ca2+ uptake in cardiac disease can exacerbate proarrhythmic disturbances in Ca2+ homeostasis via ROS and enhanced activity of oxidized RyRs, while strategies to reduce mitochondrial Ca2+ accumulation can be protective.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, United States
| | - Radmila Terentyeva
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, United States
| | - Tae Yun Kim
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, United States
| | - Peter Bronk
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, United States
| | - Richard T. Clements
- Department of Surgery, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, United States
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, United States
| | - Jin O-Uchi
- Lillehei Heart Institute University of Minnesota, Cancer and Cardiovascular Research Building, Minneapolis, MN, United States
| | - György Csordás
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Bum-Rak Choi
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, United States
| | - Dmitry Terentyev
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, United States
| |
Collapse
|
44
|
Nikolaienko R, Bovo E, Zima AV. Redox Dependent Modifications of Ryanodine Receptor: Basic Mechanisms and Implications in Heart Diseases. Front Physiol 2018; 9:1775. [PMID: 30574097 PMCID: PMC6291498 DOI: 10.3389/fphys.2018.01775] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 11/23/2018] [Indexed: 12/12/2022] Open
Abstract
Heart contraction vitally depends on tightly controlled intracellular Ca regulation. Because contraction is mainly driven by Ca released from the sarcoplasmic reticulum (SR), this organelle plays a particularly important role in Ca regulation. The type two ryanodine receptor (RyR2) is the major SR Ca release channel in ventricular myocytes. Several cardiac pathologies, including myocardial infarction and heart failure, are associated with increased RyR2 activity and diastolic SR Ca leak. It has been suggested that the increased RyR2 activity plays an important role in arrhythmias and contractile dysfunction. Several studies have linked increased SR Ca leak during myocardial infarction and heart failure to the activation of RyR2 in response to oxidative stress. This activation might include direct oxidation of RyR2 as well as indirect activation via phosphorylation or altered interactions with regulatory proteins. Out of ninety cysteine residues per RyR2 subunit, twenty one were reported to be in reduced state that could be potential targets for redox modifications that include S-nitrosylation, S-glutathionylation, and disulfide cross-linking. Despite its clinical significance, molecular mechanisms of RyR dysfunction during oxidative stress are not fully understood. Herein we review the most recent insights into redox-dependent modulation of RyR2 during oxidative stress and heart diseases.
Collapse
Affiliation(s)
- Roman Nikolaienko
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, United States
| | - Elisa Bovo
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, United States
| | - Aleksey V Zima
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, United States
| |
Collapse
|
45
|
Loucks AD, O'Hara T, Trayanova NA. Degradation of T-Tubular Microdomains and Altered cAMP Compartmentation Lead to Emergence of Arrhythmogenic Triggers in Heart Failure Myocytes: An in silico Study. Front Physiol 2018; 9:1737. [PMID: 30564142 PMCID: PMC6288429 DOI: 10.3389/fphys.2018.01737] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 11/16/2018] [Indexed: 11/13/2022] Open
Abstract
Heart failure (HF) is one of the most common causes of morbidity and mortality worldwide. Although many patients suffering from HF die from sudden cardiac death caused by arrhythmias, the mechanism linking HF remodeling to an increased arrhythmogenic propensity remains incomplete. HF is typically characterized by a progressive loss of transverse tubule (T-tubule) domains, which leads to an altered distribution of L-type calcium channels (LTCCs). Microdomain degradation also causes the disruption of the β2 adrenergic receptor (β2AR) and phosphodiesterase (PDE) signaling localization, normally confined to the dyadic space. The goal of this study was to analyze how these subcellular changes affect the function of LTCCs and lead to the emergence of ventricular cell-level triggers of arrhythmias. To accomplish this, we developed a novel computational model of a human ventricular HF myocyte in which LTCCs were divided into six different populations, based on their location and signaling environment they experience. To do so, we included T-tubular microdomain remodeling which led to a subset of LTCCs to be redistributed from the T-tubular to the surface membrane and allowed for different levels of phosphorylation of LTCCs by PKA, based on the presence of β2ARs and PDEs. The model was used to study the behavior of the LTCC current (ICaL) under basal and sympathetic stimulation and its effect on cellular action potential. Our results showed that channels redistributed from the T-tubular membrane to the bulk of the sarcolemma displayed an altered function in their new, non-native signaling domain. Incomplete calcium dependent inactivation, which resulted in a longer-lasting and larger-in-magnitude LTCC current, was observed when we decoupled LTCCs from ryanodine receptors and removed them from the dyadic space. The magnitude of the LTCC current, especially in the surface sarcolemma, was also increased via phosphorylation by the redistributed β2ARs and PDEs. These changes in LTCC current led to the development of early afterdepolarizations. Thus, our study shows that altered LTCC function is a potential cause for the emergence of cell-level triggers of arrhythmia, and that β2ARs and PDEs present useful therapeutic targets for treatment of HF and prevention of sudden cardiac death.
Collapse
Affiliation(s)
- Alexandra D Loucks
- Institute for Computational Medicine and Department of Biomedical Engineering at Johns Hopkins University, Baltimore, MD, United States
| | - Thomas O'Hara
- Institute for Computational Medicine and Department of Biomedical Engineering at Johns Hopkins University, Baltimore, MD, United States
| | - Natalia A Trayanova
- Institute for Computational Medicine and Department of Biomedical Engineering at Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
46
|
Hamilton S, Terentyev D. Proarrhythmic Remodeling of Calcium Homeostasis in Cardiac Disease; Implications for Diabetes and Obesity. Front Physiol 2018. [PMID: 30425651 DOI: 10.3389/fphys.2018.01517, 10.3389/fpls.2018.01517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A rapid growth in the incidence of diabetes and obesity has transpired to a major heath issue and economic burden in the postindustrial world, with more than 29 million patients affected in the United States alone. Cardiovascular defects have been established as the leading cause of mortality and morbidity of diabetic patients. Over the last decade, significant progress has been made in delineating mechanisms responsible for the diminished cardiac contractile function and enhanced propensity for malignant cardiac arrhythmias characteristic of diabetic disease. Rhythmic cardiac contractility relies upon the precise interplay between several cellular Ca2+ transport protein complexes including plasmalemmal L-type Ca2+ channels (LTCC), Na+-Ca2+ exchanger (NCX1), Sarco/endoplasmic Reticulum (SR) Ca2+-ATPase (SERCa2a) and ryanodine receptors (RyR2s), the SR Ca2+ release channels. Here we provide an overview of changes in Ca2+ homeostasis in diabetic ventricular myocytes and discuss the therapeutic potential of targeting Ca2+ handling proteins in the prevention of diabetes-associated cardiomyopathy and arrhythmogenesis.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States.,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States
| | - Dmitry Terentyev
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States.,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States
| |
Collapse
|
47
|
Hamilton S, Terentyev D. Proarrhythmic Remodeling of Calcium Homeostasis in Cardiac Disease; Implications for Diabetes and Obesity. Front Physiol 2018; 9:1517. [PMID: 30425651 PMCID: PMC6218530 DOI: 10.3389/fphys.2018.01517] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/09/2018] [Indexed: 12/28/2022] Open
Abstract
A rapid growth in the incidence of diabetes and obesity has transpired to a major heath issue and economic burden in the postindustrial world, with more than 29 million patients affected in the United States alone. Cardiovascular defects have been established as the leading cause of mortality and morbidity of diabetic patients. Over the last decade, significant progress has been made in delineating mechanisms responsible for the diminished cardiac contractile function and enhanced propensity for malignant cardiac arrhythmias characteristic of diabetic disease. Rhythmic cardiac contractility relies upon the precise interplay between several cellular Ca2+ transport protein complexes including plasmalemmal L-type Ca2+ channels (LTCC), Na+-Ca2+ exchanger (NCX1), Sarco/endoplasmic Reticulum (SR) Ca2+-ATPase (SERCa2a) and ryanodine receptors (RyR2s), the SR Ca2+ release channels. Here we provide an overview of changes in Ca2+ homeostasis in diabetic ventricular myocytes and discuss the therapeutic potential of targeting Ca2+ handling proteins in the prevention of diabetes-associated cardiomyopathy and arrhythmogenesis.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States.,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States
| | - Dmitry Terentyev
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States.,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States
| |
Collapse
|
48
|
Hamilton S, Terentyev D. Proarrhythmic Remodeling of Calcium Homeostasis in Cardiac Disease; Implications for Diabetes and Obesity. Front Physiol 2018; 9:1517. [PMID: 30425651 PMCID: PMC6218530 DOI: 10.3389/fphys.2018.01517,+10.3389/fpls.2018.01517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022] Open
Abstract
A rapid growth in the incidence of diabetes and obesity has transpired to a major heath issue and economic burden in the postindustrial world, with more than 29 million patients affected in the United States alone. Cardiovascular defects have been established as the leading cause of mortality and morbidity of diabetic patients. Over the last decade, significant progress has been made in delineating mechanisms responsible for the diminished cardiac contractile function and enhanced propensity for malignant cardiac arrhythmias characteristic of diabetic disease. Rhythmic cardiac contractility relies upon the precise interplay between several cellular Ca2+ transport protein complexes including plasmalemmal L-type Ca2+ channels (LTCC), Na+-Ca2+ exchanger (NCX1), Sarco/endoplasmic Reticulum (SR) Ca2+-ATPase (SERCa2a) and ryanodine receptors (RyR2s), the SR Ca2+ release channels. Here we provide an overview of changes in Ca2+ homeostasis in diabetic ventricular myocytes and discuss the therapeutic potential of targeting Ca2+ handling proteins in the prevention of diabetes-associated cardiomyopathy and arrhythmogenesis.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States
| | - Dmitry Terentyev
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States,*Correspondence: Dmitry Terentyev,
| |
Collapse
|
49
|
van Opbergen CJ, van der Voorn SM, Vos MA, de Boer TP, van Veen TA. Cardiac Ca2+ signalling in zebrafish: Translation of findings to man. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 138:45-58. [DOI: 10.1016/j.pbiomolbio.2018.05.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/09/2018] [Accepted: 05/04/2018] [Indexed: 02/07/2023]
|
50
|
Blanch i Salvador J, Egger M. Obstruction of ventricular Ca 2+ -dependent arrhythmogenicity by inositol 1,4,5-trisphosphate-triggered sarcoplasmic reticulum Ca 2+ release. J Physiol 2018; 596:4323-4340. [PMID: 30004117 PMCID: PMC6138286 DOI: 10.1113/jp276319] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 07/06/2018] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Augmented inositol 1,4,5-trisphosphate (IP3 ) receptor (IP3 R2) expression has been linked to a variety of cardiac pathologies. Although cardiac IP3 R2 function has been in the focus of research for some time, a detailed understanding of its potential role in ventricular myocyte excitation-contraction coupling under pathophysiological conditions remains elusive. The present study focuses on mechanisms of IP3 R2-mediated sarcoplasmic reticulum (SR)-Ca2+ release in ventricular excitation-contraction coupling under IP3 R2-overexpressing conditions by studying intracellular Ca2+ events. We report that, upon IP3 R2 overexpression in ventricular myocytes, IP3 -induced Ca2+ release (IP3 ICR) modulates the SR-Ca2+ content via "eventless" SR-Ca2+ release, affecting the global SR-Ca2+ leak. Thus, IP3 R2 activation could act as a SR-Ca2+ gateway mechanism to escape ominous SR-Ca2+ overload. Our approach unmasks a so far unrecognized mechanism by which "eventless" IP3 ICR plays a protective role against ventricular Ca2+ -dependent arrhythmogenicity. ABSTRACT Augmented inositol 1,4,5-trisphosphate (IP3 ) receptor (IP3 R2) function has been linked to a variety of cardiac pathologies including cardiac arrhythmias. The functional role of IP3 -induced Ca2+ release (IP3 ICR) within ventricular excitation-contraction coupling (ECC) remains elusive. As part of pathophysiological cellular remodelling, IP3 R2s are overexpressed and have been repeatedly linked to enhanced Ca2+ -dependent arrhythmogenicity. In this study we test the hypothesis that an opposite scenario might be plausible in which IP3 ICR is part of an ECC protecting mechanism, resulting in a Ca2+ -dependent anti-arrhythmogenic response on the cellular scale. IP3 R2 activation was triggered via endothelin-1 or IP3 -salt application in single ventricular myocytes from a cardiac-specific IP3 R type 2 overexpressing mouse model. Upon IP3 R2 overexpression, IP3 R activation reduced Ca2+ -wave occurrence (46 vs. 21.72%; P < 0.001) while its block increased SR-Ca2+ content (∼29.4% 2-aminoethoxydiphenyl borate, ∼16.4% xestospongin C; P < 0.001), suggesting an active role of IP3 ICR in SR-Ca2+ content regulation and anti-arrhythmogenic function. Pharmacological separation of ryanodine receptor RyR2 and IP3 R2 functions and two-dimensional Ca2+ event analysis failed to identify local IP3 ICR events (Ca2+ puffs). SR-Ca2+ leak measurements revealed that under pathophysiological conditions, "eventless" SR-Ca2+ efflux via enhanced IP3 ICR maintains the SR-Ca2+ content below Ca2+ spark threshold, preventing aberrant SR-Ca2+ release and resulting in a protective mechanism against SR-Ca2+ overload and arrhythmias. Our results support a so far unrecognized modulatory mechanism in ventricular myocytes working in an anti-arrhythmogenic fashion.
Collapse
Affiliation(s)
| | - Marcel Egger
- Department of PhysiologyUniversity of BernBuehlplatz 5CH‐3012BernSwitzerland
| |
Collapse
|