1
|
Bahrami P, Aromolaran KA, Aromolaran AS. Mechanistic Relevance of Ventricular Arrhythmias in Heart Failure with Preserved Ejection Fraction. Int J Mol Sci 2024; 25:13423. [PMID: 39769189 PMCID: PMC11677834 DOI: 10.3390/ijms252413423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/05/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is increasing at an alarming rate worldwide, with limited effective therapeutic interventions in patients. Sudden cardiac death (SCD) and ventricular arrhythmias present substantial risks for the prognosis of these patients. Obesity is a risk factor for HFpEF and life-threatening arrhythmias. Obesity and its associated metabolic dysregulation, leading to metabolic syndrome, are an epidemic that poses a significant public health problem. More than one-third of the world population is overweight or obese, leading to an enhanced risk of incidence and mortality due to cardiovascular disease (CVD). Obesity predisposes patients to atrial fibrillation and ventricular and supraventricular arrhythmias-conditions that are caused by dysfunction in the electrical activity of the heart. To date, current therapeutic options for the cardiomyopathy of obesity are limited, suggesting that there is considerable room for the development of therapeutic interventions with novel mechanisms of action that will help normalize sinus rhythms in obese patients. Emerging candidates for modulation by obesity are cardiac ion channels and Ca-handling proteins. However, the underlying molecular mechanisms of the impact of obesity on these channels and Ca-handling proteins remain incompletely understood. Obesity is marked by the accumulation of adipose tissue, which is associated with a variety of adverse adaptations, including dyslipidemia (or abnormal systemic levels of free fatty acids), increased secretion of proinflammatory cytokines, fibrosis, hyperglycemia, and insulin resistance, which cause electrical remodeling and, thus, predispose patients to arrhythmias. Furthermore, adipose tissue is also associated with the accumulation of subcutaneous and visceral fat, which is marked by distinct signaling mechanisms. Thus, there may also be functional differences in the effects of the regional distribution of fat deposits on ion channel/Ca-handling protein expression. Evaluating alterations in their functional expression in obesity will lead to progress in the knowledge of the mechanisms responsible for obesity-related arrhythmias. These advances are likely to reveal new targets for pharmacological modulation. Understanding how obesity and related mechanisms lead to cardiac electrical remodeling is likely to have a significant medical and economic impact. Nevertheless, substantial knowledge gaps remain regarding HFpEF treatment, requiring further investigations to identify potential therapeutic targets. The objective of this study is to review cardiac ion channel/Ca-handling protein remodeling in the predisposition to metabolic HFpEF and arrhythmias. This review further highlights interleukin-6 (IL-6) as a potential target, cardiac bridging integrator 1 (cBIN1) as a promising gene therapy agent, and leukotriene B4 (LTB4) as an underappreciated pathway in future HFpEF management.
Collapse
Affiliation(s)
- Pegah Bahrami
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, 95 S 2000 E, Salt Lake City, UT 84112, USA; (P.B.); (K.A.A.)
| | - Kelly A. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, 95 S 2000 E, Salt Lake City, UT 84112, USA; (P.B.); (K.A.A.)
| | - Ademuyiwa S. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, 95 S 2000 E, Salt Lake City, UT 84112, USA; (P.B.); (K.A.A.)
- Department of Surgery, Division of Cardiothoracic Surgery, Nutrition & Integrative Physiology, Biochemistry & Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| |
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 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 & Physiology Boonshoft School of Medicine and College of Science and Mathematics, Wright State University Dayton OH
| | - John A Miller
- Department of Neuroscience, Cell Biology & Physiology Boonshoft School of Medicine and College of Science and Mathematics, Wright State University Dayton OH
| | - Andrew R Ednie
- Department of Neuroscience, Cell Biology & Physiology Boonshoft School of Medicine and College of Science and Mathematics, Wright State University Dayton OH
| | - Eric S Bennett
- Department of Neuroscience, Cell Biology & Physiology Boonshoft School of Medicine and College of Science and Mathematics, Wright State University Dayton OH
| |
Collapse
|
3
|
Kaneko N, Loughrey CM, Smith G, Matsuda R, Hasunuma T, Mark PB, Toda M, Shinozaki M, Otani N, Kayley S, Da Silva Costa A, Martin TP, Dobi S, Saxena P, Shimamoto K, Ishikawa T, Kambayashi R, Riddell A, Elliott EB, McCarroll CS, Sakai T, Mitsuhisa Y, Hirano S, Kitai T, Kusano K, Inoue Y, Nakamura M, Kikuchi M, Toyoda S, Taguchi I, Fujiwara T, Sugiyama A, Kumagai Y, Iwata K. A novel ryanodine receptor 2 inhibitor, M201-A, enhances natriuresis, renal function and lusi-inotropic actions: Preclinical and phase I study. Br J Pharmacol 2024; 181:3401-3419. [PMID: 38773354 DOI: 10.1111/bph.16379] [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: 10/12/2023] [Revised: 02/17/2024] [Accepted: 02/20/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND AND PURPOSE The ryanodine receptor 2 (RyR2) is present in both the heart and kidneys, and plays a crucial role in maintaining intracellular Ca2+ homeostasis in cells in these organs. This study aimed to investigate the impact of M201-A on RyR2, as well as studying its effects on cardiac and renal functions in preclinical and clinical studies. EXPERIMENTAL APPROACH Following the administration of M201-A (1,4-benzothiazepine-1-oxide derivative), we monitored diastolic Ca2+ leak via RyR2 and intracellular Ca2+ concentration in isolated rat cardiomyocytes and in cardiac and renal function in animals. In a clinical study, M201-A was administered intravenously at doses of 0.2 and 0.4 mg·kg-1 once daily for 20 min for four consecutive days in healthy males, with the assessment of haemodynamic responses. KEY RESULTS In rat heart cells, M201-A effectively inhibited spontaneous diastolic Ca2+ leakage through RyR2 and exhibited positive lusi-inotropic effects on the rat heart. Additionally, it enhanced natriuresis and improved renal function in dogs. In human clinical studies, when administered intravenously, M201-A demonstrated an increase in natriuresis, glomerular filtration rate and creatinine clearance, while maintaining acceptable levels of drug safety and tolerability. CONCLUSIONS AND IMPLICATIONS The novel drug M201-A inhibited diastolic Ca2+ leak via RyR2, improved cardiac lusi-inotropic effects in rats, and enhanced natriuresis and renal function in humans. These findings suggest that this drug may offer a potential new treatment option for chronic kidney disease and heart failure.
Collapse
Affiliation(s)
- Noboru Kaneko
- Department of Medicine, Dokkyo Medical University, Tochigi, Japan
- AETAS Pharma Co., Ltd., Tokyo, Japan
| | | | - Godfrey Smith
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Ryuko Matsuda
- AETAS Pharma Co., Ltd., Tokyo, Japan
- Nojima Hospital, Tottori, Japan
| | | | - Patric B Mark
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | | | | | - Naoyuki Otani
- Dokkyo Medical University Nikko Medical Center, Tochigi, Japan
| | - Scott Kayley
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Ana Da Silva Costa
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Tamara P Martin
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Sara Dobi
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Priyanka Saxena
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Ken Shimamoto
- Division of Cardiovascular Medicine, Sendai Cardiovascular Center, Miyagi, Japan
| | - Tetsuya Ishikawa
- Department of Cardiology, Dokkyo Medical University, Saitama Medical Center, Saitama, Japan
| | - Ryuichi Kambayashi
- Department of Pharmacology, Faculty of Medicine, Toho University, Tokyo, Japan
| | - Alexandra Riddell
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Elspeth B Elliott
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | | | | | | | - Sayuri Hirano
- Process Research & Development Laboratories Technology Research & Development Division, Sumitomo Dainippon Pharma Co. Ltd., Osaka, Japan
| | - Takeshi Kitai
- Department of Heart Failure and Transplantation, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Kengo Kusano
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yuko Inoue
- Department of Heart Failure and Transplantation, National Cerebral and Cardiovascular Center, Osaka, Japan
| | | | - Migaku Kikuchi
- Department of Cardiovascular Medicine, Dokkyo Medical University, Tochigi, Japan
| | - Shigeru Toyoda
- Department of Cardiovascular Medicine, Dokkyo Medical University, Tochigi, Japan
| | - Isao Taguchi
- Department of Cardiology, Dokkyo Medical University, Saitama Medical Center, Saitama, Japan
| | | | - Atsushi Sugiyama
- Department of Pharmacology, Faculty of Medicine, Toho University, Tokyo, Japan
| | | | | |
Collapse
|
4
|
Starnes L, Hall A, Etal D, Cavallo AL, Grabowski P, Gallon J, Kha M, Hicks R, Pointon A. RYR2 deficient human model identifies calcium handling and metabolic dysfunction impacting pharmacological responses. Front Cardiovasc Med 2024; 11:1357315. [PMID: 39041002 PMCID: PMC11260679 DOI: 10.3389/fcvm.2024.1357315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 06/18/2024] [Indexed: 07/24/2024] Open
Abstract
Creation of disease models utilizing hiPSCs in combination with CRISPR/Cas9 gene editing enable mechanistic insights into differential pharmacological responses. This allows translation of efficacy and safety findings from a healthy to a diseased state and provides a means to predict clinical outcome sooner during drug discovery. Calcium handling disturbances including reduced expression levels of the type 2 ryanodine receptor (RYR2) are linked to cardiac dysfunction; here we have created a RYR2 deficient human cardiomyocyte model that mimics some aspects of heart failure. RYR2 deficient cardiomyocytes show differential pharmacological responses to L-type channel calcium inhibitors. Phenotypic and proteomic characterization reveal novel molecular insights with altered expression of structural proteins including CSRP3, SLMAP, and metabolic changes including upregulation of the pentose phosphate pathway and increased sensitivity to redox alterations. This genetically engineered in vitro cardiovascular model of RYR2 deficiency supports the study of pharmacological responses in the context of calcium handling and metabolic dysfunction enabling translation of drug responses from healthy to perturbed cellular states.
Collapse
Affiliation(s)
- Linda Starnes
- Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Andrew Hall
- Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, United Kingdom
| | - Damla Etal
- Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Piotr Grabowski
- Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, United Kingdom
| | - John Gallon
- Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, United Kingdom
| | - Michelle Kha
- Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Ryan Hicks
- BioPharmaceuticals R&D Cell Therapy Department, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- School of Cardiovascular and Metabolic Medicine & Sciences, King’s College London, London, United Kingdom
| | - Amy Pointon
- Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, United Kingdom
| |
Collapse
|
5
|
Taweechat P, Boonamnaj P, Samsó M, Sompornpisut P. Significance of Zn 2+ in RyR1 for Structural Integrity and Ligand Binding: Insight from Molecular Dynamics. J Phys Chem B 2024; 128:4670-4684. [PMID: 38717304 PMCID: PMC11103704 DOI: 10.1021/acs.jpcb.4c01189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/22/2024]
Abstract
Ryanodine receptor type 1 (RyR1) is a Ca2+-release channel central to skeletal muscle excitation-contraction (EC) coupling. RyR1's cryo-EM structures reveal a zinc-finger motif positioned within the cytoplasmic C-terminal domain (CTD). Yet, owing to limitations in cryo-EM resolution, RyR1 structures lack precision in detailing the metal coordination structure, prompting the need for an accurate model. In this study, we employed molecular dynamics (MD) simulations and the density functional theory (DFT) method to refine the binding characteristics of Zn2+ in the zinc-finger site of the RyR1 channel. Our findings also highlight substantial conformational changes in simulations conducted in the absence of Zn2+. Notably, we observed a loss of contact at the interface between protein domains proximal to the zinc-finger site, indicating a crucial role of Zn2+ in maintaining structural integrity and interdomain interactions within RyR1. Furthermore, this study provides valuable insights into the modulation of ATP, Ca2+, and caffeine binding, shedding light on the intricate relationship between Zn2+ coordination and the dynamic behavior of RyR1. Our integrative approach combining MD simulations and DFT calculations enhances our understanding of the molecular mechanisms governing ligand binding in RyR1.
Collapse
Affiliation(s)
- Panyakorn Taweechat
- Center
of Excellence in Computational Chemistry, Department of Chemistry,
Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Panisak Boonamnaj
- Center
of Excellence in Computational Chemistry, Department of Chemistry,
Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Montserrat Samsó
- Department
of Physiology and Biophysics, Virginia Commonwealth
University, Richmond, Virginia 23298, United States
| | - Pornthep Sompornpisut
- Center
of Excellence in Computational Chemistry, Department of Chemistry,
Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
6
|
Reisqs JB, Qu YS, Boutjdir M. Ion channel trafficking implications in heart failure. Front Cardiovasc Med 2024; 11:1351496. [PMID: 38420267 PMCID: PMC10899472 DOI: 10.3389/fcvm.2024.1351496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 01/25/2024] [Indexed: 03/02/2024] Open
Abstract
Heart failure (HF) is recognized as an epidemic in the contemporary world, impacting around 1%-2% of the adult population and affecting around 6 million Americans. HF remains a major cause of mortality, morbidity, and poor quality of life. Several therapies are used to treat HF and improve the survival of patients; however, despite these substantial improvements in treating HF, the incidence of HF is increasing rapidly, posing a significant burden to human health. The total cost of care for HF is USD 69.8 billion in 2023, warranting a better understanding of the mechanisms involved in HF. Among the most serious manifestations associated with HF is arrhythmia due to the electrophysiological changes within the cardiomyocyte. Among these electrophysiological changes, disruptions in sodium and potassium currents' function and trafficking, as well as calcium handling, all of which impact arrhythmia in HF. The mechanisms responsible for the trafficking, anchoring, organization, and recycling of ion channels at the plasma membrane seem to be significant contributors to ion channels dysfunction in HF. Variants, microtubule alterations, or disturbances of anchoring proteins lead to ion channel trafficking defects and the alteration of the cardiomyocyte's electrophysiology. Understanding the mechanisms of ion channels trafficking could provide new therapeutic approaches for the treatment of HF. This review provides an overview of the recent advances in ion channel trafficking in HF.
Collapse
Affiliation(s)
- Jean-Baptiste Reisqs
- Cardiovascular Research Program, VA New York Harbor Healthcare System, New York, NY, United States
| | - Yongxia Sarah Qu
- Cardiovascular Research Program, VA New York Harbor Healthcare System, New York, NY, United States
- Department of Cardiology, New York Presbyterian Brooklyn Methodist Hospital, New York, NY, United States
| | - Mohamed Boutjdir
- Cardiovascular Research Program, VA New York Harbor Healthcare System, New York, NY, United States
- Department of Medicine, Cell Biology and Pharmacology, State University of New York Downstate Health Sciences University, New York, NY, United States
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
7
|
Schreckenberg R, Woitasky N, Itani N, Czech L, Ferdinandy P, Schulz R. Cardiac side effects of RNA-based SARS-CoV-2 vaccines: Hidden cardiotoxic effects of mRNA-1273 and BNT162b2 on ventricular myocyte function and structure. Br J Pharmacol 2024; 181:345-361. [PMID: 37828636 DOI: 10.1111/bph.16262] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/16/2023] [Accepted: 08/03/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND AND PURPOSE To protect against SARS-CoV-2 infection, the first mRNA-based vaccines, Spikevax (mRNA-1273, Moderna) and Comirnaty (BNT162b2, Pfizer/Biontech), were approved in 2020. The structure and assembly of the immunogen-in both cases, the SARS-CoV-2 spike (S) glycoprotein-are determined by a messenger RNA sequence that is translated by endogenous ribosomes. Cardiac side-effects, which for the most part can be classified by their clinical symptoms as myo- and/or pericarditis, can be caused by both mRNA-1273 and BNT162b2. EXPERIMENTAL APPROACH As persuasive theories for the underlying pathomechanisms have yet to be developed, this study investigated the effect of mRNA-1273 and BNT162b2 on the function, structure, and viability of isolated adult rat cardiomyocytes over a 72 h period. KEY RESULTS In the first 24 h after application, both mRNA-1273 and BNT162b2 caused neither functional disturbances nor morphological abnormalities. After 48 h, expression of the encoded spike protein was detected in ventricular cardiomyocytes for both mRNAs. At this point in time, mRNA-1273 induced arrhythmic as well as completely irregular contractions associated with irregular as well as localized calcium transients, which provide indications of significant dysfunction of the cardiac ryanodine receptor (RyR2). In contrast, BNT162b2 increased cardiomyocyte contraction via significantly increased protein kinase A (PKA) activity at the cellular level. CONCLUSION AND IMPLICATIONS Here, we demonstrated for the first time, that in isolated cardiomyocytes, both mRNA-1273 and BNT162b2 induce specific dysfunctions that correlate pathophysiologically to cardiomyopathy. Both RyR2 impairment and sustained PKA activation may significantly increase the risk of acute cardiac events.
Collapse
Affiliation(s)
- Rolf Schreckenberg
- Institute of Physiology, Faculty of Medicine, Justus-Liebig University, Gießen, Germany
| | - Nadine Woitasky
- Institute of Physiology, Faculty of Medicine, Justus-Liebig University, Gießen, Germany
| | - Nadja Itani
- Institute of Physiology, Faculty of Medicine, Justus-Liebig University, Gießen, Germany
| | - Laureen Czech
- Institute of Physiology, Faculty of Medicine, Justus-Liebig University, Gießen, Germany
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, National Heart Laboratory, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Rainer Schulz
- Institute of Physiology, Faculty of Medicine, Justus-Liebig University, Gießen, Germany
| |
Collapse
|
8
|
Telles TM, May BM, Pimentel M, Pereira BLDS, Andrades M, Rohde LE, Dos Santos KG. Non‑synonymous polymorphisms in the HRC and ADRB1 genes may be associated with all‑cause death in patients with non‑ischemic heart failure. Exp Ther Med 2024; 27:48. [PMID: 38144921 PMCID: PMC10739235 DOI: 10.3892/etm.2023.12337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 11/07/2023] [Indexed: 12/26/2023] Open
Abstract
Sudden cardiac death (SCD) is an unpredictable and common mode of death in patients with heart failure (HF). Alterations in calcium handling may lead to malignant arrhythmias, resulting in SCD, and variants in calcium signaling-related genes have a significant association with SCD. Therefore, the aim of the present retrospective cohort study was to investigate the association of Ser96Ala [histidine-rich calcium-binding protein (HRC)], Ser49Gly [β1-adrenergic receptor (ADRB1)], Arg389Gly (ADRB1) and Gly1886Ser [ryanodine receptor 2 (RYR2)] polymorphisms with serious arrhythmic events and overall mortality in patients with HF with reduced left ventricular ejection fraction of non-ischemic etiology. In total, 136 patients with HF underwent physical examination, routine laboratory tests, non-invasive assessment of cardiac function and an invasive electrophysiological study. The primary outcome was the occurrence of serious arrhythmic events, set as either SCD or appropriate implantable cardioverter-defibrillator (ICD) therapy, and the secondary outcome was all-cause death. During a median follow-up of 37 months, arrhythmic events occurred in 26 patients (19%) and 41 patients (30%) died. Patients carrying the Ser allele of the Ser96Ala polymorphism in HRC had worse survival than those with the Ala/Ala genotype (log-rank P=0.043). Despite the difference in survival time, the Ala/Ala genotype was not associated with all-cause death in the regression analysis [unadjusted hazard ratio (HR)=0.17; 95% CI, 0.02-1.21]. Regarding the Ser49Gly and Arg389Gly polymorphisms in ADRB1, homozygosity for the major alleles at both sites (Ser49Ser and Arg389Arg) was associated with a two-fold increased risk of all-cause death compared with the other genotype combinations (unadjusted HR=1.98; 95% CI, 1.02-3.82). However, this association was lost after controlling for clinical covariates. No association was observed for the Gly1886Ser polymorphism in RYR2. Overall, the present findings are concurrent with the hypothesis that the Ser96Ala (HRC), Ser49Gly (ADRB1) and Arg389Gly (ADRB1) polymorphisms may be associated with HF prognosis. In particular, the Ser96Ala polymorphism might aid in risk stratification and patient selection for ICD implantation.
Collapse
Affiliation(s)
- Tanise Machado Telles
- Laboratory of Human Molecular Genetics, Lutheran University of Brazil, Canoas, Rio Grande do Sul 92425-900, Brazil
| | - Bruna Miers May
- Cardiology Division, Clinical Hospital of Porto Alegre, Porto Alegre, Rio Grande do Sul 90035-903, Brazil
| | - Mauricio Pimentel
- Cardiology Division, Clinical Hospital of Porto Alegre, Porto Alegre, Rio Grande do Sul 90035-903, Brazil
| | - Bruna Letícia Da Silva Pereira
- Cells, Tissues and Genes Laboratory, Clinical Hospital of Porto Alegre, Porto Alegre, Rio Grande do Sul 90035-903, Brazil
| | - Michael Andrades
- Cardiovascular Research Laboratory, Clinical Hospital of Porto Alegre, Porto Alegre, Rio Grande do Sul 90035-903, Brazil
| | - Luis Eduardo Rohde
- Cardiology Division, Clinical Hospital of Porto Alegre, Porto Alegre, Rio Grande do Sul 90035-903, Brazil
- Department of Internal Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90035-003, Brazil
| | - Kátia Gonçalves Dos Santos
- Laboratory of Human Molecular Genetics, Lutheran University of Brazil, Canoas, Rio Grande do Sul 92425-900, Brazil
| |
Collapse
|
9
|
Kourampi I, Katsioupa M, Oikonomou E, Tsigkou V, Marinos G, Goliopoulou A, Katsarou O, Kalogeras K, Theofilis P, Tsatsaragkou A, Siasos G, Tousoulis D, Vavuranakis M. The Role of Ranolazine in Heart Failure-Current Concepts. Am J Cardiol 2023; 209:92-103. [PMID: 37844876 DOI: 10.1016/j.amjcard.2023.09.066] [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: 07/08/2023] [Revised: 09/09/2023] [Accepted: 09/15/2023] [Indexed: 10/18/2023]
Abstract
Heart failure is a complex clinical syndrome with a detrimental impact on mortality and morbidity. Energy substrate utilization and myocardial ion channel regulation have gained research interest especially after the introduction of sodium-glucose co-transporter 2 inhibitors in the treatment of heart failure. Ranolazine or N-(2,6-dimethylphenyl)-2-(4-[2-hydroxy-3-(2-methoxyphenoxy) propyl] piperazin-1-yl) acetamide hydrochloride is an active piperazine derivative which inhibits late sodium current thus minimizing calcium overload in the ischemic cardiomyocytes. Ranolazine also prevents fatty acid oxidation and favors glycose utilization ameliorating the "energy starvation" of the failing heart. Heart failure with preserved ejection fraction is characterized by diastolic impairment; according to the literature ranolazine could be beneficial in the management of increased left ventricular end-diastolic pressure, right ventricular systolic dysfunction and wall shear stress which is reflected by the high natriuretic peptides. Fewer data is evident regarding the effects of ranolazine in heart failure with reduced ejection fraction and mainly support the control of the sodium-calcium exchanger and function of sarcoendoplasmic reticulum calcium adenosine triphosphatase. Ranolazine's therapeutic mechanisms in myocardial ion channels and energy utilization are documented in patients with chronic coronary syndromes. Nevertheless, ranolazine might have a broader effect in the therapy of heart failure and further mechanistic research is required.
Collapse
Affiliation(s)
- Islam Kourampi
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Maria Katsioupa
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece.
| | - Vasiliki Tsigkou
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Georgios Marinos
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Athina Goliopoulou
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Ourania Katsarou
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Konstantinos Kalogeras
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Panagiotis Theofilis
- 1st Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Aikaterini Tsatsaragkou
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece; Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston Massachusetts
| | - Dimitris Tousoulis
- 1st Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Manolis Vavuranakis
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| |
Collapse
|
10
|
Takenaka M, Kodama M, Murayama T, Ishigami-Yuasa M, Mori S, Ishida R, Suzuki J, Kanemaru K, Sugihara M, Iino M, Miura A, Nishio H, Morimoto S, Kagechika H, Sakurai T, Kurebayashi N. Screening for Novel Type 2 Ryanodine Receptor Inhibitors by Endoplasmic Reticulum Ca 2+ Monitoring. Mol Pharmacol 2023; 104:275-286. [PMID: 37678938 DOI: 10.1124/molpharm.123.000720] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/21/2023] [Accepted: 08/29/2023] [Indexed: 09/09/2023] Open
Abstract
Type 2 ryanodine receptor (RyR2) is a Ca2+ release channel on the endoplasmic (ER)/sarcoplasmic reticulum that plays a central role in the excitation-contraction coupling in the heart. Hyperactivity of RyR2 has been linked to ventricular arrhythmias in patients with catecholaminergic polymorphic ventricular tachycardia and heart failure, where spontaneous Ca2+ release via hyperactivated RyR2 depolarizes diastolic membrane potential to induce triggered activity. In such cases, drugs that suppress RyR2 activity are expected to prevent the arrhythmias, but there is no clinically available RyR2 inhibitors at present. In this study, we searched for RyR2 inhibitors from a well-characterized compound library using a recently developed ER Ca2+-based assay, where the inhibition of RyR2 activity was detected by the increase in ER Ca2+ signals from R-CEPIA1er, a genetically encoded ER Ca2+ indicator, in RyR2-expressing HEK293 cells. By screening 1535 compounds in the library, we identified three compounds (chloroxylenol, methyl orsellinate, and riluzole) that greatly increased the ER Ca2+ signal. All of the three compounds suppressed spontaneous Ca2+ oscillations in RyR2-expressing HEK293 cells and correspondingly reduced the Ca2+-dependent [3H]ryanodine binding activity. In cardiomyocytes from RyR2-mutant mice, the three compounds effectively suppressed abnormal Ca2+ waves without substantial effects on the action-potential-induced Ca2+ transients. These results confirm that ER Ca2+-based screening is useful for identifying modulators of ER Ca2+ release channels and suggest that RyR2 inhibitors have potential to be developed as a new category of antiarrhythmic drugs. SIGNIFICANCE STATEMENT: We successfully identified three compounds having RyR2 inhibitory action from a well-characterized compound library using an endoplasmic reticulum Ca2+-based assay, and demonstrated that these compounds suppressed arrhythmogenic Ca2+ wave generation without substantially affecting physiological action-potential induced Ca2+ transients in cardiomyocytes. This study will facilitate the development of RyR2-specific inhibitors as a potential new class of drugs for life-threatening arrhythmias induced by hyperactivation of RyR2.
Collapse
Affiliation(s)
- Mai Takenaka
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Masami Kodama
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Takashi Murayama
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Mari Ishigami-Yuasa
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Shuichi Mori
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Ryosuke Ishida
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Junji Suzuki
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Kazunori Kanemaru
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Masami Sugihara
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Masamitsu Iino
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Aya Miura
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Hajime Nishio
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Sachio Morimoto
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Hiroyuki Kagechika
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Takashi Sakurai
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Nagomi Kurebayashi
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| |
Collapse
|
11
|
Kiyomoto K, Matsuo I, Suita K, Ohnuki Y, Ishikawa M, Ito A, Mototani Y, Tsunoda M, Morii A, Nariyama M, Hayakawa Y, Amitani Y, Gomi K, Okumura S. Oral angiotensin-converting enzyme inhibitor captopril protects the heart from Porphyromonas gingivalis LPS-induced cardiac dysfunction in mice. PLoS One 2023; 18:e0292624. [PMID: 37983238 PMCID: PMC10659197 DOI: 10.1371/journal.pone.0292624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 09/25/2023] [Indexed: 11/22/2023] Open
Abstract
Although angiotensin converting enzyme (ACE) inhibitors are considered useful for the treatment of human heart failure, some experimental failing-heart models have shown little beneficial effect of ACE inhibitors in animals with poor oral health, particularly periodontitis. In this study, we examined the effects of the ACE inhibitor captopril (Cap; 0.1 mg/mL in drinking water) on cardiac dysfunction in mice treated with Porphyromonas gingivalis lipopolysaccharide (PG-LPS) at a dose (0.8 mg/kg/day) equivalent to the circulating level in patients with periodontal disease. Mice were divided into four groups: 1) Control, 2) PG-LPS, 3) Cap, and 4) PG-LPS + Cap. After1 week, we evaluated cardiac function by echocardiography. The left ventricular ejection fraction was significantly decreased in PG-LPS-treated mice compared to the control (from 66 ± 1.8 to 59 ± 2.5%), while Cap ameliorated the dysfunction (63 ± 1.1%). The area of cardiac fibrosis was significantly increased (approximately 2.9-fold) and the number of apoptotic myocytes was significantly increased (approximately 5.6-fold) in the heart of PG-LPS-treated group versus the control, and these changes were suppressed by Cap. The impairment of cardiac function in PG-LPS-treated mice was associated with protein kinase C δ phosphorylation (Tyr-311), leading to upregulation of NADPH oxidase 4 and xanthine oxidase, and calmodulin kinase II phosphorylation (Thr-286) with increased phospholamban phosphorylation (Thr-17). These changes were also suppressed by Cap. Our results suggest that the renin-angiotensin system might play an important role in the development of cardiac diseases induced by PG-LPS.
Collapse
Affiliation(s)
- Kenichi Kiyomoto
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Ichiro Matsuo
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Kenji Suita
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshiki Ohnuki
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Misao Ishikawa
- Department of Oral Anatomy, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Aiko Ito
- Department of Orthodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yasumasa Mototani
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Michinori Tsunoda
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Akinaka Morii
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Megumi Nariyama
- Department of Pediatric Dentistry, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshio Hayakawa
- Department of Dental Anesthesiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yasuharu Amitani
- Department of Mathematics, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Kazuhiro Gomi
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Satoshi Okumura
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| |
Collapse
|
12
|
Ito A, Ohnuki Y, Suita K, Matsuo I, Ishikawa M, Mitsubayashi T, Mototani Y, Kiyomoto K, Tsunoda M, Morii A, Nariyama M, Hayakawa Y, Tomonari H, Okumura S. Effects of the angiotensin-converting enzyme inhibitor captopril on occlusal-disharmony-induced cardiac dysfunction in mice. Sci Rep 2023; 13:19927. [PMID: 37968296 PMCID: PMC10651878 DOI: 10.1038/s41598-023-43099-6] [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: 11/23/2022] [Accepted: 09/19/2023] [Indexed: 11/17/2023] Open
Abstract
Occlusal disharmony is known to affect not only the oral cavity environment, but also the autonomic nervous system in the heart. Since the renin-angiotensin system (RAS) inhibitor captopril (Cap) is one of the first-line drugs for preventing cardiac remodeling in patients with heart failure, we hypothesized that Cap might prevent cardiac dysfunction induced by occlusal disharmony. Here, to test this idea, we used our bite-opening (BO) mouse model, which was developed by cementing a suitable appliance onto the mandibular incisor. Mice were divided into four groups: (1) Control, (2) BO, (3) Cap, and (4) BO + Cap. After 2 weeks, we evaluated cardiac function by echocardiography and confirmed that cardiac function was significantly decreased in the BO group compared to the control, while Cap ameliorated the dysfunction. Cardiac fibrosis, myocyte apoptosis and oxidative stress-induced myocardial damage in the BO group were significantly increased versus the control, and these increases were suppressed by Cap. Cardiac dysfunction induced by BO was associated with dual phosphorylation on PKCδ (Tyr-311/Thr-505), leading to activation of CaMKII with increased phosphorylation of RyR2 and phospholamban. Our results suggest that the RAS might play an important role in the development of cardiac diseases induced by occlusal anomalies.
Collapse
Affiliation(s)
- Aiko Ito
- Department of Orthodontics, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Yoshiki Ohnuki
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
| | - Kenji Suita
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
| | - Ichiro Matsuo
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Misao Ishikawa
- Department of Oral Anatomy, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Takao Mitsubayashi
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
| | - Yasumasa Mototani
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
| | - Kenichi Kiyomoto
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Michinori Tsunoda
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Akinaka Morii
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Megumi Nariyama
- Department of Pediatric Dentistry, Tsurumi University School of Dental Medicine, Yokohama, 236-8501, Japan
| | - Yoshio Hayakawa
- Department of Dental Anesthesiology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Hiroshi Tomonari
- Department of Orthodontics, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Satoshi Okumura
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan.
| |
Collapse
|
13
|
Dries E, Gilbert G, Roderick HL, Sipido KR. The ryanodine receptor microdomain in cardiomyocytes. Cell Calcium 2023; 114:102769. [PMID: 37390591 DOI: 10.1016/j.ceca.2023.102769] [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: 05/02/2023] [Revised: 06/11/2023] [Accepted: 06/12/2023] [Indexed: 07/02/2023]
Abstract
The ryanodine receptor type 2 (RyR) is a key player in Ca2+ handling during excitation-contraction coupling. During each heartbeat, RyR channels are responsible for linking the action potential with the contractile machinery of the cardiomyocyte by releasing Ca2+ from the sarcoplasmic reticulum. RyR function is fine-tuned by associated signalling molecules, arrangement in clusters and subcellular localization. These parameters together define RyR function within microdomains and are subject to disease remodelling. This review describes the latest findings on RyR microdomain organization, the alterations with disease which result in increased subcellular heterogeneity and emergence of microdomains with enhanced arrhythmogenic potential, and presents novel technologies that guide future research to study and target RyR channels within specific microdomains.
Collapse
Affiliation(s)
- Eef Dries
- Lab of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium.
| | - Guillaume Gilbert
- Lab of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Laboratoire ORPHY EA 4324, Université de Brest, Brest, France
| | - H Llewelyn Roderick
- Lab of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Karin R Sipido
- Lab of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
14
|
Liu T, Wu J, Shi S, Cui B, Xiong F, Yang S, Yan M. Dapagliflozin attenuates cardiac remodeling and dysfunction in rats with β-adrenergic receptor overactivation through restoring calcium handling and suppressing cardiomyocyte apoptosis. Diab Vasc Dis Res 2023; 20:14791641231197106. [PMID: 37589258 PMCID: PMC10437211 DOI: 10.1177/14791641231197106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/18/2023] Open
Abstract
Background: Long-term β-adrenergic receptor (β-AR) activation can impair myocardial structure and function. Dapagliflozin (DAPA) has been reported to improve clinical prognosis in heart failure patients, whereas the exact mechanism remains unclear. Here, we investigated the effects of DAPA against β-AR overactivation toxicity and explored the underlying mechanism.Methods and Results: Rats were randomized to receive saline + placebo, isoproterenol (ISO, 5 mg/kg/day, intraperitoneally) + placebo, or ISO + DAPA (1 mg/kg/day, intragastrically) for 2-week. DAPA treatment improved cardiac function, alleviated myocardial fibrosis, prevented cardiomyocytes (CMs) apoptosis, and decreased the expression of ER stress-mediated apoptosis markers in ISO-treated hearts. In isolated CMs, 2-week ISO stimulation resulted in deteriorated kinetics of cellular contraction and relaxation, increased diastolic intracellular Ca2+ level and decay time constant of Ca2+ transient (CaT) but decreased CaT amplitude and sarcoplasmic reticulum (SR) Ca2+ level. However, DAPA treatment prevented abnormal Ca2+ handling and contractile dysfunction in CMs from ISO-treated hearts. Consistently, DAPA treatment upregulated the expression of SR Ca2+-ATPase protein and ryanodine receptor 2 (RyR2) but reduced the expression of phosphorylated-RyR2, Ca2+/calmodulin-dependent protein kinase II (CaMKII), and phosphorylated-CaMKII in ventricles from ISO-treated rats.Conclusion: DAPA prevented myocardial remodeling and cardiac dysfunction in rats with β-AR overactivation via restoring calcium handling and suppressing ER stress-related CMs apoptosis.
Collapse
Affiliation(s)
- Tao Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jinchun Wu
- Department of Cardiology, Qinghai Provincial People's Hospital, Xining, China
| | - Shaobo Shi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Bo Cui
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Feng Xiong
- Montreal Heart Institute (MHI), Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Shuang Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Min Yan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
15
|
MacLeod KT. Changes in cellular Ca 2+ and Na + regulation during the progression towards heart failure. J Physiol 2023; 601:905-921. [PMID: 35946572 PMCID: PMC10952717 DOI: 10.1113/jp283082] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/02/2022] [Indexed: 11/08/2022] Open
Abstract
In adapting to disease and loss of tissue, the heart shows great phenotypic plasticity that involves changes to its structure, composition and electrophysiology. Together with parallel whole body cardiovascular adaptations, the initial decline in cardiac function resulting from the insult is compensated. However, in the long term, the heart muscle begins to fail and patients with this condition have a very poor prognosis, with many dying from disturbances of rhythm. The surviving myocytes of these hearts gain Na+ , which is positively inotropic because of alterations to Ca2+ fluxes mediated by the Na+ /Ca2+ exchange, but compromises Ca2+ -dependent energy metabolism in mitochondria. Uptake of Ca2+ into the sarcoplasmic reticulum (SR) is reduced because of diminished function of SR Ca2+ ATPases. The result of increased Ca2+ influx and reduced SR Ca2+ uptake is an increase in the diastolic cytosolic Ca2+ concentration, which promotes spontaneous SR Ca2+ release and induces delayed afterdepolarisations. Action potential duration prolongs because of increased late Na+ current and changes in expression and function of other ion channels and transporters increasing the probability of the formation of early afterdepolarisations. There is a reduction in T-tubule density and so the normal spatial arrangements required for efficient excitation-contraction coupling are compromised and lead to temporal delays in Ca2+ release from the SR. Therefore, the structural and electrophysiological responses that occur to provide compensation do so at the expense of (1) increasing the likelihood of arrhythmogenesis; (2) activating hypertrophic, apoptotic and Ca2+ signalling pathways; and (3) decreasing the efficiency of SR Ca2+ release.
Collapse
Affiliation(s)
- Kenneth T. MacLeod
- National Heart & Lung InstituteImperial Centre for Translational and Experimental MedicineImperial CollegeHammersmith HospitalLondonUK
| |
Collapse
|
16
|
Wang YX, Reyes-García J, Di Mise A, Zheng YM. Role of ryanodine receptor 2 and FK506-binding protein 12.6 dissociation in pulmonary hypertension. J Gen Physiol 2023; 155:213798. [PMID: 36625865 PMCID: PMC9836826 DOI: 10.1085/jgp.202213100] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 07/29/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Pulmonary hypertension (PH) is a devastating disease characterized by a progressive increase in pulmonary arterial pressure leading to right ventricular failure and death. A major cellular response in this disease is the contraction of smooth muscle cells (SMCs) of the pulmonary vasculature. Cell contraction is determined by the increase in intracellular Ca2+ concentration ([Ca2+]i), which is generated and regulated by various ion channels. Several studies by us and others have shown that ryanodine receptor 2 (RyR2), a Ca2+-releasing channel in the sarcoplasmic reticulum (SR), is an essential ion channel for the control of [Ca2+]i in pulmonary artery SMCs (PASMCs), thereby mediating the sustained vasoconstriction seen in PH. FK506-binding protein 12.6 (FKBP12.6) strongly associates with RyR2 to stabilize its functional activity. FKBP12.6 can be dissociated from RyR2 by a hypoxic stimulus to increase channel function and Ca2+ release, leading to pulmonary vasoconstriction and PH. More specifically, dissociation of the RyR2-FKBP12.6 complex is a consequence of increased mitochondrial ROS generation mediated by the Rieske iron-sulfur protein (RISP) at the mitochondrial complex III after hypoxia. Overall, RyR2/FKBP12.6 dissociation and the corresponding signaling pathway may be an important factor in the development of PH. Novel drugs and biologics targeting RyR2, FKBP12.6, and related molecules may become unique effective therapeutics for PH.
Collapse
Affiliation(s)
- Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA,Correspondence to Yong-Xiao Wang:
| | - Jorge Reyes-García
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA,Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México,Ciudad de México, México
| | - Annarita Di Mise
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA,Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Yun-Min Zheng
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA,Yun-Min Zheng:
| |
Collapse
|
17
|
Yan M, Liu T, Zhong P, Xiong F, Cui B, Wu J, Wu G. Chronic catestatin treatment reduces atrial fibrillation susceptibility via improving calcium handling in post-infarction heart failure rats. Peptides 2023; 159:170904. [PMID: 36375660 DOI: 10.1016/j.peptides.2022.170904] [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: 08/06/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND Abnormal Ca2+ handling is a pivotal element of atrial fibrillation (AF) substrates. Catestatin (CST) modulates intracellular Ca2+ handling in cardiomyocytes (CMs). We investigated the effects of CST administration on atrial Ca2+ handling and AF susceptibility in rats with post-infarction heart failure (HF). METHODS Myocardial infarction (MI) was established by ligation of the left anterior descending coronary artery in rats. Two-week later, rats with post-infarction HF were randomly treated with saline (MI group) or CST (MI + CST group) for 4-week. Cellular Ca2+ imaging was performed by incubating atrial CMs with Fura-2 AM. An in vitro electrophysiological study was performed to assess the vulnerability to action potential duration (APD) alternans and AF. Ca2+ handling proteins expression was determined using western blotting. RESULTS In atrial CMs, compared with the sham group, the sarcoplasmic reticulum (SR) Ca2+ load, Ca2+ transient (CaT) amplitude, and threshold for Ca2+ alternans were significantly decreased, but the diastolic intracellular Ca2+ level, SR Ca2+ leakage, and spontaneous Ca2+ events were markedly increased in the MI group. However, CST attenuated these Ca2+-handling abnormalities induced by post-infarction HF. Moreover, vulnerability to atrial APD alternans and AF was significantly increased in isolated hearts from the MI group compared to the sham group, whereas all effects were prevented by CST. CST treatment also preserved SR Ca2+-ATPase protein expression but decreased the protein levels of phosphorylated-ryanodine receptor 2 and phosphorylated-Ca2+/calmodulin-dependent protein kinase II in atria from post-infarction HF rats. CONCLUSION Chronic CST treatment reduces AF vulnerability in rats with MI-induced HF by improving Ca2+ handling.
Collapse
Affiliation(s)
- Min Yan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Tao Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China.
| | - Peng Zhong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China.
| | - Feng Xiong
- Montreal Heart Institute, Department of Medicine, University of Montreal, Montreal H1T 1C8, Quebec, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal H3G 1Y6, Quebec, Canada
| | - Bo Cui
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Jinchun Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Gang Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China.
| |
Collapse
|
18
|
Parra-Lucares A, Romero-Hernández E, Villa E, Weitz-Muñoz S, Vizcarra G, Reyes M, Vergara D, Bustamante S, Llancaqueo M, Toro L. New Opportunities in Heart Failure with Preserved Ejection Fraction: From Bench to Bedside… and Back. Biomedicines 2022; 11:70. [PMID: 36672578 PMCID: PMC9856156 DOI: 10.3390/biomedicines11010070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/07/2022] [Accepted: 12/13/2022] [Indexed: 12/29/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a growing public health problem in nearly 50% of patients with heart failure. Therefore, research on new strategies for its diagnosis and management has become imperative in recent years. Few drugs have successfully improved clinical outcomes in this population. Therefore, numerous attempts are being made to find new pharmacological interventions that target the main mechanisms responsible for this disease. In recent years, pathological mechanisms such as cardiac fibrosis and inflammation, alterations in calcium handling, NO pathway disturbance, and neurohumoral or mechanic impairment have been evaluated as new pharmacological targets showing promising results in preliminary studies. This review aims to analyze the new strategies and mechanical devices, along with their initial results in pre-clinical and different phases of ongoing clinical trials for HFpEF patients. Understanding new mechanisms to generate interventions will allow us to create methods to prevent the adverse outcomes of this silent pandemic.
Collapse
Affiliation(s)
- Alfredo Parra-Lucares
- Critical Care Unit, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago 8380420, Chile
- MD PhD Program, Faculty of Medicine, Universidad de Chile, Santiago 8380420, Chile
| | - Esteban Romero-Hernández
- MD PhD Program, Faculty of Medicine, Universidad de Chile, Santiago 8380420, Chile
- Division of Internal Medicine, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago 8380420, Chile
| | - Eduardo Villa
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago 8380420, Chile
| | - Sebastián Weitz-Muñoz
- Division of Internal Medicine, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago 8380420, Chile
| | - Geovana Vizcarra
- Division of Internal Medicine, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago 8380420, Chile
| | - Martín Reyes
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago 8380420, Chile
| | - Diego Vergara
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago 8380420, Chile
| | - Sergio Bustamante
- Coronary Care Unit, Cardiovascular Department, Hospital Clínico Universidad de Chile, Santiago 8380420, Chile
| | - Marcelo Llancaqueo
- Coronary Care Unit, Cardiovascular Department, Hospital Clínico Universidad de Chile, Santiago 8380420, Chile
| | - Luis Toro
- Division of Nephrology, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago 8380420, Chile
- Centro de Investigación Clínica Avanzada, Hospital Clínico, Universidad de Chile, Santiago 8380420, Chile
| |
Collapse
|
19
|
Alomar FA, Tian C, Bidasee SR, Venn ZL, Schroder E, Palermo NY, AlShabeeb M, Edagwa BJ, Payne JJ, Bidasee KR. HIV-Tat Exacerbates the Actions of Atazanavir, Efavirenz, and Ritonavir on Cardiac Ryanodine Receptor (RyR2). Int J Mol Sci 2022; 24:ijms24010274. [PMID: 36613717 PMCID: PMC9820108 DOI: 10.3390/ijms24010274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/05/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
The incidence of sudden cardiac death (SCD) in people living with HIV infection (PLWH), especially those with inadequate viral suppression, is high and the reasons for this remain incompletely characterized. The timely opening and closing of type 2 ryanodine receptor (RyR2) is critical for ensuring rhythmic cardiac contraction-relaxation cycles, and the disruption of these processes can elicit Ca2+ waves, ventricular arrhythmias, and SCD. Herein, we show that the HIV protein Tat (HIV-Tat: 0-52 ng/mL) and therapeutic levels of the antiretroviral drugs atazanavir (ATV: 0-25,344 ng/mL), efavirenz (EFV: 0-11,376 ng/mL), and ritonavir (RTV: 0-25,956 ng/mL) bind to and modulate the opening and closing of RyR2. Abacavir (0-14,315 ng/mL), bictegravir (0-22,469 ng/mL), Rilpivirine (0-14,360 ng/mL), and tenofovir disoproxil fumarate (0-18,321 ng/mL) did not alter [3H]ryanodine binding to RyR2. Pretreating RyR2 with low HIV-Tat (14 ng/mL) potentiated the abilities of ATV and RTV to bind to open RyR2 and enhanced their ability to bind to EFV to close RyR2. In silico molecular docking using a Schrodinger Prime protein-protein docking algorithm identified three thermodynamically favored interacting sites for HIV-Tat on RyR2. The most favored site resides between amino acids (AA) 1702-1963; the second favored site resides between AA 467-1465, and the third site resides between AA 201-1816. Collectively, these new data show that HIV-Tat, ATV, EFV, and RTV can bind to and modulate the activity of RyR2 and that HIV-Tat can exacerbate the actions of ATV, EFV, and RTV on RyR2. Whether the modulation of RyR2 by these agents increases the risk of arrhythmias and SCD remains to be explored.
Collapse
Affiliation(s)
- Fadhel A. Alomar
- Department of Pharmacology and Toxicology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Chengju Tian
- Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sean R. Bidasee
- Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Zachary L. Venn
- Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Evan Schroder
- Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Nicholas Y. Palermo
- Vice Chancellor for Research Cores, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mohammad AlShabeeb
- Population Health Research Section, King Abdullah International Medical Research Center, King Saudi bin Abdulaziz University for Health Sciences, Riyadh 11426, Saudi Arabia
| | - Benson J. Edagwa
- Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jason J. Payne
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Keshore R. Bidasee
- Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Environment and Occupational Health, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Nebraska Redox Biology Center, Lincoln, NE 68588, USA
- Correspondence: ; Tel.: +402-559-9018; Fax: +402-559-7495
| |
Collapse
|
20
|
Wu J, Liu T, Shi S, Fan Z, Hiram R, Xiong F, Cui B, Su X, Chang R, Zhang W, Yan M, Tang Y, Huang H, Wu G, Huang C. Dapagliflozin reduces the vulnerability of rats with pulmonary arterial hypertension-induced right heart failure to ventricular arrhythmia by restoring calcium handling. Cardiovasc Diabetol 2022; 21:197. [PMID: 36171554 PMCID: PMC9516842 DOI: 10.1186/s12933-022-01614-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 09/01/2022] [Indexed: 12/20/2022] Open
Abstract
Background Malignant ventricular arrhythmia (VA) is a major contributor to sudden cardiac death (SCD) in patients with pulmonary arterial hypertension (PAH)-induced right heart failure (RHF). Recently, dapagliflozin (DAPA), a sodium/glucose cotransporter-2 inhibitor (SGLT2i), has been found to exhibit cardioprotective effects in patients with left ventricular systolic dysfunction. In this study, we examined the effects of DAPA on VA vulnerability in a rat model of PAH-induced RHF. Methods Rats randomly received monocrotaline (MCT, 60 mg/kg) or vehicle via a single intraperitoneal injection. A day later, MCT-injected rats were randomly treated with placebo, low-dose DAPA (1 mg/kg/day), or high-dose (3 mg/kg/day) DAPA orally for 35 days. Echocardiographic analysis, haemodynamic experiments, and histological assessments were subsequently performed to confirm the presence of PAH-induced RHF. Right ventricle (RV) expression of calcium (Ca2+) handling proteins were detected via Western blotting. RV expression of connexin 43 (Cx43) was determined via immunohistochemical staining. An optical mapping study was performed to assess the electrophysiological characteristics in isolated hearts. Cellular Ca2+ imaging from RV cardiomyocytes (RVCMs) was recorded using Fura-2 AM or Fluo-4 AM. Results High-dose DAPA treatment attenuated RV structural remodelling, improved RV function, alleviated Cx43 remodelling, increased the conduction velocity, restored the expression of key Ca2+ handling proteins, increased the threshold for Ca2+ and action potential duration (APD) alternans, decreased susceptibility to spatially discordant APD alternans and spontaneous Ca2+ events, promoted cellular Ca2+ handling, and reduced VA vulnerability in PAH-induced RHF rats. Low-dose DAPA treatment also showed antiarrhythmic effects in hearts with PAH-induced RHF, although with a lower level of efficacy. Conclusion DAPA administration reduced VA vulnerability in rats with PAH-induced RHF by improving RVCM Ca2+ handling. Supplementary Information The online version contains supplementary material available at 10.1186/s12933-022-01614-5.
Collapse
Affiliation(s)
- Jinchun Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Department of Cardiology, Qinghai Provincial People's Hospital, No.2 Gong He Road, Xining, 810007, People's Republic of China
| | - Tao Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China. .,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China. .,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.
| | - Shaobo Shi
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China
| | - Zhixing Fan
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China
| | - Roddy Hiram
- Department of Medicine, Faculty of Medicine, Montreal Heart Institute (MHI), Université de Montréal, Montreal, QC, Canada
| | - Feng Xiong
- Department of Medicine, Faculty of Medicine, Montreal Heart Institute (MHI), Université de Montréal, Montreal, QC, Canada
| | - Bo Cui
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China
| | - Xiaoling Su
- Department of Cardiology, Qinghai Provincial People's Hospital, No.2 Gong He Road, Xining, 810007, People's Republic of China
| | - Rong Chang
- Department of Cardiology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, No. 187 Guanlan Road, Longhua District, Shenzhen, 518109, China
| | - Wei Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China
| | - Min Yan
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China
| | - Gang Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China. .,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China. .,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China. .,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China. .,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.
| |
Collapse
|
21
|
Molecular, Subcellular, and Arrhythmogenic Mechanisms in Genetic RyR2 Disease. Biomolecules 2022; 12:biom12081030. [PMID: 35892340 PMCID: PMC9394283 DOI: 10.3390/biom12081030] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/22/2022] [Accepted: 07/24/2022] [Indexed: 11/17/2022] Open
Abstract
The ryanodine receptor (RyR2) has a critical role in controlling Ca2+ release from the sarcoplasmic reticulum (SR) throughout the cardiac cycle. RyR2 protein has multiple functional domains with specific roles, and four of these RyR2 protomers are required to form the quaternary structure that comprises the functional channel. Numerous mutations in the gene encoding RyR2 protein have been identified and many are linked to a wide spectrum of arrhythmic heart disease. Gain of function mutations (GoF) result in a hyperactive channel that causes excessive spontaneous SR Ca2+ release. This is the predominant cause of the inherited syndrome catecholaminergic polymorphic ventricular tachycardia (CPVT). Recently, rare hypoactive loss of function (LoF) mutations have been identified that produce atypical effects on cardiac Ca2+ handling that has been termed calcium release deficiency syndrome (CRDS). Aberrant Ca2+ release resulting from both GoF and LoF mutations can result in arrhythmias through the Na+/Ca2+ exchange mechanism. This mini-review discusses recent findings regarding the role of RyR2 domains and endogenous regulators that influence RyR2 gating normally and with GoF/LoF mutations. The arrhythmogenic consequences of GoF/LoF mutations will then be discussed at the macromolecular and cellular level.
Collapse
|
22
|
Wu J, Tan Y, Kang D, Yu J, Qi J, Wu J, Zhang M. Xiaoyu Jiangzhi capsule protects against heart failure via Ca2+/CaMKII signaling pathways in mice. JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2022. [DOI: 10.1016/j.jtcms.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
23
|
Groenendyk J, Wang WA, Robinson A, Michalak M. Calreticulin and the Heart. Cells 2022; 11:cells11111722. [PMID: 35681417 PMCID: PMC9179554 DOI: 10.3390/cells11111722] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 11/30/2022] Open
Abstract
Calreticulin is an endoplasmic Ca2+ binding protein and molecular chaperone. As a cardiac embryonic gene, calreticulin is essential for heart development. The protein supports Ca2+-dependent signaling events that are critical to cardiomyocyte differentiation and cardiogenesis. The increased expression of calreticulin and endoplasmic reticulum/sarcoplasmic reticulum Ca2+ capacity produces cardiomyocytes with enhanced efficiency, and detrimental mechanical stretching of cardiac fibroblasts, leading to cardiac pathology. Deletion of the calreticulin gene in adult cardiomyocytes results in left ventricle dilation, an impaired electrocardiogram, and heart failure. These observations indicate that a well-adjusted endoplasmic reticulum and calreticulin-dependent Ca2+ pool in cardiomyocytes are critical for the maintenance of proper cardiac function.
Collapse
Affiliation(s)
- Jody Groenendyk
- Correspondence: (J.G.); (M.M.); Tel.: +1-780-492-2256 (M.M.)
| | | | | | - Marek Michalak
- Correspondence: (J.G.); (M.M.); Tel.: +1-780-492-2256 (M.M.)
| |
Collapse
|
24
|
Wang J, Shi Q, Wang Y, Dawson LW, Ciampa G, Zhao W, Zhang G, Chen B, Weiss RM, Grueter CE, Hall DD, Song LS. Gene Therapy With the N-Terminus of Junctophilin-2 Improves Heart Failure in Mice. Circ Res 2022; 130:1306-1317. [PMID: 35317607 PMCID: PMC9050933 DOI: 10.1161/circresaha.121.320680] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/11/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Transcriptional remodeling is known to contribute to heart failure (HF). Targeting stress-dependent gene expression mechanisms may represent a clinically relevant gene therapy option. We recently uncovered a salutary mechanism in the heart whereby JP2 (junctophilin-2), an essential component of the excitation-contraction coupling apparatus, is site-specifically cleaved and releases an N-terminal fragment (JP2NT [N-terminal fragment of JP2]) that translocates into the nucleus and functions as a transcriptional repressor of HF-related genes. This study aims to determine whether JP2NT can be leveraged by gene therapy techniques for attenuating HF progression in a preclinical pressure overload model. METHODS We intraventricularly injected adeno-associated virus (AAV) (2/9) vectors expressing eGFP (enhanced green fluorescent protein), JP2NT, or DNA-binding deficient JP2NT (JP2NTΔbNLS/ARR) into neonatal mice and induced cardiac stress by transaortic constriction (TAC) 9 weeks later. We also treated mice with established moderate HF from TAC stress with either AAV-JP2NT or AAV-eGFP. RNA-sequencing analysis was used to reveal changes in hypertrophic and HF-related gene transcription by JP2NT gene therapy after TAC. Echocardiography, confocal imaging, and histology were performed to evaluate heart function and pathological myocardial remodeling following stress. RESULTS Mice preinjected with AAV-JP2NT exhibited ameliorated cardiac remodeling following TAC. The JP2NT DNA-binding domain is required for cardioprotection as its deletion within the AAV-JP2NT vector prevented improvement in TAC-induced cardiac dysfunction. Functional and histological data suggest that JP2NT gene therapy after the onset of cardiac dysfunction is effective at slowing the progression of HF. RNA-sequencing analysis further revealed a broad reversal of hypertrophic and HF-related gene transcription by JP2NT overexpression after TAC. CONCLUSIONS Our prevention- and intervention-based approaches here demonstrated that AAV-mediated delivery of JP2NT into the myocardium can attenuate stress-induced transcriptional remodeling and the development of HF when administered either before or after cardiac stress initiation. Our data indicate that JP2NT gene therapy holds great potential as a novel therapeutic for treating hypertrophy and HF.
Collapse
Affiliation(s)
- Jinxi Wang
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242
| | - Qian Shi
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242
| | - Yihui Wang
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242
| | - Logan W. Dawson
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, IA 52242
| | - Grace Ciampa
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, IA 52242
| | - Weiyang Zhao
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242
| | - Guangqin Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242
| | - Biyi Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242
| | - Robert M. Weiss
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242
| | - Chad E. Grueter
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242
| | - Duane D. Hall
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242
| | - Long-Sheng Song
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, IA 52242
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
- Department of Veterans Affairs, Iowa City Medical Center, IA 52242
| |
Collapse
|
25
|
Hulsurkar MM, Lahiri SK, Karch J, Wang MC, Wehrens XHT. Targeting calcium-mediated inter-organellar crosstalk in cardiac diseases. Expert Opin Ther Targets 2022; 26:303-317. [PMID: 35426759 PMCID: PMC9081256 DOI: 10.1080/14728222.2022.2067479] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/14/2022] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Abnormal calcium signaling between organelles such as the sarcoplasmic reticulum (SR), mitochondria and lysosomes is a key feature of heart diseases. Calcium serves as a secondary messenger mediating inter-organellar crosstalk, essential for maintaining the cardiomyocyte function. AREAS COVERED This article examines the available literature related to calcium channels and transporters involved in inter-organellar calcium signaling. The SR calcium-release channels ryanodine receptor type-2 (RyR2) and inositol 1,4,5-trisphosphate receptor (IP3R), and calcium-transporter SR/ER-ATPase 2a (SERCA2a) are illuminated. The roles of mitochondrial voltage-dependent anion channels (VDAC), the mitochondria Ca2+ uniporter complex (MCUC), and the lysosomal H+/Ca2+ exchanger, two pore channels (TPC), and transient receptor potential mucolipin (TRPML) are discussed. Furthermore, recent studies showing calcium-mediated crosstalk between the SR, mitochondria, and lysosomes as well as how this crosstalk is dysregulated in cardiac diseases are placed under the spotlight. EXPERT OPINION Enhanced SR calcium release via RyR2 and reduced SR reuptake via SERCA2a, increased VDAC and MCUC-mediated calcium uptake into mitochondria, and enhanced lysosomal calcium-release via lysosomal TPC and TRPML may all contribute to aberrant calcium homeostasis causing heart disease. While mechanisms of this crosstalk need to be studied further, interventions targeting these calcium channels or combinations thereof might represent a promising therapeutic strategy.
Collapse
Affiliation(s)
- Mohit M Hulsurkar
- Baylor College of Medicine, Houston TX USA
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Satadru K Lahiri
- Baylor College of Medicine, Houston TX USA
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Jason Karch
- Baylor College of Medicine, Houston TX USA
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Meng C Wang
- Baylor College of Medicine, Houston TX USA
- Huffington Center on Aging, Baylor College of Medicine, Houston TX USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Xander H T Wehrens
- Baylor College of Medicine, Houston TX USA
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine (Cardiology), Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics (Cardiology), Baylor College of Medicine, Houston, TX, USA
- Center for Space Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
26
|
Dixon RE. Nanoscale Organization, Regulation, and Dynamic Reorganization of Cardiac Calcium Channels. Front Physiol 2022; 12:810408. [PMID: 35069264 PMCID: PMC8769284 DOI: 10.3389/fphys.2021.810408] [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: 11/06/2021] [Accepted: 11/30/2021] [Indexed: 12/19/2022] Open
Abstract
The architectural specializations and targeted delivery pathways of cardiomyocytes ensure that L-type Ca2+ channels (CaV1.2) are concentrated on the t-tubule sarcolemma within nanometers of their intracellular partners the type 2 ryanodine receptors (RyR2) which cluster on the junctional sarcoplasmic reticulum (jSR). The organization and distribution of these two groups of cardiac calcium channel clusters critically underlies the uniform contraction of the myocardium. Ca2+ signaling between these two sets of adjacent clusters produces Ca2+ sparks that in health, cannot escalate into Ca2+ waves because there is sufficient separation of adjacent clusters so that the release of Ca2+ from one RyR2 cluster or supercluster, cannot activate and sustain the release of Ca2+ from neighboring clusters. Instead, thousands of these Ca2+ release units (CRUs) generate near simultaneous Ca2+ sparks across every cardiomyocyte during the action potential when calcium induced calcium release from RyR2 is stimulated by depolarization induced Ca2+ influx through voltage dependent CaV1.2 channel clusters. These sparks summate to generate a global Ca2+ transient that activates the myofilaments and thus the electrical signal of the action potential is transduced into a functional output, myocardial contraction. To generate more, or less contractile force to match the hemodynamic and metabolic demands of the body, the heart responds to β-adrenergic signaling by altering activity of calcium channels to tune excitation-contraction coupling accordingly. Recent accumulating evidence suggests that this tuning process also involves altered expression, and dynamic reorganization of CaV1.2 and RyR2 channels on their respective membranes to control the amplitude of Ca2+ entry, SR Ca2+ release and myocardial function. In heart failure and aging, altered distribution and reorganization of these key Ca2+ signaling proteins occurs alongside architectural remodeling and is thought to contribute to impaired contractile function. In the present review we discuss these latest developments, their implications, and future questions to be addressed.
Collapse
Affiliation(s)
- Rose E Dixon
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|