1
|
Seo SJ, Jin Y. Enhancing Cardiomyocyte Purity through Lactate-Based Metabolic Selection. Tissue Eng Regen Med 2025:10.1007/s13770-024-00696-4. [PMID: 39820961 DOI: 10.1007/s13770-024-00696-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/20/2024] [Accepted: 12/22/2024] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND Direct reprogramming of fibroblasts into chemically induced cardiomyocyte-like cells (CiCMs) through small molecules presents a promising cell source for cardiac regeneration and therapeutic development. However, the contaminating non-cardiomyocytes, primarily unconverted fibroblasts, reduce the effectiveness of CiCMs in various applications. This study investigated a metabolic selection approach using lactate to enrich CiCMs by exploiting the unique metabolic capability of cardiomyocytes to utilize lactate as an alternative energy source. METHODS Primary mouse embryonic fibroblasts (pMEFs) were reprogrammed into CiCMs and subjected to a glucose-depleted, lactate-supplemented medium for 4 days. Afterward, cell viability was analyzed, and cardiomyocyte efficiency was assessed through the expression of cardiac-specific markers. Additionally, electrophysiological function was evaluated by examining drug-induced responses. RESULTS The lactate treatment led to a significant decrease in the viability of non-cardiomyocytes (pMEF-LAC), while CiCMs (CiCM-LAC) showed minimal cell death. Specifically, the expression of all cardiac-related markers was increased in CiCM-LAC. Metabolically purified CiCMs exhibited enhanced contractile force and increased contraction frequency compared to non-purified CiCMs, as well as an elevated responsiveness to drugs. CONCLUSION This study demonstrates that lactate-based metabolic selection is an effective and practical approach for enriching CiCMs, offering a cost-effective alternative to other purification methods. The application of this strategy could potentially broaden the accessibility and utility of reprogrammed cardiomyocytes in cardiac regeneration and therapeutic development.
Collapse
Affiliation(s)
- Seung Ju Seo
- Department of Physiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yoonhee Jin
- Department of Physiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| |
Collapse
|
2
|
Toms L, FitzPatrick L, Auckland P. Super-resolution microscopy as drug discovery tool. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2025:100209. [PMID: 39824440 DOI: 10.1016/j.slasd.2025.100209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/02/2025] [Indexed: 01/20/2025]
Abstract
At the turn of the century a fundamental resolution barrier in fluorescence microscopy known as the diffraction limit was broken, giving rise to the field of super-resolution microscopy. Subsequent nanoscopic investigation with visible light revolutionised our understanding of how previously unknown molecular features give rise to the emergent behaviour of cells. It transpires that the devil is in these fine molecular details, and essential nanoscale processes were found everywhere researchers chose to look. Now, after nearly two decades, super-resolution microscopy has begun to address previously unmet challenges in the study of human disease and is poised to become a pivotal tool in drug discovery.
Collapse
Affiliation(s)
- Lauren Toms
- Medicines Discovery Catapult, Block 35, Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4ZF.
| | - Lorna FitzPatrick
- Medicines Discovery Catapult, Block 35, Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4ZF
| | - Philip Auckland
- Medicines Discovery Catapult, Block 35, Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4ZF.
| |
Collapse
|
3
|
Wang W, Su W, Han J, Song W, Li X, Xu C, Sun Y, Wang L. Microfluidic platforms for monitoring cardiomyocyte electromechanical activity. MICROSYSTEMS & NANOENGINEERING 2025; 11:4. [PMID: 39788940 PMCID: PMC11718118 DOI: 10.1038/s41378-024-00751-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/04/2024] [Accepted: 06/26/2024] [Indexed: 01/12/2025]
Abstract
Cardiovascular diseases account for ~40% of global deaths annually. This situation has revealed the urgent need for the investigation and development of corresponding drugs for pathogenesis due to the complexity of research methods and detection techniques. An in vitro cardiomyocyte model is commonly used for cardiac drug screening and disease modeling since it can respond to microphysiological environmental variations through mechanoelectric feedback. Microfluidic platforms are capable of accurate fluid control and integration with analysis and detection techniques. Therefore, various microfluidic platforms (i.e., heart-on-a-chip) have been applied for the reconstruction of the physiological environment and detection of signals from cardiomyocytes. They have demonstrated advantages in mimicking the cardiovascular structure and function in vitro and in monitoring electromechanical signals. This review presents a summary of the methods and technologies used to monitor the contractility and electrophysiological signals of cardiomyocytes within microfluidic platforms. Then, applications in common cardiac drug screening and cardiovascular disease modeling are presented, followed by design strategies for enhancing physiology studies. Finally, we discuss prospects in the tissue engineering and sensing techniques of microfluidic platforms.
Collapse
Affiliation(s)
- Wei Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), 250353, Jinan, China
- Shandong Institute of Mechanical Design and Research, 250353, Jinan, China
| | - Weiguang Su
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), 250353, Jinan, China
- Shandong Institute of Mechanical Design and Research, 250353, Jinan, China
| | - Junlei Han
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), 250353, Jinan, China
- Shandong Institute of Mechanical Design and Research, 250353, Jinan, China
| | - Wei Song
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021, Jinan, China
| | - Xinyu Li
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021, Jinan, China
| | - Chonghai Xu
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), 250353, Jinan, China
- Shandong Institute of Mechanical Design and Research, 250353, Jinan, China
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, M5S3G8, Canada.
| | - Li Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), 250353, Jinan, China.
- Shandong Institute of Mechanical Design and Research, 250353, Jinan, China.
| |
Collapse
|
4
|
Carter BA, Parker VE. Role of MicroRNAs in Regulating Sarcoplasmic Reticulum Calcium Handling and Their Implications for Cardiomyocyte Function and Heart Disease. Curr Probl Cardiol 2025:102980. [PMID: 39788467 DOI: 10.1016/j.cpcardiol.2025.102980] [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: 12/19/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
The regulation of calcium signaling within cardiomyocytes is pivotal for maintaining cardiac function, with disruptions in sarcoplasmic reticulum (SR) calcium handling linked to various heart diseases. This review explores the emerging role of microRNAs (miRNAs) in modulating SR calcium dynamics, highlighting their influence on cardiomyocyte maturation, function, and disease progression. We present a comprehensive overview of the mechanisms by which specific miRNAs, such as miR-1, miR-24, and miR-22, regulate key components of calcium handling, including ryanodine receptors, SERCA, and NCX. Notably, we identify critical research gaps, particularly the inconsistent findings regarding miRNA expression in heart disease and the need for standardized experimental conditions. Furthermore, we emphasize the potential of miRNAs as therapeutic targets, given their ability to influence calcium handling pathways and cardiac remodeling. The review also discusses the challenges in translating miRNA research into clinical applications, including the need for safe and effective delivery methods. By synthesizing current knowledge and identifying areas for future investigation, this review aims to provide insights into the therapeutic potential of miRNAs in diagnosing and treating heart diseases, ultimately contributing to improved patient outcomes.
Collapse
Affiliation(s)
- Benjamin Alexander Carter
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
| | - Victoria Elizabeth Parker
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
| |
Collapse
|
5
|
Chan JA, Munro ML. Time-dependent effect of FKBP12 loss in the development of dilated cardiomyopathy. J Gen Physiol 2025; 157:e202413673. [PMID: 39665747 PMCID: PMC11636550 DOI: 10.1085/jgp.202413673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024] Open
Abstract
Hanna et al. reveal that early, but not late, developmental cardiac FKBP12 deficiency leads to dilated cardiomyopathy in the adult heart.
Collapse
Affiliation(s)
- Joan A. Chan
- Department of Physiology, School of Biomedical Sciences and HeartOtago, University of Otago, Dunedin, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Michelle L. Munro
- Department of Physiology, School of Biomedical Sciences and HeartOtago, University of Otago, Dunedin, New Zealand
| |
Collapse
|
6
|
Chavda VR, Bhatt SB, Umaretiya VR, Gajera HP, Padhiyar SM, Kandoliya UK, Parakhia MV. Characterization and metabolomic profiling of endophytic bacteria isolated from Moringa oleifera and Piper betel leaves. Sci Rep 2025; 15:632. [PMID: 39753876 PMCID: PMC11698722 DOI: 10.1038/s41598-024-84840-z] [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: 08/08/2024] [Accepted: 12/27/2024] [Indexed: 01/06/2025] Open
Abstract
Endophytes are microorganisms residing in plant tissues without causing harm and their relevance in medicinal plants has grown due to their biomolecules used in pharmaceuticals. This study isolated two endophytic bacterial strains from the leaves of M. oleifera and P. betel collected from Junagadh Agricultural University. The isolates were characterized morphologically and physio-biochemically, confirming them as gram-positive or gram-negative rods and cocci. Identification using 16S rRNA gene sequencing identified isolates belonging to various genera, including Priestia aryabhattai and Kocuria rhizophila The SEM characterization of the five selected isolates revealed diverse morphological structures, including coccus and rod shapes, organized in various formations. Isolates varied in size, with N3 (Kocuria rhizophila) cocci and S5 (Priestia aryabhattai) rods. Metabolomic analysis using GC/MS and LC-MS revealed diverse metabolic profiles with key compounds like n-Hexadecanoic acid, Pyrrolo[1,2-a]pyrazine-1,4-dione, Dihydrocapsaicin, and β-Homoproline, highlighting the potential of these endophytic bacteria in agricultural applications due to their roles in membrane integrity, antioxidant properties, stress response, and antibacterial activity.
Collapse
Affiliation(s)
- V R Chavda
- Department of Biotechnology, College of Agriculture, Junagadh Agricultural University, Junagadh, India
| | - S B Bhatt
- Department of Biotechnology, College of Agriculture, Junagadh Agricultural University, Junagadh, India.
| | - V R Umaretiya
- Department of Biotechnology, College of Agriculture, Junagadh Agricultural University, Junagadh, India
| | - H P Gajera
- Department of Biotechnology, College of Agriculture, Junagadh Agricultural University, Junagadh, India
| | - S M Padhiyar
- Department of Biotechnology, College of Agriculture, Junagadh Agricultural University, Junagadh, India
| | - U K Kandoliya
- Department of Biotechnology, College of Agriculture, Junagadh Agricultural University, Junagadh, India
| | - M V Parakhia
- Department of Biotechnology, College of Agriculture, Junagadh Agricultural University, Junagadh, India
| |
Collapse
|
7
|
Tang Y, Feng S, Yao K, Cheung SW, Wang K, Zhou X, Xiang L. Exogenous electron generation techniques for biomedical applications: Bridging fundamentals and clinical practice. Biomaterials 2025; 317:123083. [PMID: 39798242 DOI: 10.1016/j.biomaterials.2025.123083] [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: 07/07/2024] [Revised: 12/14/2024] [Accepted: 01/01/2025] [Indexed: 01/15/2025]
Abstract
Endogenous bioelectrical signals are quite crucial in biological development, governing processes such as regeneration and disease progression. Exogenous stimulation, which mimics endogenous bioelectrical signals, has demonstrated significant potential to modulate complex biological processes. Consequently, increasing scientific efforts have focused on developing methods to generate exogenous electrons for biological applications, primarily relying on piezoelectric, acoustoelectric, optoelectronic, magnetoelectric, and thermoelectric principles. Given the expanding body of literature on this topic, a systematic and comprehensive review is essential to foster a deeper understanding and facilitate clinical applications of these techniques. This review synthesizes and compares these methods for generating exogenous electrical signals, their underlying principles (e.g., semiconductor deformation, photoexcitation, vibration and relaxation, and charge separation), biological mechanisms, potential clinical applications, and device designs, highlighting their advantages and limitations. By offering a comprehensive perspective on the critical role of exogenous electrons in biological systems, elucidating the principles of various electron-generation techniques, and exploring possible pathways for developing medical devices utilizing exogenous electrons, this review aims to advance the field and support therapeutic innovation.
Collapse
Affiliation(s)
- Yufei Tang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Shuqi Feng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Keyi Yao
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan, China
| | - Sze Wing Cheung
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Kai Wang
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan, China
| | - Xuemei Zhou
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan, China.
| | - Lin Xiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
8
|
Meng F, Jin S, Liu N. Cardiac selectivity in pulsed field ablation. Curr Opin Cardiol 2025; 40:37-41. [PMID: 39611738 PMCID: PMC11623377 DOI: 10.1097/hco.0000000000001183] [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] [Indexed: 11/30/2024]
Abstract
PURPOSE OF REVIEW This review examines the selective cardiac injury induced by pulsed electric fields during atrial fibrillation ablation. It consolidates findings from both preclinical and clinical studies on cardiac selectivity and explores the potential mechanisms behind this selectivity. RECENT FINDINGS Preclinical studies indicate that pulsed electric fields cause significantly more myocardial injury compared with other tissues. Clinical studies have similarly shown that complication rates for pulsed field ablation are notably lower than those for radiofrequency and cryoballoon ablation. SUMMARY Pulsed field ablation demonstrates a notable selectivity for myocardial injury, likely because of the unique functional and metabolic characteristics of cardiomyocytes. This review delves into the underlying principles of cardiac selectivity and proposes future directions for improving this selectivity. It is important to note that while pulsed field ablation shows promise, its cardiac selectivity is not absolute, as some complications still occur, necessitating further research.
Collapse
Affiliation(s)
- Fanchao Meng
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University
- National Clinical Research Center for Cardiovascular Diseases, Beijing, China
| | - Shuqi Jin
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University
- National Clinical Research Center for Cardiovascular Diseases, Beijing, China
| | - Nian Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University
- National Clinical Research Center for Cardiovascular Diseases, Beijing, China
| |
Collapse
|
9
|
Marimon X, Esquinas F, Ferrer M, Cerrolaza M, Portela A, Benítez R. A Novel non-invasive optical framework for simultaneous analysis of contractility and calcium in single-cell cardiomyocytes. J Mech Behav Biomed Mater 2025; 161:106812. [PMID: 39566161 DOI: 10.1016/j.jmbbm.2024.106812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/13/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
The use of a video method based on the Digital Image Correlation (DIC) algorithm from experimental mechanics to estimate the displacements, strain field, and sarcolemma length in a beating single-cell cardiomyocyte is proposed in this work. The obtained deformation is then correlated with the calcium signal, from calcium imaging where fluorescent dyes sensitive to calcium Ca2+ are used. Our proposed video-based method for simultaneous contraction and intracellular calcium analysis results in a low-cost, non-invasive, and label-free method. This technique has shown great advantages in long-term observations because this type of intervention-free measurement neutralizes the possible alteration in the beating cardiomyocyte introduced by other techniques for measuring cell contractility (e.g., Traction Force Microscopy, Atomic Force Microscopy, Microfabrication or Optical tweezers). Three tests were performed with synthetically augmented data from cardiomyocyte images to validate the robustness of the algorithm. First, a simulated rigid translation of a referenced image is applied, then a rotation, and finally a controlled longitudinal deformation of the referenced image, thus simulating a native realistic deformation. Finally, the proposed framework is evaluated with real experimental data. To validate contraction induced by intracellular calcium concentration, this signal is correlated with a new deformation measure proposed in this article, which is independent of cell orientation in the imaging setup. Finally, based on the displacements obtained by the DIC algorithm, the change in sarcolemma length in a contracting cardiomyocyte is calculated and its temporal correlation with the calcium signal is obtained.
Collapse
Affiliation(s)
- Xavier Marimon
- Automatic Control Department, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IRSJD), Spain; Bioengineering Institute of Technology, Universitat Internacional de Catalunya (UIC), Barcelona, Spain.
| | - Ferran Esquinas
- Automatic Control Department, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain
| | - Miquel Ferrer
- Department of Strength of Materials and Structural Engineering, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain
| | - Miguel Cerrolaza
- School of Engineering, Science and Technology, Valencian International University (VIU), Valencia, Spain
| | - Alejandro Portela
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya (UIC), Barcelona, Spain
| | - Raúl Benítez
- Automatic Control Department, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IRSJD), Spain
| |
Collapse
|
10
|
Zhang XH, Morad M. Regulation of SR and mitochondrial Ca 2+ signaling by L-type Ca 2+ channels and Na/Ca exchanger in hiPSC-CMs. Cell Calcium 2025; 125:102985. [PMID: 39693912 DOI: 10.1016/j.ceca.2024.102985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 12/20/2024]
Abstract
RATIONALE & METHODS While signaling of cardiac SR by surface membrane proteins (ICa & INCX) is well studied, the regulation of mitochondrial Ca2+ by plasmalemmal proteins remains less explored. Here we have examined the signaling of mitochondria and SR by surface-membrane calcium-transporting proteins, using genetically engineered targeted fluorescent probes, mito-GCamP6 and R-CEPIA1er. RESULTS In voltage-clamped and TIRF-imaged cardiomyocytes, low Na+ induced SR Ca2+ release was suppressed by short pre-exposures to ∼100 nM FCCP, suggesting mitochondrial Ca2+ contribution to low Na+ triggered SR Ca2+release. Even though low Na+- or caffeine-triggered SR Ca2+ release activated global mitochondrial Ca2+ uptake, focal mitochondrial Ca2+ signals varied in kinetics and magnitude, showing uptake or release of calcium, depending on cellular location of mitochondria. In spontaneously pacing cells, sustained caffeine exposures depleted the SR Ca2+ content activating mitochondrial Ca2+ uptake followed by sustained mitochondrial pacing. Spontaneous hiPSCCMs pacing was strongly suppressed by L-type calcium channels blockers, but not by inhibiting SERCA2a by CPA. CONCLUSION Spontaneous hiPSCCMs pacing is triggered by influx of calcium through L-type Ca2+ channel that gates the release of SR pools supplemented by NCX-mediated mitochondrial calcium contribution.
Collapse
Affiliation(s)
- Xiao-Hua Zhang
- Cardiac Signaling Center of USC, MUSC and Clemson University, 68 President St BEB 306, Charleston, SC 29425, USA
| | - Martin Morad
- Cardiac Signaling Center of USC, MUSC and Clemson University, 68 President St BEB 306, Charleston, SC 29425, USA.
| |
Collapse
|
11
|
Irving M. Functional control of myosin motors in the cardiac cycle. Nat Rev Cardiol 2025; 22:9-19. [PMID: 39030271 DOI: 10.1038/s41569-024-01063-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/02/2024] [Indexed: 07/21/2024]
Abstract
Contraction of the heart is driven by cyclical interactions between myosin and actin filaments powered by ATP hydrolysis. The modular structure of heart muscle and the organ-level synchrony of the heartbeat ensure tight reciprocal coupling between this myosin ATPase cycle and the macroscopic cardiac cycle. The myosin motors respond to the cyclical activation of the actin and myosin filaments to drive the pressure changes that control the inflow and outflow valves of the heart chambers. Opening and closing of the valves in turn switches the myosin motors between roughly isometric and roughly isotonic contraction modes. Peak filament stress in the heart is much smaller than in fully activated skeletal muscle, although the myosin filaments in the two muscle types have the same number of myosin motors. Calculations indicate that only ~5% of the myosin motors in the heart are needed to generate peak systolic pressure, although many more motors are needed to drive ejection. Tight regulation of the number of active motors is essential for the efficient functioning of the healthy heart - this control is commonly disrupted by gene variants associated with inherited heart disease, and its restoration might be a useful end point in the development of novel therapies.
Collapse
Affiliation(s)
- Malcolm Irving
- Randall Centre for Cell and Molecular Biophysics and BHF Centre for Research Excellence, King's College London, London, UK.
| |
Collapse
|
12
|
Leite LB, Soares LL, Portes AMO, da Silva BAF, Dias TR, Soares TI, Assis MQ, Guimarães-Ervilha LO, Carneiro-Júnior MA, Forte P, Machado-Neves M, Reis ECC, Natali AJ. Combined exercise hinders the progression of pulmonary and right heart harmful remodeling in monocrotaline-induced pulmonary arterial hypertension. J Appl Physiol (1985) 2025; 138:182-194. [PMID: 39611819 DOI: 10.1152/japplphysiol.00379.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/30/2024] [Accepted: 11/20/2024] [Indexed: 11/30/2024] Open
Abstract
The aim of this study was to test whether combined physical exercise training of moderate intensity executed during the development of monocrotaline (MCT)-induced pulmonary arterial hypertension (PAH) hinders the progression of pulmonary and right heart harmful functional and structural remodeling in rats. Wistar rats were injected with MCT (60 mg/kg) and after 24 h were exposed to a combined exercise training program: aerobic exercise (treadmill running-60 min/day; 60% of maximum running speed); and resistance exercise (vertical ladder climbing-15 climbs; 60% of maximum carrying load), on alternate days, 5 days/wk, for ∼3 wk. After euthanasia, the lung and right ventricle (RV) were excised and processed for histological, single myocyte, and biochemical analyses. Combined exercise increased the tolerance to physical effort (time until fatigue and relative maximum load) and prevented increases in pulmonary artery resistance (acceleration time (TA)/ejection time (TE)] and reductions in RV function [tricuspid annular plane systolic excursion (TAPSE)]. Moreover, in myocytes isolated from the RV, combined exercise preserved contraction amplitude, as well as contraction and relaxation velocities, and inhibited reductions in the amplitude and maximum speeds to peak and to decay of the intracellular Ca2+ transient. Furthermore, combined exercise avoided RV (RV weight, cardiomyocyte, extracellular matrix, collagen, inflammatory infiltrate, and extracellular matrix) and lung (pulmonary alveoli and alveolar septum) harmful structural remodeling. In addition, combined exercise restricted RV [nitric oxide (NO) and carbonyl protein (CP)] and lung [catalase (CAT), glutathione S-transferase (GST), and NO] oxidative stress. In conclusion, the applied combined exercise regime hinders the progression of pulmonary and right heart functional and structural harmful remodeling in rats with MCT-induced PAH.NEW & NOTEWORTHY This study reveals that combined exercise improves tolerance to physical effort, prevents increases in pulmonary artery resistance, and conserves the right heart function during the progression of pulmonary arterial hypertension. Our analyses show that combined exercise hinders harmful right ventricular and lung structural remodeling and oxidative stress, which reflects in the maintenance of right ventricular myocytes' contractile function by preserving the intracellular calcium cycling. An attenuated progression of the disease impacts positively on its prognosis.
Collapse
Affiliation(s)
- Luciano Bernardes Leite
- Department of Physical Education, Laboratory of Exercise Biology, Federal University of Viçosa, Viçosa, Brazil
| | - Leôncio Lopes Soares
- Department of Physical Education, Laboratory of Exercise Biology, Federal University of Viçosa, Viçosa, Brazil
| | | | | | - Taís Rodrigues Dias
- Department of Physical Education, Laboratory of Exercise Biology, Federal University of Viçosa, Viçosa, Brazil
| | - Thayana Inácia Soares
- Department of Physical Education, Laboratory of Exercise Biology, Federal University of Viçosa, Viçosa, Brazil
| | - Mirian Quintão Assis
- Department of General Biology, Laboratory of Structural Biology, Federal University of Viçosa, Viçosa, Brazil
| | | | | | - Pedro Forte
- Research Center for Physical Activity and Wellbeing (Livewell), Polytechnic Institute of Bragança, Bragança, Portugal
- CI-ISCE, Higher Instituto of Educational Sciences of the Douro, Penafiel, Portugal
- Department of Sports, Higher Institute of Educational Sciences of the Douro, Penafiel, Portugal
| | - Mariana Machado-Neves
- Department of General Biology, Laboratory of Structural Biology, Federal University of Viçosa, Viçosa, Brazil
| | | | - Antônio José Natali
- Department of Physical Education, Laboratory of Exercise Biology, Federal University of Viçosa, Viçosa, Brazil
| |
Collapse
|
13
|
Gaburjakova J, Domsicova M, Poturnayova A, Gaburjakova M. Flecainide Specifically Targets the Monovalent Countercurrent Through the Cardiac Ryanodine Receptor, While a Dominant Opposing Ca 2+/Ba 2+ Current Is Present. Int J Mol Sci 2024; 26:203. [PMID: 39796059 PMCID: PMC11719481 DOI: 10.3390/ijms26010203] [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: 11/05/2024] [Revised: 12/17/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
Catecholaminergic polymorphic ventricular tachycardia (CPVT) is a highly arrhythmogenic syndrome triggered by stress, primarily linked to gain-of-function point mutations in the cardiac ryanodine receptor (RyR2). Flecainide, as an effective therapy for CPVT, is a known blocker of the surface-membrane Na+ channel, also affecting the intracellular RyR2 channel. The therapeutic relevance of the flecainide-RyR2 interaction remains controversial, as flecainide blocks only the RyR2 current flowing in the opposite direction to the physiological Ca2+ release from the sarcoplasmic reticulum (SR). However, it has been proposed that charge-compensating countercurrent from the cytosol to SR lumen plays a critical role, and its reduction may indeed suppress excessive diastolic SR Ca2+ release through RyR2 channels in CPVT. Monitoring single-channel properties, we examined whether flecainide can target intracellular pathways for charge-balancing currents carried by RyR2 and SR Cl- channels under cell-like conditions. Particularly, the Tris+ countercurrent flowed through the RyR2 channel simultaneously with a dominant reverse Ca2+/Ba2+ current. We demonstrate that flecainide blocked the RyR2-mediated countercurrent without affecting channel activity. In contrast, the SR Cl- channel was completely resistant to flecainide. Based on these findings, it is reasonable to propose that the primary intracellular target of flecainide in vivo is the RyR2-mediated countercurrent.
Collapse
Affiliation(s)
| | | | | | - Marta Gaburjakova
- Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dubravska cesta 9, 840 05 Bratislava, Slovakia; (J.G.); (M.D.); (A.P.)
| |
Collapse
|
14
|
Rahmani M, Pham T, Crossman DJ, Tran K, Taberner AJ, Han JC. Sex differences in cardiac energetics in the rat ventricular muscle. Sci Rep 2024; 14:31242. [PMID: 39732777 DOI: 10.1038/s41598-024-82604-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 12/06/2024] [Indexed: 12/30/2024] Open
Abstract
Cardiac sex-difference functional studies have centred on measurements of twitch force and Ca2+ dynamics. The energy expenditures from these two cellular processes: activation (Ca2+ handling) and contraction (cross-bridge cycling), have not been assessed, and compared, between sexes. Whole-heart studies measuring oxygen consumption do not directly measure the energy expenditure of these activation-contraction processes. In this study, we directly quantified these energy expenditures in terms of heat production. Left-ventricular trabeculae were dissected from rats aged 9-13 weeks. Mechano-energetics of trabeculae were characterized using our work-loop calorimeter under various conditions including varying muscle lengths, stimulus frequencies, and afterloads. Each trabecula was subjected to protocols that allowed it to contract either isometrically or shorten to perform work-loops. Force production, length change, and heat output were simultaneously measured. We extracted various metrics: twitch kinetics, shortening kinetics, mechanical work, and heat associated with cross-bridge cycling and Ca2+ cycling, and quantified mechanical efficiency. Results show no sex differences in any of the metrics. Peak mechanical efficiency was not affected by sex (10.25 ± 0.57% in female trabeculae; 10.93 ± 0.87% in male trabeculae). We conclude that cardiac mechanics and energetics are not affected by sex at the muscle level, within the rat age range studied.
Collapse
Affiliation(s)
- Maryam Rahmani
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand.
| | - Toan Pham
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - David J Crossman
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Kenneth Tran
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Andrew J Taberner
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
- Department of Engineering Science and Biomedical Engineering, The University of Auckland, Auckland, New Zealand
| | - June-Chiew Han
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
15
|
Zhou Y, Tian L, Wang L, Wu W, Liang B, Xiong W, Zhang L, Li X, Chen J. Bisphenol S exposure interrupted human embryonic stem cell derived cardiomyocytes differentiation through ER-NF-κB/ERK signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 290:117576. [PMID: 39729939 DOI: 10.1016/j.ecoenv.2024.117576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 12/29/2024]
Abstract
Bisphenol S (BPS) has been put into production as a wide range of Bisphenol A (BPA) alternatives, while little is known regarding its cardiac developmental toxicity. To explore the effect of BPS on cardiomyocyte differentiation and its mechanism, our study established the human embryonic stem cell-cardiomyocyte differentiation model (hESC-CM), which was divided into early period of differentiation (DP1:1-8d), anaphase period of differentiation (DP2:9-16d) and whole stage of differentiation (DP3:1-16d) exposed to human-related levels of BPS. We found that the survival rate of cardiomyocytes was more sensitive to BPS at the early stage of differentiation than at the anaphase stage of differentiation, and exposure to higher than 30 µg/mL BPS throughout the differentiation period decreased the expression of cTnT. BPS may affect cardiomyocyte differentiation by activating ERβ-NF-κB/ERK signaling pathway, and the signaling pathway of each stage might be different. During DP1, 3 µg/mL of BPS may increase the inflammatory effect of cardiomyocytes mainly through the ERβ-NF-κB signaling pathway, thereby inhibiting cell proliferation, and leading to impaired cardiac function in early differentiation. During DP2, BPS may activate the ERβ-ERK signaling pathway, increase cardiomyocyte apoptosis, alter the establishment of the outer matrix, and thus affect myocardial differentiation. However, exposure to BPS throughout the differentiation stage may disrupt the immune response and cell differentiation, which in turn interrupts heart function. The benchmark dose lower confidence limit (BMDL) of the relative expression of cTnT mRNA exposed by BPS during DP3 was the lowest among all the BMDLs of a good fit, with BMDL5 of 1.96 × 10-2 µg/mL, which is lower than the current reported exposure levels of BPS in maternal serum (0.03-0.07 ng/mL) and maternal umbilical cord serum (0.03-0.12 ng/mL).
Collapse
Affiliation(s)
- Yongru Zhou
- Department of Nutrition and Food Safety, West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| | - Lin Tian
- Department of Nutrition and Food Safety, West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| | - Liang Wang
- Department of Nutrition and Food Safety, West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| | - Wenjing Wu
- Department of Nutrition and Food Safety, West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| | - Baofang Liang
- Department of Nutrition and Food Safety, West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| | - Wei Xiong
- Department of Nutrition and Food Safety, West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| | - Lishi Zhang
- Department of Nutrition and Food Safety, West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| | - Xiaomeng Li
- Department of Nutrition and Food Safety, West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China.
| | - Jinyao Chen
- Department of Nutrition and Food Safety, West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China.
| |
Collapse
|
16
|
Königstein D, Fender H, Plačkić J, Kisko TM, Wöhr M, Kockskämper J. Altered Protein Kinase A-Dependent Phosphorylation of Cav1.2 in Left Ventricular Myocardium from Cacna1c Haploinsufficient Rat Hearts. Int J Mol Sci 2024; 25:13713. [PMID: 39769475 PMCID: PMC11678006 DOI: 10.3390/ijms252413713] [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: 11/06/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
CACNA1C encodes the α1c subunit of the L-type Ca2+ channel, Cav1.2. Ventricular myocytes from haploinsufficient Cacna1c (Cacna1c+/-) rats exhibited reduced expression of Cav1.2 but an apparently normal sarcolemmal Ca2+ influx with an impaired response to sympathetic stress. We tested the hypothesis that the altered phosphorylation of Cav1.2 might underlie the sarcolemmal Ca2+ influx phenotype in Cacna1c+/- myocytes using immunoblotting of the left ventricular (LV) tissue from Cacna1c+/- versus wildtype (WT) hearts. Activation of cAMP-dependent protein kinase A (PKA) increases L-type Ca2+ current and phosphorylates Cav1.2 at serine-1928. Using an antibody directed against this phosphorylation site, we observed elevated phosphorylation of Cav1.2 at serine-1928 in LV myocardium from Cacna1c+/- rats under basal conditions (+110% versus WT). Sympathetic stress was simulated by isoprenaline (100 nM) in Langendorff-perfused hearts. Isoprenaline increased the phosphorylation of serine-1928 in Cacna1c+/- LV myocardium by ≈410%, but the increase was significantly smaller than in WT myocardium (≈650%). In conclusion, our study reveals altered PKA-dependent phosphorylation of Cav1.2 with elevated phosphorylation of serine-1928 under basal conditions and a diminished phosphorylation reserve during β-adrenergic stimulation. These alterations in the phosphorylation of Cav1.2 may explain the apparently normal sarcolemmal Ca2+ influx in Cacna1c+/- myocytes under basal conditions as well as the impaired response to sympathetic stimulation.
Collapse
Affiliation(s)
- David Königstein
- Institute of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Biochemical and Pharmacological Center (BPC) Marburg, University of Marburg, 35032 Marburg, Germany; (D.K.); (H.F.); (J.P.)
| | - Hauke Fender
- Institute of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Biochemical and Pharmacological Center (BPC) Marburg, University of Marburg, 35032 Marburg, Germany; (D.K.); (H.F.); (J.P.)
| | - Jelena Plačkić
- Institute of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Biochemical and Pharmacological Center (BPC) Marburg, University of Marburg, 35032 Marburg, Germany; (D.K.); (H.F.); (J.P.)
| | - Theresa M. Kisko
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, 35032 Marburg, Germany; (T.M.K.); (M.W.)
- Behavioral Neuroscience, Experimental and Biological Psychology, University of Marburg, 35032 Marburg, Germany
- KU Leuven, Faculty of Psychology and Educational Sciences, Research Unit Brain and Cognition, Laboratory of Biological Psychology, Social and Affective Neuroscience Research Group, B-3000 Leuven, Belgium
- KU Leuven, Leuven Brain Institute, B-3000 Leuven, Belgium
| | - Markus Wöhr
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, 35032 Marburg, Germany; (T.M.K.); (M.W.)
- Behavioral Neuroscience, Experimental and Biological Psychology, University of Marburg, 35032 Marburg, Germany
- KU Leuven, Faculty of Psychology and Educational Sciences, Research Unit Brain and Cognition, Laboratory of Biological Psychology, Social and Affective Neuroscience Research Group, B-3000 Leuven, Belgium
- KU Leuven, Leuven Brain Institute, B-3000 Leuven, Belgium
| | - Jens Kockskämper
- Institute of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Biochemical and Pharmacological Center (BPC) Marburg, University of Marburg, 35032 Marburg, Germany; (D.K.); (H.F.); (J.P.)
| |
Collapse
|
17
|
Cao H, Yue L, Shao J, Kong F, Liu S, Huai H, He Z, Mao Z, Yang Y, Tan Y, Wang H. Small extracellular vesicles derived from umbilical cord mesenchymal stem cells alleviate radiation-induced cardiac organoid injury. Stem Cell Res Ther 2024; 15:493. [PMID: 39707562 DOI: 10.1186/s13287-024-04115-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Radiation-induced heart disease (RIHD) is one of the most serious complications of radiation therapy (RT) for thoracic tumors, and new interventions are needed for its prevention and treatment. Small extracellular vesicles (sEVs) from stem cells have attracted much attention due to their ability to repair injury. However, the role of umbilical cord mesenchymal stem cell (UCMSC)-derived sEVs in protecting cardiac organoids from radiation-induced injury and the underlying mechanisms are largely unknown. METHODS A radiation-induced cardiac organoid injury model was established by using X-ray radiation, and the optimal radiation dose of 20 Gy was determined by live/dead staining. After radiation, the cardiac organoids were treated with sEVs derived from UCMSCs, and energy metabolism, calcium transient changes and the ultrastructure of the organoids were assessed through Seahorse analysis, optical mapping and transmission electron microscopy, respectively. Confocal microscopy was used to observe the changes in mitochondrial ROS and mitochondrial membrane potential (ΔΨm). Furthermore, real-time quantitative PCR was used to verify the RNA-seq results. RESULTS After X-ray radiation, the mortality of cardiac organoids significantly increased, energy metabolism decreased, and calcium transients changed. We also observed that the mitochondrial structure of cardiac organoids was disrupted and that ΔΨm was decreased. These effects could be inhibited by sEVs treatment. sEVs may protect against radiation-induced cardiac organoid injury by regulating oxidative phosphorylation and the p53 signaling pathway. CONCLUSION sEVs derived from UCMSCs can be used as a potential therapeutic strategy for radiation-induced heart disease.
Collapse
Affiliation(s)
- Hu Cao
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Liang Yue
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, 27 Taiping Road, Beijing, 100850, P.R. China
| | - Jingyuan Shao
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Fanxuan Kong
- PLA Strategic Support Force Characteristic Medical Center, Beijing, 100101, China
| | - Shenghua Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Hongyu Huai
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Zhichao He
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Zhuang Mao
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yuefeng Yang
- Department of Experimental Medical Science, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Yingxia Tan
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, 27 Taiping Road, Beijing, 100850, P.R. China.
| | - Hua Wang
- Beijing Institute of Radiation Medicine, Beijing, 100850, China.
- Beijing Key Laboratory for Radiobiology, Beijing, 100850, China.
- Department of Experimental Haematology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, P.R. China.
| |
Collapse
|
18
|
Cleary SR, Teng ACT, Kongmeneck AD, Fang X, Phillips TA, Cho EE, Smith RA, Karkut P, Makarewich CA, Kekenes-Huskey PM, Gramolini AO, Robia SL. Dilated cardiomyopathy variant R14del increases phospholamban pentamer stability, blunting dynamic regulation of calcium. J Biol Chem 2024:108118. [PMID: 39710323 DOI: 10.1016/j.jbc.2024.108118] [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: 06/01/2024] [Revised: 10/28/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024] Open
Abstract
The sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA) is a membrane transporter that creates and maintains intracellular Ca2+ stores. In the heart, SERCA is regulated by an inhibitory interaction with the monomeric form of the transmembrane micropeptide phospholamban (PLB). PLB also forms avid homo-pentamers, and dynamic exchange of PLB between pentamers and SERCA is an important determinant of cardiac responsiveness to exercise. Here, we investigated two naturally occurring pathogenic variants of PLB: a cysteine substitution of Arg9 (R9C) and an in-frame deletion of Arg14 (R14del). Both variants are associated with dilated cardiomyopathy. We previously showed that the R9C mutation causes disulfide crosslinking and hyperstabilization of pentamers. While the pathogenic mechanism of R14del is unclear, we hypothesized this mutation may also alter pentamer stability. Immunoblots revealed a significantly increased pentamer:monomer ratio for R14del-PLB compared to WT-PLB. We quantified homo-oligomerization and SERCA-binding in live cells using fluorescence resonance energy transfer (FRET) microscopy. R14del-PLB showed increased affinity for homo-oligomerization and decreased binding affinity for SERCA compared to WT. The data suggest that, like R9C, the R14del mutation stabilizes PLB in pentamers, decreasing its ability to regulate SERCA. The R14del mutation reduces the rate of PLB unbinding from pentamers after transient elevations of Ca2+, limiting the recovery of PLB-SERCA complexes. A computational model predicted that hyperstabilization of PLB pentamers by R14del impairs the ability of cardiac Ca2+ handling to respond to changing heart rates between rest and exercise. We postulate that impaired responsiveness to physiological stress contributes to arrhythmogenesis in human carriers of the R14del mutation.
Collapse
Affiliation(s)
- Sean R Cleary
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
| | - Allen C T Teng
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | | | - Xuan Fang
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
| | - Taylor A Phillips
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
| | - Ellen E Cho
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
| | - Rhys A Smith
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
| | - Patryk Karkut
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
| | - Catherine A Makarewich
- Division of Molecular Cardiovascular Biology of the Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Peter M Kekenes-Huskey
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
| | | | - Seth L Robia
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA.
| |
Collapse
|
19
|
Venkateshappa R, Yildirim Z, Zhao SR, Wu MA, Vacante F, Abilez OJ, Wu JC. Protocol to study electrophysiological properties of hPSC-derived 3D cardiac organoids using MEA and sharp electrode techniques. STAR Protoc 2024; 5:103406. [PMID: 39514393 PMCID: PMC11584947 DOI: 10.1016/j.xpro.2024.103406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/31/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024] Open
Abstract
Continuing advancements in human pluripotent stem cell (hPSC)-derived complex three-dimensional (3D) cardiac tissues require the development of novel technologies or adaptation of existing technologies to understand the physiology of the derived 3D cardiac tissues. In this protocol, we describe the use of multielectrode array (MEA) and sharp electrode electrophysiology techniques to investigate the electrical properties of 3D cardiac organoids. This protocol deciphers the electrical behavior of 3D cardiac organoids at both the single-cell level and tissue level.
Collapse
Affiliation(s)
- Ravichandra Venkateshappa
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Zehra Yildirim
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shane R Zhao
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Matthew A Wu
- Greenstone Biosciences, Palo Alto, CA 94304, USA
| | - Francesca Vacante
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Oscar J Abilez
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Pediatric Cardiac Surgery, Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
20
|
Nordén ES, Bendiksen BA, Bergo KK, Espe EKS, McGinley G, Hasic A, Hauge-Iversen IM, Ugland HK, Shen X, Frisk M, Mabotuwana NS, Louch WE, Hussain RI, Zhang L, Sjaastad I, Cataliotti A, Christensen G. Sacubitril/valsartan preserves regional cardiac function following myocardial infarction in rats. ESC Heart Fail 2024. [PMID: 39696842 DOI: 10.1002/ehf2.15145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/30/2024] [Accepted: 10/11/2024] [Indexed: 12/20/2024] Open
Abstract
AIMS Sacubitril/valsartan (Sac/Val) is used for treatment of heart failure. The effect of Sac/Val on regional dysfunction following myocardial infarction (MI) remains uncertain. This study aimed at understanding the effects of Sac/Val on regional function after MI. METHODS AND RESULTS MI or sham surgery was performed in Sprague-Dawley rats. Animals were randomized to treatment with Sac/Val, valsartan (Val) or vehicle (Veh). Magnetic resonance imaging was used to acquire left ventricular volumes and strain. Left ventricular tissue was obtained for wesern blotting, PCR and Masson's trichrome staining. Isolated cardiac fibroblasts were cultured with Veh, atrial natriuretic peptide (ANP), adrenomedullin (ADM) and sacubitrilat, and collagen expression assessed with droplet digital PCR. RESULTS Sac/Val reduced ventricular end-diastolic volume by 18% compared with Veh, and preserved circumferential systolic strain in the zone proximal to infarction compared with sham after 42 days of treatment (peak strain ± SEM: sham: -0.19 ± 0.01%; Sac/Val: -0.14 ± 0.02%; Val: -0.10 ± 0.02%; Veh: -0.10 ± 0.02%). Masson's trichrome staining demonstrated lower fibrotic deposition in the intermediate zone with Sac/Val treatment than Veh (sham: 2.29 ± 0.17%; Sac/Val: 2.31 ± 0.27%; Val: 3.22 ± 0.60%; Veh: 4.14 ± 0.48%). The amounts of the pro-apoptotic caspase 3 cleavage fragments p19/17 were 89% higher in Val than sham, with Sac/Val showing no significant increase compared with sham. Collagen expression in human fibroblast culture was lower in cells co-treated with sacubitrilat and ANP, an effect not observed with sacubitrilat/ADM co-treatment. CONCLUSIONS Sac/Val preserves in vivo myocardial function in the region most proximal to MI in rats and reduces left ventricular dilatation. These effects may be related to a reduction in both fibrosis and pro-apoptotic signalling.
Collapse
Affiliation(s)
- Einar Sjaastad Nordén
- University of Oslo, Oslo University Hospital and Oslo New University College, Oslo, Norway
| | - Bård Andre Bendiksen
- University of Oslo, Oslo University Hospital and Oslo New University College, Oslo, Norway
| | | | | | - Gary McGinley
- University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Almira Hasic
- University of Oslo and Oslo University Hospital, Oslo, Norway
| | | | | | - Xin Shen
- University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Michael Frisk
- University of Oslo and Oslo University Hospital, Oslo, Norway
| | | | - William E Louch
- University of Oslo and Oslo University Hospital, Oslo, Norway
| | | | - Lili Zhang
- University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Ivar Sjaastad
- University of Oslo and Oslo University Hospital, Oslo, Norway
| | | | | |
Collapse
|
21
|
Bedioune I, Gandon-Renard M, Dessillons M, Barthou A, Varin A, Mika D, Bichali S, Cellier J, Lechène P, Karam S, Dia M, Gomez S, Pereira de Vasconcelos W, Mercier-Nomé F, Mateo P, Dubourg A, Stratakis CA, Mercadier JJ, Benitah JP, Algalarrondo V, Leroy J, Fischmeister R, Gomez AM, Vandecasteele G. Essential Role of the RIα Subunit of cAMP-Dependent Protein Kinase in Regulating Cardiac Contractility and Heart Failure Development. Circulation 2024; 150:2031-2045. [PMID: 39355927 DOI: 10.1161/circulationaha.124.068858] [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: 01/23/2024] [Accepted: 08/16/2024] [Indexed: 10/03/2024]
Abstract
BACKGROUND The heart expresses 2 main subtypes of cAMP-dependent protein kinase (PKA; type I and II) that differ in their regulatory subunits, RIα and RIIα. Embryonic lethality of RIα knockout mice limits the current understanding of type I PKA function in the myocardium. The objective of this study was to test the role of RIα in adult heart contractility and pathological remodeling. METHODS We measured PKA subunit expression in human heart and developed a conditional mouse model with cardiomyocyte-specific knockout of RIα (RIα-icKO). Myocardial structure and function were evaluated by echocardiography, histology, and ECG and in Langendorff-perfused hearts. PKA activity and cAMP levels were determined by immunoassay, and phosphorylation of PKA targets was assessed by Western blot. L-type Ca2+ current (ICa,L), sarcomere shortening, Ca2+ transients, Ca2+ sparks and waves, and subcellular cAMP were recorded in isolated ventricular myocytes (VMs). RESULTS RIα protein was decreased by 50% in failing human heart with ischemic cardiomyopathy and by 75% in the ventricles and in VMs from RIα-icKO mice but not in atria or sinoatrial node. Basal PKA activity was increased ≈3-fold in RIα-icKO VMs. In young RIα-icKO mice, left ventricular ejection fraction was increased and the negative inotropic effect of propranolol was prevented, whereas heart rate and the negative chronotropic effect of propranolol were not modified. Phosphorylation of phospholamban, ryanodine receptor, troponin I, and cardiac myosin-binding protein C at PKA sites was increased in propranolol-treated RIα-icKO mice. Hearts from RIα-icKO mice were hypercontractile, associated with increased ICa,L, and [Ca2+]i transients and sarcomere shortening in VMs. These effects were suppressed by the PKA inhibitor, H89. Global cAMP content was decreased in RIα-icKO hearts, whereas local cAMP at the phospholamban/sarcoplasmic reticulum Ca2+ ATPase complex was unchanged in RIα-icKO VMs. RIα-icKO VMs had an increased frequency of Ca2+ sparks and proarrhythmic Ca2+ waves, and RIα-icKO mice had an increased susceptibility to ventricular tachycardia. On aging, RIα-icKO mice showed progressive contractile dysfunction, cardiac hypertrophy, and fibrosis, culminating in congestive heart failure with reduced ejection fraction that caused 50% mortality at 1 year. CONCLUSIONS These results identify RIα as a key negative regulator of cardiac contractile function, arrhythmia, and pathological remodeling.
Collapse
Affiliation(s)
- Ibrahim Bedioune
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S1180 (I.B., M.G.-R., M.D., A.B., A.V., D.M., S.B., J.C., P.L., S.K., M.D., S.G., W.P.d.V., P.M., A.D., J.-J.M., J.-P.B., V.A., J.L., R.F., A.-M.G., G.V.), Orsay, France
| | - Marine Gandon-Renard
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S1180 (I.B., M.G.-R., M.D., A.B., A.V., D.M., S.B., J.C., P.L., S.K., M.D., S.G., W.P.d.V., P.M., A.D., J.-J.M., J.-P.B., V.A., J.L., R.F., A.-M.G., G.V.), Orsay, France
| | - Matthieu Dessillons
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S1180 (I.B., M.G.-R., M.D., A.B., A.V., D.M., S.B., J.C., P.L., S.K., M.D., S.G., W.P.d.V., P.M., A.D., J.-J.M., J.-P.B., V.A., J.L., R.F., A.-M.G., G.V.), Orsay, France
| | - Aurélien Barthou
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S1180 (I.B., M.G.-R., M.D., A.B., A.V., D.M., S.B., J.C., P.L., S.K., M.D., S.G., W.P.d.V., P.M., A.D., J.-J.M., J.-P.B., V.A., J.L., R.F., A.-M.G., G.V.), Orsay, France
| | - Audrey Varin
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S1180 (I.B., M.G.-R., M.D., A.B., A.V., D.M., S.B., J.C., P.L., S.K., M.D., S.G., W.P.d.V., P.M., A.D., J.-J.M., J.-P.B., V.A., J.L., R.F., A.-M.G., G.V.), Orsay, France
| | - Delphine Mika
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S1180 (I.B., M.G.-R., M.D., A.B., A.V., D.M., S.B., J.C., P.L., S.K., M.D., S.G., W.P.d.V., P.M., A.D., J.-J.M., J.-P.B., V.A., J.L., R.F., A.-M.G., G.V.), Orsay, France
| | - Saïd Bichali
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S1180 (I.B., M.G.-R., M.D., A.B., A.V., D.M., S.B., J.C., P.L., S.K., M.D., S.G., W.P.d.V., P.M., A.D., J.-J.M., J.-P.B., V.A., J.L., R.F., A.-M.G., G.V.), Orsay, France
| | - Joffrey Cellier
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S1180 (I.B., M.G.-R., M.D., A.B., A.V., D.M., S.B., J.C., P.L., S.K., M.D., S.G., W.P.d.V., P.M., A.D., J.-J.M., J.-P.B., V.A., J.L., R.F., A.-M.G., G.V.), Orsay, France
| | - Patrick Lechène
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S1180 (I.B., M.G.-R., M.D., A.B., A.V., D.M., S.B., J.C., P.L., S.K., M.D., S.G., W.P.d.V., P.M., A.D., J.-J.M., J.-P.B., V.A., J.L., R.F., A.-M.G., G.V.), Orsay, France
| | - Sarah Karam
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S1180 (I.B., M.G.-R., M.D., A.B., A.V., D.M., S.B., J.C., P.L., S.K., M.D., S.G., W.P.d.V., P.M., A.D., J.-J.M., J.-P.B., V.A., J.L., R.F., A.-M.G., G.V.), Orsay, France
| | - Maya Dia
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S1180 (I.B., M.G.-R., M.D., A.B., A.V., D.M., S.B., J.C., P.L., S.K., M.D., S.G., W.P.d.V., P.M., A.D., J.-J.M., J.-P.B., V.A., J.L., R.F., A.-M.G., G.V.), Orsay, France
| | - Susana Gomez
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S1180 (I.B., M.G.-R., M.D., A.B., A.V., D.M., S.B., J.C., P.L., S.K., M.D., S.G., W.P.d.V., P.M., A.D., J.-J.M., J.-P.B., V.A., J.L., R.F., A.-M.G., G.V.), Orsay, France
| | - Walma Pereira de Vasconcelos
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S1180 (I.B., M.G.-R., M.D., A.B., A.V., D.M., S.B., J.C., P.L., S.K., M.D., S.G., W.P.d.V., P.M., A.D., J.-J.M., J.-P.B., V.A., J.L., R.F., A.-M.G., G.V.), Orsay, France
| | | | - Philippe Mateo
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S1180 (I.B., M.G.-R., M.D., A.B., A.V., D.M., S.B., J.C., P.L., S.K., M.D., S.G., W.P.d.V., P.M., A.D., J.-J.M., J.-P.B., V.A., J.L., R.F., A.-M.G., G.V.), Orsay, France
| | - Audrey Dubourg
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S1180 (I.B., M.G.-R., M.D., A.B., A.V., D.M., S.B., J.C., P.L., S.K., M.D., S.G., W.P.d.V., P.M., A.D., J.-J.M., J.-P.B., V.A., J.L., R.F., A.-M.G., G.V.), Orsay, France
| | - Constantine A Stratakis
- Human Genetics and Precision Medicine, IMBB, FORTH, Heraklion, Crete, Greece (C.A.S.)
- ELPEN Research Institute, Athens, Greece (C.A.S.)
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD (C.A.S.)
| | - Jean-Jacques Mercadier
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S1180 (I.B., M.G.-R., M.D., A.B., A.V., D.M., S.B., J.C., P.L., S.K., M.D., S.G., W.P.d.V., P.M., A.D., J.-J.M., J.-P.B., V.A., J.L., R.F., A.-M.G., G.V.), Orsay, France
- Xavier Bichat school of Medicine, Paris, France (J.-J.M.)
| | - Jean-Pierre Benitah
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S1180 (I.B., M.G.-R., M.D., A.B., A.V., D.M., S.B., J.C., P.L., S.K., M.D., S.G., W.P.d.V., P.M., A.D., J.-J.M., J.-P.B., V.A., J.L., R.F., A.-M.G., G.V.), Orsay, France
| | - Vincent Algalarrondo
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S1180 (I.B., M.G.-R., M.D., A.B., A.V., D.M., S.B., J.C., P.L., S.K., M.D., S.G., W.P.d.V., P.M., A.D., J.-J.M., J.-P.B., V.A., J.L., R.F., A.-M.G., G.V.), Orsay, France
| | - Jérôme Leroy
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S1180 (I.B., M.G.-R., M.D., A.B., A.V., D.M., S.B., J.C., P.L., S.K., M.D., S.G., W.P.d.V., P.M., A.D., J.-J.M., J.-P.B., V.A., J.L., R.F., A.-M.G., G.V.), Orsay, France
| | - Rodolphe Fischmeister
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S1180 (I.B., M.G.-R., M.D., A.B., A.V., D.M., S.B., J.C., P.L., S.K., M.D., S.G., W.P.d.V., P.M., A.D., J.-J.M., J.-P.B., V.A., J.L., R.F., A.-M.G., G.V.), Orsay, France
| | - Ana-Maria Gomez
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S1180 (I.B., M.G.-R., M.D., A.B., A.V., D.M., S.B., J.C., P.L., S.K., M.D., S.G., W.P.d.V., P.M., A.D., J.-J.M., J.-P.B., V.A., J.L., R.F., A.-M.G., G.V.), Orsay, France
| | - Grégoire Vandecasteele
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S1180 (I.B., M.G.-R., M.D., A.B., A.V., D.M., S.B., J.C., P.L., S.K., M.D., S.G., W.P.d.V., P.M., A.D., J.-J.M., J.-P.B., V.A., J.L., R.F., A.-M.G., G.V.), Orsay, France
| |
Collapse
|
22
|
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
|
23
|
Dominic KL, Schmidt AV, Granzier H, Campbell KS, Stelzer JE. Mechanism-based myofilament manipulation to treat diastolic dysfunction in HFpEF. Front Physiol 2024; 15:1512550. [PMID: 39726859 PMCID: PMC11669688 DOI: 10.3389/fphys.2024.1512550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/21/2024] [Indexed: 12/28/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a major public health challenge, affecting millions worldwide and placing a significant burden on healthcare systems due to high hospitalization rates and limited treatment options. HFpEF is characterized by impaired cardiac relaxation, or diastolic dysfunction. However, there are no therapies that directly treat the primary feature of the disease. This is due in part to the complexity of normal diastolic function, and the challenge of isolating the mechanisms responsible for dysfunction in HFpEF. Without a clear understanding of the mechanisms driving diastolic dysfunction, progress in treatment development has been slow. In this review, we highlight three key areas of molecular dysregulation directly underlying impaired cardiac relaxation in HFpEF: altered calcium sensitivity in the troponin complex, impaired phosphorylation of myosin-binding protein C (cMyBP-C), and reduced titin compliance. We explore how targeting these pathways can restore normal relaxation, improve diastolic function, and potentially provide new therapeutic strategies for HFpEF treatment. Developing effective HFpEF therapies requires precision targeting to balance systolic and diastolic function, avoiding both upstream non-specificity and downstream rigidity. This review highlights three rational molecular targets with a strong mechanistic basis and potential for therapeutic success.
Collapse
Affiliation(s)
- Katherine L. Dominic
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Alexandra V. Schmidt
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
| | - Kenneth S. Campbell
- Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY, United States
- Department of Physiology, University of Kentucky, Lexington, KY, United States
| | - Julian E. Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
24
|
Girault-Sotias PE, Deloux R, De Mota N, Riche S, Daubeuf F, Iturrioz X, Parlakian A, Berdeaux A, Agbulut O, Bonnet D, Boitard S, Llorens-Cortes C. The metabolically resistant apelin-17 analog LIT01-196 reduces cardiac dysfunction and remodeling in heart failure after myocardial infarction. Can J Cardiol 2024:S0828-282X(24)01258-3. [PMID: 39674544 DOI: 10.1016/j.cjca.2024.11.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 11/20/2024] [Accepted: 11/24/2024] [Indexed: 12/16/2024] Open
Abstract
BACKGROUND To protect patients after myocardial infarction (MI) and preserve cardiac function, the development of new therapeutics remains an important issue. Apelin, a neuro-vasoactive peptide, increases aqueous diuresis and cardiac contractility while reducing vascular resistance. However, its in vivo half-life is very short. We therefore developed a metabolically resistant apelin-17 analog, LIT01-196 and investigated its effects on cardiac function and remodeling in a murine MI model. METHODS The selectivity of LIT01-196 towards ApelinR was checked in vitro. Its in vivo half-life was assessed in male Swiss mice by radioimmunoassay. After permanent coronary artery ligation to induce MI, mice received subcutaneous administration of LIT01-196 (MI+LIT01-196, 9 mg/kg/day) or saline (MI+Vehicle) for 4 weeks. LV function was assessed using echocardiography and Millar catheter, vascular density by immunofluorescence and cardiac fibrosis by Sirius red staining. Real-time quantitative PCR measured mRNA expression of HF and fibrosis biomarkers and SERCA2. RESULTS The in vivo half-life of LIT01-196, a specific and selective ApelinR agonist, was two and a half hours. MI+LIT01-196 mice showed significantly improved LV function, reduced HF biomarkers and enhanced cardiac contractility and SERCA2 expression compared with MI+Vehicle. LIT01-196 treatment almost doubled cardiac vascular density and maintained LV wall thickness post-MI. It also significantly reduced cardiac fibrosis and fibrosis biomarkers, without decreasing arterial blood pressure. CONCLUSIONS Chronic LIT01-196 treatment post-MI improves LV function without decreasing blood pressure, increases cardiac vascular density and reduces cardiac remodeling. This suggests that Apelin-R activation by LIT01-196, may constitute an original pharmacological approach for HF treatment after MI.
Collapse
Affiliation(s)
- Pierre-Emmanuel Girault-Sotias
- Laboratory of Central Neuropeptides in the Regulation of Water Balance and Cardiovascular Functions, College de France, CIRB, INSERM U1050/CNRS UMR7241, 75005 Paris, France
| | - Robin Deloux
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 75005, Paris, France
| | - Nadia De Mota
- Laboratory of Central Neuropeptides in the Regulation of Water Balance and Cardiovascular Functions, College de France, CIRB, INSERM U1050/CNRS UMR7241, 75005 Paris, France
| | - Stephanie Riche
- Laboratoire d'Innovation Thérapeutique, UMR7200 CNRS/Université de Strasbourg, Labex MEDALIS, Faculté de Pharmacie, 67401 Illkirch, France
| | - François Daubeuf
- Laboratoire d'Innovation Thérapeutique, UMR7200 CNRS/Université de Strasbourg, Labex MEDALIS, Faculté de Pharmacie, 67401 Illkirch, France
| | - Xavier Iturrioz
- Université Paris Saclay, CEA, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, 91191 Gif-sur-Yvette, France
| | - A Parlakian
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 75005, Paris, France
| | - Alain Berdeaux
- INSERM U955-IMRB Equipe 03 Université Paris Est Créteil, 94010 Créteil, France
| | - Onnik Agbulut
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 75005, Paris, France
| | - Dominique Bonnet
- Laboratoire d'Innovation Thérapeutique, UMR7200 CNRS/Université de Strasbourg, Labex MEDALIS, Faculté de Pharmacie, 67401 Illkirch, France
| | - Solene Boitard
- Laboratory of Central Neuropeptides in the Regulation of Water Balance and Cardiovascular Functions, College de France, CIRB, INSERM U1050/CNRS UMR7241, 75005 Paris, France
| | - Catherine Llorens-Cortes
- Laboratory of Central Neuropeptides in the Regulation of Water Balance and Cardiovascular Functions, College de France, CIRB, INSERM U1050/CNRS UMR7241, 75005 Paris, France; Université Paris Saclay, CEA, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, 91191 Gif-sur-Yvette, France.
| |
Collapse
|
25
|
Nikolaienko R, Bovo E, Zima AV. Expression level of cardiac ryanodine receptors dictates properties of Ca 2+-induced Ca 2+ release. BIOPHYSICAL REPORTS 2024; 4:100183. [PMID: 39341600 PMCID: PMC11532243 DOI: 10.1016/j.bpr.2024.100183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/10/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
The type 2 ryanodine receptor (RyR2) is the major Ca2+ release channel required for Ca2+-induced Ca2+ release (CICR) and cardiac excitation-contraction coupling. The cluster organization of RyR2 at the dyad is critical for efficient CICR. Despite its central role in cardiac Ca2+ signaling, the mechanisms that control CICR are not fully understood. As a single RyR2 Ca2+ flux dictates local CICR that underlies Ca2+ sparks, RyR2 density in a cluster, and therefore the distance between RyR2s, should have a profound impact on local CICR. Here, we studied the effect of the RyR2 expression level ([RyR2]) on CICR activation, termination, and amplitude. The endoplasmic reticulum (ER)-targeted Ca2+ sensor RCEPIA-1er was used to directly measure the ER [Ca2+] (Ca2+]ER) in the T-Rex-293 the sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2a) stable cell line expressing human RyR2. Cells coexpressing RyR2 and SERCA2a produced periodic [Ca2+]ER depletions in the form of spontaneous Ca2+ waves due to propagating CICR. For each studied cell, the [Ca2+]ER at which Ca2+ waves are activated and terminated was analyzed as a function of [RyR2]. CICR parameters, such as [Ca2+]ER activation, termination, and amplitude, were inversely proportional to [RyR2] at low-intermediate levels. Increasing the sensitivity of RyR2 to cytosolic Ca2+ lowered the [Ca2+]ER at which CICR is activated and terminated. Decreasing the sensitivity of RyR2 to cytosolic Ca2+ had the opposite effect on CICR. These results suggest that RyR2 density in the release cluster should have a significant impact on local CICR activation and termination. Since SR Ca2+ load is evenly distributed throughout the SR network, clusters with higher RyR2 density would have a higher probability of initiating spontaneous CICR.
Collapse
Affiliation(s)
- Roman Nikolaienko
- Department of Cell and Molecular Physiology, Strich School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Elisa Bovo
- Department of Cell and Molecular Physiology, Strich School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Aleksey V Zima
- Department of Cell and Molecular Physiology, Strich School of Medicine, Loyola University Chicago, Maywood, Illinois.
| |
Collapse
|
26
|
Li L, Shanmugasundaram A, Kim J, Oyunbaatar NE, Kanade PP, Cha SE, Lim D, Lee CH, Kim YB, Lee BK, Kim ES, Lee DW. Graphene SU-8 Platform for Enhanced Cardiomyocyte Maturation and Intercellular Communication in Cardiac Drug Screening. ACS NANO 2024; 18:33293-33309. [PMID: 39591586 DOI: 10.1021/acsnano.4c05365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2024]
Abstract
Cell culture substrates designed for myocardial applications are pivotal in promoting the maturation and functional integration of cardiomyocytes. However, traditional in vitro models often inadequately mimic the diverse biochemical signals and electrophysiological properties of mature cardiomyocytes. Herein, we propose the application of monolayer graphene, transferred onto SU-8 cantilevers integrated with a microelectrode array, to evaluate its influence on the structural, functional, and electro-mechano-physiological properties of cardiomyocytes. The monolayer graphene, prepared using chemical vapor deposition, is adeptly transferred to the target substrates via thermal release tape. The electrical conductivity of these graphene-enhanced SU-8 substrates is about 1600 S/cm, markedly surpassing that of previously reported cell culture substrates. Immunofluorescence staining and Western blot analyses reveal that the electrically conductive graphene significantly enhances cardiomyocyte maturation and cardiac marker expression compared to bare SU-8 substrates. Cardiomyocytes cultured on graphene-transferred substrates exhibit conduction velocity approximately 3.4 times greater than that of the control group. Such improvements in cardiac marker expression, mechano-electrophysiological performance lead to better responsiveness to cardiovascular drugs, such as Verapamil and Isoproterenol. While the graphene monolayer does not fully replicate the complex environment found in native cardiac tissue, its use on SU-8 substrates offers a feasible approach for accelerating cardiomyocyte maturation and facilitating drug screening applications.
Collapse
Affiliation(s)
- Longlong Li
- Department of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Arunkumar Shanmugasundaram
- Department of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jongyun Kim
- Department of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
- Center for Next-Generation Sensor Research and Development, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Nomin-Erdene Oyunbaatar
- Department of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Pooja P Kanade
- Centre for Quantum Materials and Technology, School of Mathematics and Physics, Queen's University Belfast, Northern Ireland, Belfast BT7 1NN, U.K
| | - Seong-Eung Cha
- Department of Biological Sciences, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Daeyun Lim
- Green Energy & Nano Technology R&D Group, Korea Institute of Industrial Technology, Gwangju 61012, Republic of Korea
| | - Chil-Hyoung Lee
- Green Energy & Nano Technology R&D Group, Korea Institute of Industrial Technology, Gwangju 61012, Republic of Korea
| | - Young-Baek Kim
- Green Energy & Nano Technology R&D Group, Korea Institute of Industrial Technology, Gwangju 61012, Republic of Korea
| | - Bong-Kee Lee
- Department of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Eung-Sam Kim
- Department of Biological Sciences, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Dong-Weon Lee
- Department of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
- Center for Next-Generation Sensor Research and Development, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
27
|
Wu Z, Park J, Steiner PR, Zhu B, Zhang JXJ. A Graph-Based Machine-Learning Approach Combined with Optical Measurements to Understand Beating Dynamics of Cardiomyocytes. J Comput Biol 2024. [PMID: 39648722 DOI: 10.1089/cmb.2024.0491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024] Open
Abstract
The development of computational models for the prediction of cardiac cellular dynamics remains a challenge due to the lack of first-principled mathematical models. We develop a novel machine-learning approach hybridizing physics simulation and graph networks to deliver robust predictions of cardiomyocyte dynamics. Embedded with inductive physical priors, the proposed constraint-based interaction neural projection (CINP) algorithm can uncover hidden physical constraints from sparse image data on a small set of beating cardiac cells and provide robust predictions for heterogenous large-scale cell sets. We also implement an in vitro culture and imaging platform for cellular motion and calcium transient analysis to validate the model. We showcase our model's efficacy by predicting complex organoid cellular behaviors in both in silico and in vitro settings.
Collapse
Affiliation(s)
- Ziqian Wu
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Jiyoon Park
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Paul R Steiner
- Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Bo Zhu
- School of Interactive Computing, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - John X J Zhang
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| |
Collapse
|
28
|
Trus M, Atlas D. Non-ionotropic voltage-gated calcium channel signaling. Channels (Austin) 2024; 18:2341077. [PMID: 38601983 PMCID: PMC11017947 DOI: 10.1080/19336950.2024.2341077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/04/2024] [Indexed: 04/12/2024] Open
Abstract
Voltage-gated calcium channels (VGCCs) are the major conduits for calcium ions (Ca2+) within excitable cells. Recent studies have highlighted the non-ionotropic functionality of VGCCs, revealing their capacity to activate intracellular pathways independently of ion flow. This non-ionotropic signaling mode plays a pivotal role in excitation-coupling processes, including gene transcription through excitation-transcription (ET), synaptic transmission via excitation-secretion (ES), and cardiac contraction through excitation-contraction (EC). However, it is noteworthy that these excitation-coupling processes require extracellular calcium (Ca2+) and Ca2+ occupancy of the channel ion pore. Analogous to the "non-canonical" characterization of the non-ionotropic signaling exhibited by the N-methyl-D-aspartate receptor (NMDA), which requires extracellular Ca2+ without the influx of ions, VGCC activation requires depolarization-triggered conformational change(s) concomitant with Ca2+ binding to the open channel. Here, we discuss the contributions of VGCCs to ES, ET, and EC coupling as Ca2+ binding macromolecules that transduces external stimuli to intracellular input prior to elevating intracellular Ca2+. We emphasize the recognition of calcium ion occupancy within the open ion-pore and its contribution to the excitation coupling processes that precede the influx of calcium. The non-ionotropic activation of VGCCs, triggered by the upstroke of an action potential, provides a conceptual framework to elucidate the mechanistic aspects underlying the microseconds nature of synaptic transmission, cardiac contractility, and the rapid induction of first-wave genes.
Collapse
Affiliation(s)
- Michael Trus
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Daphne Atlas
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
29
|
Lu HH, Ege D, Salehi S, Boccaccini AR. Ionic medicine: Exploiting metallic ions to stimulate skeletal muscle tissue regeneration. Acta Biomater 2024; 190:1-23. [PMID: 39454933 DOI: 10.1016/j.actbio.2024.10.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
The regeneration of healthy and functional skeletal muscle at sites of injuries and defects remains a challenge. Mimicking the natural environment surrounding skeletal muscle cells and the application of electrical and mechanical stimuli are approaches being investigated to promote muscle tissue regeneration. Likewise, chemical stimulation with therapeutic (biologically active) ions is an emerging attractive alternative in the tissue engineering and regenerative medicine fields, specifically to trigger myoblast proliferation, myogenic differentiation, myofiber formation, and ultimately to promote new muscle tissue growth. The present review covers the specialized literature focusing on the biochemical stimulation of muscle tissue repair by applying inorganic ions (bioinorganics). Extracting information from the literature, different ions and their potential influence as chemical cues on skeletal muscle regeneration are discussed. It is revealed that different ions and their varied doses have an individual effect at different stages of muscle cellular development. The dose-dependent effects of ions, as well as applications of ions alone and in combination with biomaterials, are also summarized. Some ions, such as boron, silicon, magnesium, selenium and zinc, are reported to exhibit a beneficial effect on skeletal muscle cells in carefully controlled doses, while the effects of other ions such as iron and copper appear to be contradictory. In addition, calcium is an essential regulatory ion for the differentiation of myoblasts. On the other hand, some ions such as phosphate have been shown to inhibit muscle cell behavior. This review thus provides a complete overview of the application of ionic stimulation for skeletal muscle tissue engineering applications, highlighting the importance of inorganic ions as an attractive alternative to the application of small molecules and growth factors to stimulate muscle tissue repair. STATEMENT OF SIGNIFICANCE: Ionic medicine (IM) is emerging as a promising and attractive approach in the field of tissue engineering, including muscle tissue regeneration. IM is based on the delivery of biologically active ions to injury sites, acting as stimulants for the repair process. This method offers a potentially simpler and more affordable alternative to conventional biomolecule-based regulators such as growth factors. Different biologically active ions, depending on their specific doping concentrations, can have varying effects on cellular development, which could be either beneficial or inhibitory. This literature review covers the field of IM in muscle regeneration with focus on the impact of various ions on skeletal muscle regeneration. The paper is thus a critical summary for guiding future research in ionic-related regenerative medicine, highlighting the potential and challenges of this approach for muscle regeneration.
Collapse
Affiliation(s)
- Hsuan-Heng Lu
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Duygu Ege
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany; Institute of Biomedical Engineering, Bogazici University, Rasathane St., Kandilli 34684, Istanbul, Turkey
| | - Sahar Salehi
- Department of Biomaterials, Faculty of Engineering Science, University of Bayreuth, 95447 Bayreuth, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany.
| |
Collapse
|
30
|
Zhong M, Karma A. Role of ryanodine receptor cooperativity in Ca 2+-wave-mediated triggered activity in cardiomyocytes. J Physiol 2024; 602:6745-6787. [PMID: 39565684 DOI: 10.1113/jp286145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 09/23/2024] [Indexed: 11/22/2024] Open
Abstract
Ca2+ waves are known to trigger delayed after-depolarizations that can cause malignant cardiac arrhythmias. However, modelling Ca2+ waves using physiologically realistic models has remained a major challenge. Existing models with low Ca2+ sensitivity of ryanodine receptors (RyRs) necessitate large release currents, leading to an unrealistically large Ca2+ transient amplitude incompatible with the experimental observations. Consequently, current physiologically detailed models of delayed after-depolarizations resort to unrealistic cell architectures to produce Ca2+ waves with a normal Ca2+ transient amplitude. Here, we address these challenges by incorporating RyR cooperativity into a physiologically detailed model with a realistic cell architecture. We represent RyR cooperativity phenomenologically through a Hill coefficient within the sigmoid function of RyR open probability. Simulations in permeabilized myocytes with high Ca2+ sensitivity reveal that a sufficiently large Hill coefficient is required for Ca2+ wave propagation via the fire-diffuse-fire mechanism. In intact myocytes, propagating Ca2+ waves can occur only within an intermediate Hill coefficient range. Within this range, the spark rate is neither too low, enabling Ca2+ wave propagation, nor too high, allowing for the maintenance of a high sarcoplasmic reticulum load during diastole of the action potential. Moreover, this model successfully replicates other experimentally observed manifestations of Ca2+-wave-mediated triggered activity, including phase 2 and phase 3 early after-depolarizations and high-frequency voltage-Ca2+ oscillations. These oscillations feature an elevated take-off potential with depolarization mediated by the L-type Ca2+ current. The model also sheds light on the roles of luminal gating of RyRs and the mobile buffer ATP in the genesis of these arrhythmogenic phenomena. KEY POINTS: Existing mathematical models of Ca2+ waves use an excessively large Ca2+-release current or unrealistic diffusive coupling between release units. Our physiologically realistic model, using a Hill coefficient in the ryanodine receptor (RyR) gating function to represent RyR cooperativity, addresses these limitations and generates organized Ca2+ waves at Hill coefficients ranging from ∼5 to 10, as opposed to the traditional value of 2. This range of Hill coefficients gives a spark rate neither too low, thereby enabling Ca2+ wave propagation, nor too high, allowing for the maintenance of a high sarcoplasmic reticulum load during the plateau phase of the action potential. Additionally, the model generates Ca2+-wave-mediated phase 2 and phase 3 early after-depolarizations, and coupled membrane voltage with Ca2+ oscillations mediated by the L-type Ca2+ current. This study suggests that pharmacologically targeting RyR cooperativity could be a promising strategy for treating cardiac arrhythmias linked to Ca2+-wave-mediated triggered activity.
Collapse
Affiliation(s)
- Mingwang Zhong
- Physics Department and Center for Interdisciplinary Research in Complex Systems, Northeastern University, Boston, MA, USA
| | - Alain Karma
- Physics Department and Center for Interdisciplinary Research in Complex Systems, Northeastern University, Boston, MA, USA
| |
Collapse
|
31
|
Hassel KR, Gibson AM, Šeflová J, Cho EE, Blair NS, Van Raamsdonk CD, Anderson DM, Robia SL, Makarewich CA. Another-regulin regulates cardiomyocyte calcium handling via integration of neuroendocrine signaling with SERCA2a activity. J Mol Cell Cardiol 2024; 197:45-58. [PMID: 39437886 PMCID: PMC11588527 DOI: 10.1016/j.yjmcc.2024.10.008] [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] [Received: 06/22/2024] [Revised: 10/02/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Calcium (Ca2+) dysregulation is a hallmark feature of cardiovascular disease. Intracellular Ca2+ regulation is essential for proper heart function and is controlled by the sarco/endoplasmic reticulum Ca2+ ATPase (SERCA2a). Another-regulin (ALN) is a newly discovered cardiomyocyte-expressed SERCA2a inhibitor, suggesting cardiomyocyte Ca2+-handling is more complex than previously appreciated. To study the role of ALN in cardiomyocytes, we generated ALN null mice (knockout, KO) and found that cardiomyocytes from these animals displayed enhanced Ca2+ cycling and contractility compared to wildtype (WT) mice, indicating enhanced SERCA2a activity. In vitro and in vivo studies show that ALN is post-translationally modified via phosphorylation on Serine 19 (S19), suggesting this contributes to its ability to regulate SERCA2a. Immunoprecipitation and FRET analysis of ALN-WT, phospho-deficient ALN (S19A), or phosphomimetic ALN (S19D) revealed that S19 phosphorylation alters the SERCA2a-ALN interaction, leading to relief of its inhibitory effects. Adeno-associated virus mediated delivery of ALN-WT or phospho-mutant ALN-S19A/D in ALN KO mice showed that cardiomyocyte-specific expression of phospho-deficient ALN-S19A resulted in increased SERCA2a inhibition characterized by reduced rates of cytoplasmic Ca2+ clearance compared to ALN-WT and ALN-S19D expressing cells, further supporting a role for this phosphorylation event in controlling SERCA2a-regulation by ALN. Levels of ALN phosphorylation were markedly increased in cardiomyocytes in response to Gαq agonists (angiotensin II, endothelin-1, phenylephrine) and Gαq-mediated phosphorylation of ALN translated to increased Ca2+ cycling in cardiomyocytes from WT but not ALN KO mice. Collectively, these results indicate that ALN uniquely regulates Ca2+ handling in cardiomyocytes via integration of neuroendocrine signaling with SERCA2a activity.
Collapse
Affiliation(s)
- Keira R Hassel
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Aaron M Gibson
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jaroslava Šeflová
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
| | - Ellen E Cho
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
| | - N Scott Blair
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Catherine D Van Raamsdonk
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, B.C., Canada
| | - Douglas M Anderson
- Department of Medicine, Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Seth L Robia
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
| | - Catherine A Makarewich
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA.
| |
Collapse
|
32
|
Kavalenia TA, Lapshina EA, Ilyich TV, Zhao HC, Zavodnik IB. Functional activity and morphology of isolated rat cardiac mitochondria under calcium overload. Effect of naringin. Mol Cell Biochem 2024; 479:3329-3340. [PMID: 38332449 DOI: 10.1007/s11010-024-04935-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 01/08/2024] [Indexed: 02/10/2024]
Abstract
The function of mitochondria as a regulator of myocyte calcium homeostasis has been extensively discussed. The aim of the present work was further clarification of the details of modulation of the functional activity of rat cardiac mitochondria by exogenous Ca2+ ions either in the absence or in the presence of the plant flavonoid naringin. Low free Ca2+ concentrations (40-250 nM) effectively inhibited the respiratory activity of heart mitochondria, remaining unaffected the efficacy of oxygen consumption. In the presence of high exogenous Ca2+ ion concentrations (Ca2+ free was 550 µM), we observed a dramatic increase in mitochondrial heterogeneity in size and electron density, which was related to calcium-induced opening of the mitochondrial permeability transition pores (MPTP) and membrane depolarization (Ca2+free ions were from 150 to 750 µM). Naringin partially prevented Ca2+-induced cardiac mitochondrial morphological transformations (200 µM) and dose-dependently inhibited the respiratory activity of mitochondria (10-75 µM) in the absence or in the presence of calcium ions. Our data suggest that naringin (75 µM) promoted membrane potential dissipation, diminishing the potential-dependent accumulation of calcium ions by mitochondria and inhibiting calcium-induced MPTP formation. The modulating effect of the flavonoid on Ca2+-induced mitochondria alterations may be attributed to the weak-acidic nature of the flavonoid and its protonophoric/ionophoric properties. Our results show that the sensitivity of rat heart mitochondria to Ca2+ ions was much lower in the case of MPTP opening and much higher in the case of respiration inhibition as compared to liver mitochondria.
Collapse
Affiliation(s)
- T A Kavalenia
- Department of Biochemistry, Yanka Kupala State University of Grodno, Bulvar Leninskogo Komsomola, 5, 230009, Grodno, Belarus
| | - E A Lapshina
- Department of Biochemistry, Yanka Kupala State University of Grodno, Bulvar Leninskogo Komsomola, 5, 230009, Grodno, Belarus
| | - T V Ilyich
- Department of Biochemistry, Yanka Kupala State University of Grodno, Bulvar Leninskogo Komsomola, 5, 230009, Grodno, Belarus
| | - Hu-Cheng Zhao
- Institute of Biomechanics and Medical Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - I B Zavodnik
- Department of Biochemistry, Yanka Kupala State University of Grodno, Bulvar Leninskogo Komsomola, 5, 230009, Grodno, Belarus.
| |
Collapse
|
33
|
De Waard S, Hinnen H, Kucera JP. Stochastic and alternating pacing paradigms to assess the stability of cardiac conduction. J Mol Cell Cardiol 2024; 197:20-33. [PMID: 39437883 DOI: 10.1016/j.yjmcc.2024.10.007] [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: 05/13/2024] [Revised: 10/14/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Reentry, the most common cause of severe arrhythmias, is initiated by slow conduction and conduction block. Hence, evaluating conduction velocity and conduction block is of primary importance. However, the assessment of cardiac conduction safety in experimental and clinical settings remains elusive. To identify markers of conduction instability that can be determined experimentally, we developed an approach based on new pacing paradigms. Conduction across a cardiac tissue expansion was assessed in computer simulations and in experiments using cultures of neonatal murine cardiomyocytes on microelectrode arrays. Simulated and in vitro tissues were paced at a progressively increasing rate, with stochastic or alternating variations of cycle length, until conduction block occurred. Increasing pacing rate led to conduction block near the expansion. When stochastic or alternating variations were introduced into the pacing protocol, the standard deviation and the amplitude of alternating variations of local conduction times emerged as markers of unstable conduction prone to block. In both simulations and experiments, conduction delays were prolonged at the expansion but increased only slightly during the pacing protocol. In contrast, these markers of instability increased several-fold, early before block occurrence. The first and second moments of these two metrics provided an estimation of the site of block and the accuracy of this estimation. Therefore, when beat-to-beat variations of pacing cycle length are introduced into a pacing protocol, the local variability of conduction permits to predict sites of block. Our pacing paradigms may have translational applications in clinical cardiac electrophysiology, particularly in identifying ablation targets during mapping procedures.
Collapse
Affiliation(s)
- Stephan De Waard
- Department of Physiology, University of Bern, Bühlplatz 5, CH-3012 Bern, Switzerland.
| | - Helene Hinnen
- Department of Physiology, University of Bern, Bühlplatz 5, CH-3012 Bern, Switzerland.
| | - Jan P Kucera
- Department of Physiology, University of Bern, Bühlplatz 5, CH-3012 Bern, Switzerland.
| |
Collapse
|
34
|
Florindi C, Vurro V, Moretti P, Bertarelli C, Zaza A, Lanzani G, Lodola F. Role of stretch-activated channels in light-generated action potentials mediated by an intramembrane molecular photoswitch. J Transl Med 2024; 22:1068. [PMID: 39605022 PMCID: PMC11600573 DOI: 10.1186/s12967-024-05902-4] [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: 09/05/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The use of light to control the activity of living cells is a promising approach in cardiac research due to its unparalleled spatio-temporal selectivity and minimal invasiveness. Ziapin2, a newly synthesized azobenzene compound, has recently been reported as an efficient tool for light-driven modulation of excitation-contraction coupling (ECC) in human-induced pluripotent stem cells-derived cardiomyocytes. However, the exact biophysical mechanism of this process remains incompletely understood. METHODS To address this, we performed a detailed electrophysiological characterization in a more mature cardiac model, specifically adult mouse ventricular myocytes (AMVMs). RESULTS Our in vitro results demonstrate that Ziapin2 can photomodulate cardiac ECC in mature AMVMs without affecting the main transporters and receptors located within the sarcolemma. We established a connection between Ziapin2-induced membrane thickness modulation and light-generated action potentials by showcasing the pivotal role of stretch-activated channels (SACs). Notably, our experimental findings, through pharmacological blockade, suggest that non-selective SACs might serve as the biological culprit responsible for the effect. CONCLUSIONS Taken together, these findings elucidate the intricacies of Ziapin2-mediated photostimulation mechanism and open new perspectives for its application in cardiac research.
Collapse
Affiliation(s)
- Chiara Florindi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, P.za della Scienza 2, 20126, Milan, Italy
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Milan, Italy
| | - Vito Vurro
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Milan, Italy
| | - Paola Moretti
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Milan, Italy
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Chiara Bertarelli
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Milan, Italy
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Antonio Zaza
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, P.za della Scienza 2, 20126, Milan, Italy
| | - Guglielmo Lanzani
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Milan, Italy
- Department of Physics, Politecnico di Milano, Milan, Italy
| | - Francesco Lodola
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, P.za della Scienza 2, 20126, Milan, Italy.
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Milan, Italy.
| |
Collapse
|
35
|
Panday N, Sigdel D, Adam I, Ramirez J, Verma A, Eranki AN, Wang W, Wang D, Ping P. Data-Driven Insights into the Association Between Oxidative Stress and Calcium-Regulating Proteins in Cardiovascular Disease. Antioxidants (Basel) 2024; 13:1420. [PMID: 39594561 PMCID: PMC11590986 DOI: 10.3390/antiox13111420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/10/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
A growing body of biomedical literature suggests a bidirectional regulatory relationship between cardiac calcium (Ca2+)-regulating proteins and reactive oxygen species (ROS) that is integral to the pathogenesis of various cardiac disorders via oxidative stress (OS) signaling. To address the challenge of finding hidden connections within the growing volume of biomedical research, we developed a data science pipeline for efficient data extraction, transformation, and loading. Employing the CaseOLAP (Context-Aware Semantic Analytic Processing) algorithm, our pipeline quantifies interactions between 128 human cardiomyocyte Ca2+-regulating proteins and eight cardiovascular disease (CVD) categories. Our machine-learning analysis of CaseOLAP scores reveals that the molecular interfaces of Ca2+-regulating proteins uniquely associate with cardiac arrhythmias and diseases of the cardiac conduction system, distinguishing them from other CVDs. Additionally, a knowledge graph analysis identified 59 of the 128 Ca2+-regulating proteins as involved in OS-related cardiac diseases, with cardiomyopathy emerging as the predominant category. By leveraging a link prediction algorithm, our research illuminates the interactions between Ca2+-regulating proteins, OS, and CVDs. The insights gained from our study provide a deeper understanding of the molecular interplay between cardiac ROS and Ca2+-regulating proteins in the context of CVDs. Such an understanding is essential for the innovation and development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Namuna Panday
- Department of Physiology, School of Medicine, University of California, Los Angeles, CA 90095, USA; (N.P.); (D.S.)
- NHLBI Integrated Cardiovascular Data Science Training Program (iDISCOVER), University of California, Los Angeles, CA 90095, USA; (I.A.); (J.R.); (A.V.); (A.N.E.)
| | - Dibakar Sigdel
- Department of Physiology, School of Medicine, University of California, Los Angeles, CA 90095, USA; (N.P.); (D.S.)
| | - Irsyad Adam
- NHLBI Integrated Cardiovascular Data Science Training Program (iDISCOVER), University of California, Los Angeles, CA 90095, USA; (I.A.); (J.R.); (A.V.); (A.N.E.)
| | - Joseph Ramirez
- NHLBI Integrated Cardiovascular Data Science Training Program (iDISCOVER), University of California, Los Angeles, CA 90095, USA; (I.A.); (J.R.); (A.V.); (A.N.E.)
| | - Aarushi Verma
- NHLBI Integrated Cardiovascular Data Science Training Program (iDISCOVER), University of California, Los Angeles, CA 90095, USA; (I.A.); (J.R.); (A.V.); (A.N.E.)
| | - Anirudh N. Eranki
- NHLBI Integrated Cardiovascular Data Science Training Program (iDISCOVER), University of California, Los Angeles, CA 90095, USA; (I.A.); (J.R.); (A.V.); (A.N.E.)
| | - Wei Wang
- Department of Computer Science, University of California, Los Angeles, CA 90095, USA;
- Department of Computational Medicine, University of California, Los Angeles, CA 90095, USA
- Scalable Analytics Institute (ScAi), University of California, Los Angeles, CA 90095, USA
- Department of Bioinformatics and Biomedical Informatics, University of California, Los Angeles, CA 90095, USA
| | - Ding Wang
- Department of Physiology, School of Medicine, University of California, Los Angeles, CA 90095, USA; (N.P.); (D.S.)
- NHLBI Integrated Cardiovascular Data Science Training Program (iDISCOVER), University of California, Los Angeles, CA 90095, USA; (I.A.); (J.R.); (A.V.); (A.N.E.)
| | - Peipei Ping
- Department of Physiology, School of Medicine, University of California, Los Angeles, CA 90095, USA; (N.P.); (D.S.)
- NHLBI Integrated Cardiovascular Data Science Training Program (iDISCOVER), University of California, Los Angeles, CA 90095, USA; (I.A.); (J.R.); (A.V.); (A.N.E.)
- Scalable Analytics Institute (ScAi), University of California, Los Angeles, CA 90095, USA
- Department of Bioinformatics and Biomedical Informatics, University of California, Los Angeles, CA 90095, USA
- Department of Medicine/Cardiology, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
36
|
Paudel R, Jafri MS, Ullah A. Gain-of-Function and Loss-of-Function Mutations in the RyR2-Expressing Gene Are Responsible for the CPVT1-Related Arrhythmogenic Activities in the Heart. Curr Issues Mol Biol 2024; 46:12886-12910. [PMID: 39590361 PMCID: PMC11592891 DOI: 10.3390/cimb46110767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/08/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Mutations in the ryanodine receptor (RyR2) gene have been linked to arrhythmia and possibly sudden cardiac death (SCD) during acute emotional stress, physical activities, or catecholamine perfusion. The most prevalent disorder is catecholaminergic polymorphic ventricular tachycardia (CPVT1). Four primary mechanisms have been proposed to describe CPVT1 with a RyR2 mutation: (a) gain-of-function, (b) destabilization of binding proteins, (c) store-overload-induced Ca2+ release (SOICR), and (d) loss of function. The goal of this study was to use computational models to understand these four mechanisms and how they might contribute to arrhythmia. To this end, we have developed a local control stochastic model of a ventricular cardiac myocyte and used it to investigate how the Ca2+ dynamics in the mutant RyR2 are responsible for the development of an arrhythmogenic episode under the condition of β-adrenergic (β-AR) stimulation or pauses afterward. Into the model, we have incorporated 20,000 distinct cardiac dyads consisting of stochastically gated L-type Ca2+ channels (LCCs) and ryanodine receptors (RyR2s) and the intervening dyadic cleft to analyze the alterations in Ca2+ dynamics. Recent experimental findings were incorporated into the model parameters to test these proposed mechanisms and their role in triggering arrhythmias. The model could not find any connection between SOICR and the destabilization of binding proteins as the arrhythmic mechanisms in the mutant myocyte. On the other hand, the model was able to observe loss-of-function and gain-of-function mutations resulting in EADs (Early Afterdepolarizations) and variations in action potential amplitudes and durations as the precursors to generate arrhythmia, respectively. These computational studies demonstrate how GOF and LOF mutations can lead to arrhythmia and cast doubt on the feasibility of SOICR as a mechanism of arrhythmia.
Collapse
Affiliation(s)
- Roshan Paudel
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
- School of Computer, Mathematical, and Natural Sciences, Morgan State University, Baltimore, MD 21251, USA
| | - Mohsin Saleet Jafri
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 20201, USA
| | - Aman Ullah
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
| |
Collapse
|
37
|
Paraskevaidis I, Kourek C, Farmakis D, Tsougos E. Heart Failure: A Deficiency of Energy-A Path Yet to Discover and Walk. Biomedicines 2024; 12:2589. [PMID: 39595155 PMCID: PMC11592498 DOI: 10.3390/biomedicines12112589] [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: 10/15/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
Heart failure is a complex syndrome and our understanding and therapeutic approach relies mostly on its phenotypic presentation. Notably, the heart is characterized as the most energy-consuming organ, being both a producer and consumer, in order to satisfy multiple cardiac functions: ion exchange, electromechanical coordination, excitation-contraction coupling, etc. By obtaining further knowledge of the cardiac energy field, we can probably better characterize the basic pathophysiological events occurring in heart disease patients and understand the metabolic substance changes, the relationship between the alteration of energy production/consumption, and hence energetic deficiency not only in the heart as a whole but in every single cardiac territory, which will hopefully provide us with the opportunity to uncover the beginning of the heart failure process. In this respect, using (a) newer imaging techniques, (b) biomedicine, (c) nanotechnology, and (d) artificial intelligence, we can gain a deeper understanding of this complex syndrome. This, in turn, can lead to earlier and more effective therapeutic approaches, ultimately improving human health. To date, the scientific community has not given sufficient attention to the energetic starvation model. In our view, this review aims to encourage scientists and the medical community to conduct studies for a better understanding and treatment of this syndrome.
Collapse
Affiliation(s)
- Ioannis Paraskevaidis
- 6th Department of Cardiology, Hygeia Hospital, 151 23 Athens, Greece; (I.P.); (E.T.)
| | - Christos Kourek
- Department of Cardiology, 417 Army Share Fund Hospital of Athens (NIMTS), 115 21 Athens, Greece;
| | - Dimitrios Farmakis
- Heart Failure Unit, Department of Cardiology, Attikon University Hospital, Medical School, National and Kapodistiran University of Athens, 124 62 Athens, Greece
| | - Elias Tsougos
- 6th Department of Cardiology, Hygeia Hospital, 151 23 Athens, Greece; (I.P.); (E.T.)
| |
Collapse
|
38
|
Pham LT, Mangmool S, Parichatikanond W. Sodium-Glucose Cotransporter 2 (SGLT2) Inhibitors: Guardians against Mitochondrial Dysfunction and Endoplasmic Reticulum Stress in Heart Diseases. ACS Pharmacol Transl Sci 2024; 7:3279-3298. [PMID: 39539254 PMCID: PMC11555527 DOI: 10.1021/acsptsci.4c00240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 09/11/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024]
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors are an innovative class of antidiabetic drugs that provide cardiovascular benefits to both diabetic and nondiabetic patients, surpassing those of other antidiabetic drugs. Although the roles of mitochondria and endoplasmic reticulum (ER) in cardiovascular research are increasingly recognized as promising therapeutic targets, the exact molecular mechanisms by which SGLT2 inhibitors influence mitochondrial and ER homeostasis in the heart remain incompletely elucidated. This review comprehensively summarizes and discusses the impacts of SGLT2 inhibitors on mitochondrial dysfunction and ER stress in heart diseases including heart failure, ischemic heart disease/myocardial infarction, and arrhythmia from preclinical and clinical studies. Based on the existing evidence, the effects of SGLT2 inhibitors may potentially involve the restoration of mitochondrial biogenesis and alleviation of ER stress. Such consequences are achieved by enhancing adenosine triphosphate (ATP) production, preserving mitochondrial membrane potential, improving the activity of electron transport chain complexes, maintaining mitochondrial dynamics, mitigating oxidative stress and apoptosis, influencing cellular calcium and sodium handling, and targeting the unfolded protein response (UPR) through three signaling pathways including inositol requiring enzyme 1α (IRE1α), protein kinase R like endoplasmic reticulum kinase (PERK), and activating transcription factor 6 (ATF6). Therefore, SGLT2 inhibitors have emerged as a promising target for treating heart diseases due to their potential to improve mitochondrial functions and ER stress.
Collapse
Affiliation(s)
- Linh Thi
Truc Pham
- Biopharmaceutical
Sciences Program, Faculty of Pharmacy, Mahidol
University, Bangkok, 10400 Thailand
- Department
of Pharmacology, Faculty of Pharmacy, Mahidol
University, Bangkok, 10400 Thailand
| | - Supachoke Mangmool
- Department
of Pharmaceutical Care, Faculty of Pharmacy, Chiang Mai University, Chiang
Mai, 50200 Thailand
| | | |
Collapse
|
39
|
Tripoli BA, Smyth JT. Septins regulate heart contractility through modulation of cardiomyocyte store-operated calcium entry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.621876. [PMID: 39574715 PMCID: PMC11580947 DOI: 10.1101/2024.11.04.621876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
Highly regulated cardiomyocyte Ca 2+ fluxes drive heart contractions. Recent findings from multiple organisms demonstrate that the specific Ca 2+ transport mechanism known as store-operated Ca 2+ entry (SOCE) is essential in cardiomyocytes for proper heart function, and SOCE dysregulation results in cardiomyopathy. Mechanisms that regulate SOCE in cardiomyocytes are poorly understood. Here we tested the role of cytoskeletal septin proteins in cardiomyocyte SOCE regulation. Septins are essential SOCE modulators in other cell types, but septin functions in cardiomyocytes are nearly completely unexplored. We show using targeted genetics and intravital imaging of heart contractility in Drosophila that cardiomyocyte-specific depletion of septins 1, 2, and 4 results in heart dilation that phenocopies the effects of SOCE suppression. Heart dilation caused by septin 2 depletion was suppressed by SOCE upregulation, supporting the hypothesis that septin 2 is required in cardiomyocytes for sufficient SOCE function. A major function of SOCE is to support SERCA-dependent sarco/endoplasmic reticulum (S/ER) Ca 2+ stores, and augmenting S/ER store filling by SERCA overexpression also suppressed the septin 2 phenotype. We also ruled out several potential SOCE-independent septin functions, as septin 2 phenotypes were not due to septin function during development and septin 2 was not required for z-disk organization as defined by α-actinin labeling. These results demonstrate, for the first time, an essential role of septins in cardiomyocyte physiology and heart function that is due, at least in part, to septin regulation of SOCE function.
Collapse
|
40
|
Hirakis SP, Bartol TM, Autin L, Amaro RE, Sejnowski TJ. Electrophysical cardiac remodeling at the molecular level: Insights into ryanodine receptor activation and calcium-induced calcium release from a stochastic explicit-particle model. Biophys J 2024; 123:3812-3831. [PMID: 39369273 PMCID: PMC11560313 DOI: 10.1016/j.bpj.2024.09.029] [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: 04/28/2023] [Revised: 07/03/2024] [Accepted: 09/26/2024] [Indexed: 10/07/2024] Open
Abstract
We present the first-ever, fully discrete, stochastic model of triggered cardiac Ca2+ dynamics. Using anatomically accurate subcellular cardiac myocyte geometries, we simulate the molecular players involved in Ca2+ handling using high-resolution stochastic and explicit-particle methods at the level of an individual cardiac dyadic junction. Integrating data from multiple experimental sources, the model not only replicates the findings of traditional in silico studies and complements in vitro experimental data but also reveals new insights into the molecular mechanisms driving cardiac dysfunction under stress and disease conditions. We improve upon older, nondiscrete models using the same realistic geometry by incorporating molecular mechanisms for spontaneous, as well as triggered calcium-induced calcium release (CICR). Action potentials are used to activate L-type calcium channels (LTCC), triggering CICR through ryanodine receptors (RyRs) on the surface of the sarcoplasmic reticulum. These improvements allow for the specific focus on the couplon: the structure-function relationship between LTCC and RyR. We investigate the electrophysical effects of normal and diseased action potentials on CICR and interrogate the effects of dyadic junction deformation through detubulation and orphaning of RyR. Our work demonstrates the importance of the electrophysical integrity of the calcium release unit on CICR fidelity, giving insights into the molecular basis of heart disease. Finally, we provide a unique, detailed, molecular view of the CICR process using advanced rendering techniques. This easy-to-use model comes complete with tutorials and the necessary software for use and analysis to maximize usability and reproducibility. Our work focuses on quantifying, qualifying, and visualizing the behavior of the molecular species that underlie the function and dysfunction of subcellular cardiomyocyte systems.
Collapse
Affiliation(s)
- Sophia P Hirakis
- Computational Neurobiology Lab, The Salk Institute of Biological Studies, La Jolla, California; Department of Chemistry and Biochemistry, The University of California San Diego, La Jolla, California
| | - Thomas M Bartol
- Computational Neurobiology Lab, The Salk Institute of Biological Studies, La Jolla, California
| | - Ludovic Autin
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California
| | - Rommie E Amaro
- Department of Chemistry and Biochemistry, The University of California San Diego, La Jolla, California.
| | - Terrence J Sejnowski
- Computational Neurobiology Lab, The Salk Institute of Biological Studies, La Jolla, California; Department of Chemistry and Biochemistry, The University of California San Diego, La Jolla, California.
| |
Collapse
|
41
|
Yang Y, Valencia LA, Lu CH, Nakamoto ML, Tsai CT, Liu C, Yang H, Zhang W, Jahed Z, Lee WR, Santoro F, Liou J, Wu JC, Cui B. Plasma membrane curvature regulates the formation of contacts with the endoplasmic reticulum. Nat Cell Biol 2024; 26:1878-1891. [PMID: 39289582 PMCID: PMC11567891 DOI: 10.1038/s41556-024-01511-x] [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: 11/20/2023] [Accepted: 08/19/2024] [Indexed: 09/19/2024]
Abstract
Contact sites between the endoplasmic reticulum (ER) and plasma membrane (PM) play a crucial role in governing calcium regulation and lipid homeostasis. Despite their significance, the factors regulating their spatial distribution on the PM remain elusive. Inspired by observations in cardiomyocytes, where ER-PM contact sites concentrate on tubular PM invaginations known as transverse tubules, we hypothesize that PM curvature plays a role in ER-PM contact formation. Through precise control of PM invaginations, we show that PM curvatures locally induce the formation of ER-PM contacts in cardiomyocytes. Intriguingly, the junctophilin family of ER-PM tethering proteins, specifically expressed in excitable cells, is the key player in this process, whereas the ubiquitously expressed extended synaptotagmin-2 does not show a preference for PM curvature. At the mechanistic level, we find that the low-complexity region (LCR) and membrane occupation and recognition nexus (MORN) motifs of junctophilins can bind independently to the PM, but both the LCR and MORN motifs are required for targeting PM curvatures. By examining the junctophilin interactome, we identify a family of curvature-sensing proteins-Eps15 homology domain-containing proteins-that interact with the MORN_LCR motifs and facilitate the preferential tethering of junctophilins to curved PM. These findings highlight the pivotal role of PM curvature in the formation of ER-PM contacts in cardiomyocytes and unveil a mechanism for the spatial regulation of ER-PM contacts through PM curvature modulation.
Collapse
Affiliation(s)
- Yang Yang
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Luis A Valencia
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Chih-Hao Lu
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Melissa L Nakamoto
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Ching-Ting Tsai
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Chun Liu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Departments of Physiology and Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Huaxiao Yang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Department of Biomedical Engineering, University of North Texas, Denton, TX, USA
| | - Wei Zhang
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Zeinab Jahed
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Department of Chemical and Nano Engineering, University of California, San Diego, San Diego, CA, USA
| | - Wan-Ru Lee
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Francesca Santoro
- Tissue Electronics, Istituto Italiano di Tecnologia, Naples, Italy
- Faculty of Electrical Engineering and Information Technology, RWTH Aachen University, Aachen, Germany
- Institute of Biological Information Processing-Bioelectronics (IBI-3), Forschungszentrum, Jülich, Germany
| | - Jen Liou
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Cardiology, Stanford University, Stanford, CA, USA
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
42
|
Zhang X, Wu Y, Smith CER, Louch WE, Morotti S, Dobrev D, Grandi E, Ni H. Enhanced Ca 2+-Driven Arrhythmogenic Events in Female Patients With Atrial Fibrillation: Insights From Computational Modeling. JACC Clin Electrophysiol 2024; 10:2371-2391. [PMID: 39340505 PMCID: PMC11602355 DOI: 10.1016/j.jacep.2024.07.020] [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: 04/22/2024] [Revised: 07/10/2024] [Accepted: 07/29/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Substantial sex-based differences have been reported in atrial fibrillation (AF), but the underlying mechanisms are poorly understood. OBJECTIVES This study sought to gain a mechanistic understanding of Ca2+-handling disturbances and Ca2+-driven arrhythmogenic events in male vs female atrial cardiomyocytes and establish their responses to Ca2+-targeted interventions. METHODS We integrated reported sex differences and AF-associated changes (ie, expression and phosphorylation of Ca2+-handling proteins, cardiomyocyte ultrastructural characteristics, and dimensions) into our human atrial cardiomyocyte model that couples electrophysiology with spatially detailed Ca2+-handling processes. Sex-specific responses of atrial cardiomyocytes to arrhythmia-provoking protocols and Ca2+-targeted interventions were evaluated. RESULTS Simulated quiescent cardiomyocytes showed increased incidence of Ca2+ sparks in female vs male myocytes in AF, in agreement with previous experimental reports. Additionally, our female model exhibited elevated propensity to develop pacing-induced spontaneous Ca2+ releases (SCRs) and augmented beat-to-beat variability in action potential (AP)-elicited Ca2+ transients compared with the male model. Sensitivity analysis uncovered distinct arrhythmogenic contributions of each component involved in sex and/or AF alterations. Specifically, increased ryanodine receptor phosphorylation emerged as the major SCR contributor in female AF cardiomyocytes, whereas reduced L-type Ca2+ current was protective against SCRs for male AF cardiomyocytes. Furthermore, simulated Ca2+-targeted interventions identified potential strategies (eg, t-tubule restoration, and inhibition of ryanodine receptor and sarcoplasmic/endoplasmic reticulum Ca2⁺-ATPase) to attenuate Ca2+-driven arrhythmogenic events in women, and revealed enhanced efficacy when applied in combination. CONCLUSIONS Sex-specific modeling uncovers increased Ca2+-driven arrhythmogenic events in female vs male atria in AF, and suggests combined Ca2+-targeted interventions are promising therapeutic approaches in women.
Collapse
Affiliation(s)
- Xianwei Zhang
- Department of Pharmacology, University of California-Davis, Davis, California, USA. https://twitter.com/xianweizhang1
| | - Yixuan Wu
- Department of Pharmacology, University of California-Davis, Davis, California, USA
| | - Charlotte E R Smith
- Department of Pharmacology, University of California-Davis, Davis, California, USA. https://twitter.com/Char_Smith3
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway. https://twitter.com/IEMRLouch
| | - Stefano Morotti
- Department of Pharmacology, University of California-Davis, Davis, California, USA. https://twitter.com/MorottiLab
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany; Montréal Heart Institute, Université de Montréal, Montréal, Québec, Canada; Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA. https://twitter.com/dr_dobrev
| | - Eleonora Grandi
- Department of Pharmacology, University of California-Davis, Davis, California, USA.
| | - Haibo Ni
- Department of Pharmacology, University of California-Davis, Davis, California, USA.
| |
Collapse
|
43
|
Huang J, Pan X, Yan N. Structural biology and molecular pharmacology of voltage-gated ion channels. Nat Rev Mol Cell Biol 2024; 25:904-925. [PMID: 39103479 DOI: 10.1038/s41580-024-00763-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2024] [Indexed: 08/07/2024]
Abstract
Voltage-gated ion channels (VGICs), including those for Na+, Ca2+ and K+, selectively permeate ions across the cell membrane in response to changes in membrane potential, thus participating in physiological processes involving electrical signalling, such as neurotransmission, muscle contraction and hormone secretion. Aberrant function or dysregulation of VGICs is associated with a diversity of neurological, psychiatric, cardiovascular and muscular disorders, and approximately 10% of FDA-approved drugs directly target VGICs. Understanding the structure-function relationship of VGICs is crucial for our comprehension of their working mechanisms and role in diseases. In this Review, we discuss how advances in single-particle cryo-electron microscopy have afforded unprecedented structural insights into VGICs, especially on their interactions with clinical and investigational drugs. We present a comprehensive overview of the recent advances in the structural biology of VGICs, with a focus on how prototypical drugs and toxins modulate VGIC activities. We explore how these structures elucidate the molecular basis for drug actions, reveal novel pharmacological sites, and provide critical clues to future drug discovery.
Collapse
Affiliation(s)
- Jian Huang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Xiaojing Pan
- Institute of Bio-Architecture and Bio-Interactions (IBABI), Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen, Guangdong, China.
| | - Nieng Yan
- Institute of Bio-Architecture and Bio-Interactions (IBABI), Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen, Guangdong, China.
- Beijing Frontier Research Center for Biological Structure, Tsinghua-Peking Joint Center for Life Sciences, State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, China.
| |
Collapse
|
44
|
Pizzo E, Cervantes DO, Ripa V, Filardo A, Berrettoni S, Ketkar H, Jagana V, Di Stefano V, Singh K, Ezzati A, Ghadirian K, Kouril A, Jacobson JT, Bisserier M, Jain S, Rota M. The cAMP/PKA signaling pathway conditions cardiac performance in experimental animals with metabolic syndrome. J Mol Cell Cardiol 2024; 196:35-51. [PMID: 39251059 PMCID: PMC11534532 DOI: 10.1016/j.yjmcc.2024.09.002] [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] [Received: 12/23/2023] [Revised: 07/20/2024] [Accepted: 09/05/2024] [Indexed: 09/11/2024]
Abstract
Metabolic syndrome (MetS) increases the risk of coronary artery disease, but effects of this condition on the working myocardium remain to be fully elucidated. In the present study we evaluated the consequences of diet-induced metabolic disorders on cardiac function and myocyte performance using female mice fed with Western diet. Animals maintained on regular chow were used as control (Ctrl). Mice on the Western diet (WesD) had increased body weight, impaired glucose metabolism, preserved diastolic and systolic function, but increased left ventricular (LV) mass, with respect to Ctrl animals. Moreover, WesD mice had reduced heart rate variability (HRV), indicative of altered cardiac sympathovagal balance. Myocytes from WesD mice had increased volume, enhanced cell mechanics, and faster kinetics of contraction and relaxation. Moreover, levels of cAMP and protein kinase A (PKA) activity were enhanced in WesD myocytes, and interventions aimed at stabilizing cAMP/PKA abrogated functional differences between Ctrl and WesD cells. Interestingly, in vivo β-adrenergic receptor (β-AR) blockade normalized the mechanical properties of WesD myocytes and revealed defective cardiac function in WesD mice, with respect to Ctrl. Collectively, these results indicate that metabolic disorders induced by Western diet enhance the cAMP/PKA signaling pathway, a possible adaptation required to maintain cardiac function.
Collapse
Affiliation(s)
- Emanuele Pizzo
- Department of Physiology, New York Medical College, Valhalla, NY, USA
| | | | - Valentina Ripa
- Department of Physiology, New York Medical College, Valhalla, NY, USA
| | - Andrea Filardo
- Department of Physiology, New York Medical College, Valhalla, NY, USA
| | - Silvia Berrettoni
- Department of Physiology, New York Medical College, Valhalla, NY, USA
| | - Harshada Ketkar
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Vineeta Jagana
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| | | | - Kanwardeep Singh
- Department of Physiology, New York Medical College, Valhalla, NY, USA
| | - Asha Ezzati
- Department of Physiology, New York Medical College, Valhalla, NY, USA
| | - Kash Ghadirian
- Department of Physiology, New York Medical College, Valhalla, NY, USA
| | - Anna Kouril
- Department of Physiology, New York Medical College, Valhalla, NY, USA
| | - Jason T Jacobson
- Department of Physiology, New York Medical College, Valhalla, NY, USA; Department of Cardiology, Westchester Medical Center, Valhalla, NY, USA
| | - Malik Bisserier
- Department of Physiology, New York Medical College, Valhalla, NY, USA; Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| | - Sudhir Jain
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Marcello Rota
- Department of Physiology, New York Medical College, Valhalla, NY, USA.
| |
Collapse
|
45
|
del Rivero Morfin PJ, Chavez DS, Jayaraman S, Yang L, Geisler SM, Kochiss AL, Tuluc P, Colecraft HM, Marx SO, Liu XS, Rajadhyaksha AM, Ben-Johny M. A genetically encoded actuator boosts L-type calcium channel function in diverse physiological settings. SCIENCE ADVANCES 2024; 10:eadq3374. [PMID: 39475605 PMCID: PMC11524184 DOI: 10.1126/sciadv.adq3374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/24/2024] [Indexed: 11/02/2024]
Abstract
L-type Ca2+ channels (CaV1.2/1.3) convey influx of calcium ions that orchestrate a bevy of biological responses including muscle contraction, neuronal function, and gene transcription. Deficits in CaV1 function play a vital role in cardiac and neurodevelopmental disorders. Here, we develop a genetically encoded enhancer of CaV1.2/1.3 channels (GeeCL) to manipulate Ca2+ entry in distinct physiological settings. We functionalized a nanobody that targets the CaV complex by attaching a minimal effector domain from an endogenous CaV modulator-leucine-rich repeat containing protein 10 (Lrrc10). In cardiomyocytes, GeeCL selectively increased L-type current amplitude. In neurons in vitro and in vivo, GeeCL augmented excitation-transcription (E-T) coupling. In all, GeeCL represents a powerful strategy to boost CaV1.2/1.3 function and lays the groundwork to illuminate insights on neuronal and cardiac physiology and disease.
Collapse
Affiliation(s)
| | - Diego Scala Chavez
- Pediatric Neurology, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Srinidhi Jayaraman
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Lin Yang
- Division of Cardiology, Department of Medicine, Columbia University, Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Stefanie M. Geisler
- Department of Pharmacology and Toxicology, University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Audrey L. Kochiss
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Petronel Tuluc
- Department of Pharmacology and Toxicology, University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Henry M. Colecraft
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY, USA
| | - Steven O. Marx
- Division of Cardiology, Department of Medicine, Columbia University, Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY, USA
| | - X. Shawn Liu
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
| | - Anjali M. Rajadhyaksha
- Pediatric Neurology, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Weill Cornell Autism Research Program, Weill Cornell Medicine, New York, NY, USA
| | - Manu Ben-Johny
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| |
Collapse
|
46
|
Birkedal R, Branovets J, Vendelin M. Compartmentalization in cardiomyocytes modulates creatine kinase and adenylate kinase activities. FEBS Lett 2024; 598:2623-2640. [PMID: 39112921 DOI: 10.1002/1873-3468.14994] [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: 03/14/2024] [Revised: 06/03/2024] [Accepted: 07/21/2024] [Indexed: 11/12/2024]
Abstract
Intracellular molecules are transported by motor proteins or move by diffusion resulting from random molecular motion. Cardiomyocytes are packed with structures that are crucial for function, but also confine the diffusional spaces, providing cells with a means to control diffusion. They form compartments in which local concentrations are different from the overall, average concentrations. For example, calcium and cyclic AMP are highly compartmentalized, allowing these versatile second messengers to send different signals depending on their location. In energetic compartmentalization, the ratios of AMP and ADP to ATP are different from the average ratios. This is important for the performance of ATPases fuelling cardiac excitation-contraction coupling and mechanical work. A recent study suggested that compartmentalization modulates the activity of creatine kinase and adenylate kinase in situ. This could have implications for energetic signaling through, for example, AMP-activated kinase. It highlights the importance of taking compartmentalization into account in our interpretation of cellular physiology and developing methods to assess local concentrations of AMP and ADP to enhance our understanding of compartmentalization in different cell types.
Collapse
Affiliation(s)
- Rikke Birkedal
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Estonia
| | - Jelena Branovets
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Estonia
| | - Marko Vendelin
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Estonia
| |
Collapse
|
47
|
Scott B, Greenberg L, Squarci C, Campbell KS, Greenberg MJ. Danicamtiv reduces myosin's working stroke but enhances contraction by activating the thin filament. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.09.617269. [PMID: 39416013 PMCID: PMC11482770 DOI: 10.1101/2024.10.09.617269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Heart failure is a leading cause of death worldwide, and even with current treatments, the 5-year transplant-free survival rate is only ~50-70%. As such, there is a need to develop new treatments for patients that improve survival and quality of life. Recently, there have been efforts to develop small molecules for heart failure that directly target components of the sarcomere, including cardiac myosin. One such molecule, danicamtiv, recently entered phase II clinical trials; however, its mechanism of action and direct effects on myosin's mechanics and kinetics are not well understood. Using optical trapping techniques, stopped flow transient kinetics, and in vitro reconstitution assays, we found that danicamtiv reduces the size of cardiac myosin's working stroke, and in contrast to studies in muscle fibers, we found that it does not affect actomyosin detachment kinetics at the level of individual crossbridges. We demonstrate that danicamtiv accelerates actomyosin association kinetics, leading to increased recruitment of myosin crossbridges and subsequent thin filament activation at physiologically-relevant calcium concentrations. Finally, we computationally model how the observed changes in mechanics and kinetics at the level of single crossbridges contribute to increased cardiac contraction and improved diastolic function compared to the related myotrope, omecamtiv mecarbil. Taken together, our results have important implications for the design of new sarcomeric-targeting compounds for heart failure.
Collapse
Affiliation(s)
- Brent Scott
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Lina Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Caterina Squarci
- Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY, 40506, USA
| | - Kenneth S. Campbell
- Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY, 40506, USA
| | - Michael J. Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| |
Collapse
|
48
|
Xu F, Li JJ, Yang E, Zhang Y, Xie W. Assaying sarcoplasmic reticulum Ca 2+-leak in mouse atrial myocytes. BIOPHYSICS REPORTS 2024; 10:297-303. [PMID: 39539281 PMCID: PMC11554581 DOI: 10.52601/bpr.2023.230044] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 12/26/2023] [Indexed: 11/16/2024] Open
Abstract
More and more studies have suggested an essential role of sarcoplasmic reticulum (SR) Ca2+ leak of atrial myocytes in atrial diseases such as atrial fibrillation (AF). The increasing interest in atrial Ca2+ signaling makes it necessary to develop a more accurate approach for Ca2+ measurement in atrial myocytes due to obvious differences between atrial and ventricular Ca2+ handling. In the present study, we proposed a new approach for quantifying total SR Ca2+ leak in atrial myocytes with confocal line-scan Ca2+ images. With a very precious approximation of the histogram of normalized line-scan Ca2+ images by using a modified Gaussian distribution, we separated the signal pixel components from noisy pixels and extracted two new dimensionless parameters, F signals and R signals, to reflect the summation of signal pixels and their release components, respectively. In the presence of tetracaine blocking SR Ca2+ leak, the two parameters were very close to 0, and in atrial myocytes under normal conditions, the two parameters are well positive correlative with Ca2+ spark frequency and total signal mass, the two classic readouts for SR Ca2+ leak. Consistent with Ca2+ Spark readouts, the two parameters quantified a significant increase of SR Ca2+ leak in atrial myocytes from mice harboring a leaky type 2 ryanodine receptor mutation (RyR2-R2474S+/-) compared to the WT group. Collectively, this study proposed a simple and effective approach to quantify SR Ca2+ leak in atrial myocytes, which may benefit research on calcium signaling in atrial physiology and diseases.
Collapse
Affiliation(s)
- Fan Xu
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710061, China
| | - Jing-Jing Li
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, Xi’an 710061, China
| | - Eric Yang
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, Xi’an 710061, China
| | - Yi Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Wenjun Xie
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, Xi’an 710061, China
| |
Collapse
|
49
|
Yang B, Wang SQ, Yang HQ. β-adrenergic regulation of Ca 2+ signaling in heart cells. BIOPHYSICS REPORTS 2024; 10:274-282. [PMID: 39539286 PMCID: PMC11554573 DOI: 10.52601/bpr.2024.240906] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/15/2024] [Indexed: 11/16/2024] Open
Abstract
β-adrenergic receptors (βARs) play significant roles in regulating Ca2+ signaling in cardiac myocytes, thus holding a key function in modulating heart performance. βARs regulate the influx of extracellular Ca2+ and the release and uptake of Ca2+ from the sarcoplasmic reticulum (SR) by activating key components such as L-type calcium channels (LTCCs), ryanodine receptors (RyRs) and phospholamban (PLN), mediated by the phosphorylation actions by protein kinase A (PKA). In cardiac myocytes, the presence of β2AR provides a protective mechanism against potential overstimulation of β1AR, which may aid in the restoration of cardiac dysfunctions. Understanding the Ca2+ regulatory signaling pathways of βARs in cardiac myocytes and the differences among various βAR subtypes are crucial in cardiology and hold great potential for developing treatments for heart diseases.
Collapse
Affiliation(s)
- Bo Yang
- Cyrus Tang Medical Institute, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| | - Shi-Qiang Wang
- State Key Lab of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Hua-Qian Yang
- Cyrus Tang Medical Institute, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
50
|
Su Y, Liang Y, Xu M, Gao B, Zhang S, Yang E, Yin S, Li D, Huang Z, Xie W. Modeling sarcoplasmic reticulum Ca 2+ in rat cardiomyocytes. BIOPHYSICS REPORTS 2024; 10:328-335. [PMID: 39539287 PMCID: PMC11554579 DOI: 10.52601/bpr.2024.240012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 07/10/2024] [Indexed: 11/16/2024] Open
Abstract
The sarcoplasmic reticulum (SR) primarily serves as the intracellular Ca2+ store in cardiac myocytes, mediating cellular function under cardiac physiology and diseases. However, the properties of cardiac SR Ca2+ have not yet been fully determined, particularly in rats and mice, which are the most commonly used experimental species in studies on cardiac physiology and diseases. Here, we developed a spatially detailed numerical model to deduce Ca2+ movements inside the junctional SR (jSR) cisternae of rat cardiomyocytes. Our model accurately reproduced the jSR Ca2+ kinetics of local and global SR Ca2+ releases reported in a recent experimental study. With this model, we revealed that jSR Ca2+ kinetics was mostly determined by the total release flux via type 2 ryanodine receptor (RyR2) channels but not by RyR2 positioning. Although Ca2+ diffusion in global SR was previously reported to be slow, our simulation demonstrated that Ca2+ diffused very quickly inside local jSR cisternae and the decrease in the diffusion coefficient resulted in a significant reduction of jSR Ca2+ depletion amplitude. Intracellular Ca2+ was typically experimentally detected with fluorescence dye. Our simulation revealed that when the dynamical characteristics of fluorescence dye exerted a minimal effect on actual Ca2+ mobility inside jSR, the reaction rate of the dye with Ca2+ could significantly affect apparent jSR Ca2+ kinetics. Therefore, loading a chemical fluorescence dye with fast kinetics, such as Fluo-5N, into SR is important for Ca2+ measurement inside SR. Overall, our model provides new insights into deciphering Ca2+ handling inside nanoscopic jSR cisternae in rat cardiomyocytes.
Collapse
Affiliation(s)
- Yutong Su
- Beijing Engineering Research Center for IoT Software and Systems, Beijing University of Technology, Beijing 100124, China
| | - Yongshen Liang
- Beijing Engineering Research Center for IoT Software and Systems, Beijing University of Technology, Beijing 100124, China
| | - Menghao Xu
- Beijing Engineering Research Center for IoT Software and Systems, Beijing University of Technology, Beijing 100124, China
| | - Beibei Gao
- Beijing Engineering Research Center for IoT Software and Systems, Beijing University of Technology, Beijing 100124, China
| | - Siyuan Zhang
- Beijing Engineering Research Center for IoT Software and Systems, Beijing University of Technology, Beijing 100124, China
| | - Eric Yang
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, Xi’an 710061, China
| | - Shuai Yin
- Beijing Engineering Research Center for IoT Software and Systems, Beijing University of Technology, Beijing 100124, China
| | - Da Li
- Beijing Engineering Research Center for IoT Software and Systems, Beijing University of Technology, Beijing 100124, China
| | - Zhangqin Huang
- Beijing Engineering Research Center for IoT Software and Systems, Beijing University of Technology, Beijing 100124, China
| | - Wenjun Xie
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, Xi’an 710061, China
| |
Collapse
|