1
|
Lakatta EG. Heart Rhythm Harmony Becomes Discordant as We Age. Heart Lung Circ 2025:S1443-9506(25)00324-5. [PMID: 40355300 DOI: 10.1016/j.hlc.2025.04.084] [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: 04/23/2025] [Accepted: 04/23/2025] [Indexed: 05/14/2025]
Abstract
Heartbeats are initiated by pacemaker cells within the sinoatrial node (SAN) that generate spontaneous impulses at intervals that resonate around a preferred mean frequency. A coupled-clock system (CCS) intrinsic to individual pacemaker cells, that is modulated by autonomic input, drives SAN normal automaticity. Subcellular and cell-wide mechanisms within the CCS are in "dynamic equilibrium," and never achieve a true steady state. Nanoscale electromagnetic "vibrations" caused by mechanisms intrinsic to the CCS and their autonomic modulation create heartbeat rhythm, ("heartbeat music"). A "Heart-Brain Grand Symphony" (HBGS), that emerges from this "beautiful noise" as the heart beats, is broadcast to the body surface, and its numerous motifs within the symphony can be experienced by tuning into electrocardiogram (EKG) RR-interval variability rhythms. As age increases, one or more of the components of physiologic coupling within the neuroautonomic regulatory sinus node and atrial networks begins to deteriorate, and cacophony emerges within the HBGS, manifested by reductions in the mean rate and rhythm at which the CCS within SAN cells fires action potentials. These subclinical changes in SAN structure and function as age advances become "partners" with pathophysiology that defines clinical SAN and other cardiac tissue diseases, e.g., Sick Sinus Syndrome and atrial fibrillation, and as such age-associated changes in SAN structure and function are co-morbidities of these clinical cardiac diseases. In other terms as age advances, sub-clinical age-associated changes in SAN structure and function, per se, are major shareholders in SAN disease enterprises.
Collapse
Affiliation(s)
- Edward G Lakatta
- Laboratory of Cardiovascular Science Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
2
|
Wu Y, Wang Q, Granger J, Reyes Gaido O, Lopez-Cecetaite G, Aguilar EN, Ludwig A, Moroni A, Bianchet MA, Anderson ME. HCN4 channels sense temperature and determine heart rate responses to heat. Nat Commun 2025; 16:2102. [PMID: 40025061 PMCID: PMC11873294 DOI: 10.1038/s41467-025-57358-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 02/18/2025] [Indexed: 03/04/2025] Open
Abstract
The hyperpolarization-activated cyclic nucleotide-gated ion channel 4 (HCN4) current increases due to cAMP binding and is well-recognized to contribute to adrenergically driven heart rate acceleration. HCN4 current also increases with heat by an unknown mechanism(s). We use thermodynamical and homology computational modeling, site-directed mutagenesis, and mouse models to identify a concise motif on the S4-S5 linker of HCN4 channels (M407/Y409) that determines HCN4 current (If) responses to heat. This motif is required for heat-triggered rate acceleration in cardiac pacemaker cells, isolated hearts and in vivo. Surprisingly, a loss of function M407/Y409 motif mutation prevented not only normal heat but also cAMP responses, suggesting that the heat-sensing machinery within the S4-S5 linker is essential for operating the cAMP allosteric pathway and is central to HCN4 gating modulation. The M407/Y409 motif is conserved across all HCN family members suggesting that HCN channels participate broadly in coupling heat to changes in cell membrane excitability.
Collapse
Affiliation(s)
- Yuejin Wu
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| | - Qinchuan Wang
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jonathan Granger
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Oscar Reyes Gaido
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Eric Nunez Aguilar
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andreas Ludwig
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Anna Moroni
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy
| | - Mario A Bianchet
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mark E Anderson
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Medicine and Division of Biological Sciences, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
3
|
Yang S, Xue J, Li Z, Zhang S, Zhang Z, Huang Z, Yung KKL, Lai KWC. Deep Learning-Based Ion Channel Kinetics Analysis for Automated Patch Clamp Recording. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2404166. [PMID: 39737527 PMCID: PMC12083860 DOI: 10.1002/advs.202404166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/07/2024] [Indexed: 01/01/2025]
Abstract
The patch clamp technique is a fundamental tool for investigating ion channel dynamics and electrophysiological properties. This study proposes the first artificial intelligence framework for characterizing multiple ion channel kinetics of whole-cell recordings. The framework integrates machine learning for anomaly detection and deep learning for multi-class classification. The anomaly detection excludes recordings that are incompatible with ion channel behavior. The multi-class classification combined a 1D convolutional neural network, bidirectional long short-term memory, and an attention mechanism to capture the spatiotemporal patterns of the recordings. The framework achieves an accuracy of 97.58% in classifying 124 test datasets into six categories based on ion channel kinetics. The utility of the novel framework is demonstrated in two applications: Alzheimer's disease drug screening and nanomatrix-induced neuronal differentiation. In drug screening, the framework illustrates the inhibitory effects of memantine on endogenous channels, and antagonistic interactions among potassium, magnesium, and calcium ion channels. For nanomatrix-induced differentiation, the classifier indicates the effects of differentiation conditions on sodium and potassium channels associated with action potentials, validating the functional properties of differentiated neurons for Parkinson's disease treatment. The proposed framework is promising for enhancing the efficiency and accuracy of ion channel kinetics analysis in electrophysiological research.
Collapse
Affiliation(s)
- Shengjie Yang
- Department of Biomedical EngineeringCity University of Hong KongTat Chee Avenue, Kowloon TongKowloonHong Kong SARChina
| | - Jiaqi Xue
- Department of Biomedical EngineeringCity University of Hong KongTat Chee Avenue, Kowloon TongKowloonHong Kong SARChina
| | - Ziqi Li
- Department of Biomedical EngineeringCity University of Hong KongTat Chee Avenue, Kowloon TongKowloonHong Kong SARChina
| | - Shiqing Zhang
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug ResearchCollege of PharmacyJinan University601 West Huangpu Road, TianheGuangzhou510632China
| | - Zhang Zhang
- School of Public HealthGuangzhou Medical UniversityXinzao, PanyuGuangzhou511436China
| | - Zhifeng Huang
- Department of ChemistryChinese University of Hong KongShatinNew TerritoriesHong Kong SARChina
| | - Ken Kin Lam Yung
- Department of Science and Environmental StudiesEducation University of Hong Kong10 Lo Ping RoadTai PoNew TerritoriesHong Kong SARChina
| | - King Wai Chiu Lai
- Department of Biomedical EngineeringCity University of Hong KongTat Chee Avenue, Kowloon TongKowloonHong Kong SARChina
| |
Collapse
|
4
|
Copier JS, Verkerk AO, Lodder EM. HCN4 in the atrioventricular node. Heart Rhythm 2025:S1547-5271(25)00200-0. [PMID: 39988103 DOI: 10.1016/j.hrthm.2025.02.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 02/25/2025]
Abstract
Hyperpolarization-activated cyclic nucleotide-gated channel 4 (HCN4) drives the funny current in cardiac pacemaker regions. Its involvement in sinoatrial node pacemaker generation is well known, but its function in the atrioventricular (AV) node (AVN) has not intensively been studied. HCN4 is expressed in the AVN, and its expression within the AVN seems similar across mammalian species with HCN4 presence in the inferior nodal extensions, compact node, and AV bundle. The main direct regulators of HCN4 are cAMP and protein kinase A. In addition, indirect regulators may affect HCN4 via trafficking and localization. However, these effects are underexplored in the AVN. AVN-specific effects in knockout and knockin mice include reduced funny current density and increased AV block. HCN4 expression in the AVN could be affected by aging, exercise, heart failure, and diabetes. This could underlie changes in PR interval, atria-His interval, Wenckebach cycle length, and AVN effective refractory period. Clinical reports link the HCN4 variant G1097W to AV block. Other clinical data come from studies assessing ivabradine, an HCN4 inhibitor. In animals, ivabradine resulted in prolonged PR and atrial-his intervals. To date, uncertainty regarding the role of HCN4 in the AVN remains. However, AVN-focused studies suggest HCN4's importance for AVN function. This review summarizes recent findings and highlights the involvement of HCN4 in normal and pathological AVN function.
Collapse
Affiliation(s)
- Jaël S Copier
- Experimental Cardiology, Amsterdam UMC, Amsterdam, The Netherlands; Heart Failure & Arrhythmias, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Arie O Verkerk
- Experimental Cardiology, Amsterdam UMC, Amsterdam, The Netherlands; Heart Failure & Arrhythmias, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands; Medical Biology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Elisabeth M Lodder
- Experimental Cardiology, Amsterdam UMC, Amsterdam, The Netherlands; Heart Failure & Arrhythmias, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
| |
Collapse
|
5
|
Long V, El Gebeily G, Leblanc É, Senhadji M, Fiset C. Cardiac automaticity is modulated by IKACh in sinoatrial node during pregnancy. Cardiovasc Res 2024; 120:2208-2219. [PMID: 39259837 PMCID: PMC11687396 DOI: 10.1093/cvr/cvae200] [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: 10/13/2023] [Revised: 02/29/2024] [Accepted: 07/14/2024] [Indexed: 09/13/2024] Open
Abstract
AIMS Pregnant (P) women have a significantly elevated resting heart rate (HR), which makes cardiac arrhythmias more likely to occur. Although electrical remodelling of the sinoatrial node (SAN) has been documented, the underlying mechanism is not fully understood. The acetylcholine-activated potassium current (IKACh), one of the major repolarizing currents in the SAN, plays a critical role in HR control by hyperpolarizing the maximal diastolic potential (MDP) of the SAN action potential (AP), thereby reducing SAN automaticity and HR. Thus, considering its essential role in cardiac automaticity, this study aims to determine whether changes in IKACh are potentially involved in the increased HR associated with pregnancy. METHODS AND RESULTS Experiments were conducted on non-pregnant (NP) and pregnant (P; 17-18 days gestation) female CD-1 mice aged 2 to 4 months. IKACh was recorded on spontaneously beating SAN cells using the muscarinic agonist carbachol (CCh). Voltage-clamp data showed a reduction in IKACh density during pregnancy, which returned to control values shortly after delivery. The reduction in IKACh was explained by a decrease in protein expression of Kir3.1 channel subunit and the muscarinic type 2 receptor. In agreement with these findings, current-clamp data showed that the MDP of SAN cells from P mice were less hyperpolarized following CCh administration. Surface electrocardiograms (ECGs) recorded on anaesthetized mice revealed that the cholinergic antagonist atropine and the selective KACh channel blocker tertiapin-Q increased HR in NP mice and had only a minimal effect on P mice. AP and ECG data also showed that pregnancy is associated with a decrease in beating and HR variability, respectively. CONCLUSION IKACh function and expression are decreased in the mouse SAN during pregnancy, strongly suggesting that, in addition to other electrical remodelling of the SAN, reduced IKACh also plays an important role in the pregnancy-induced increased HR.
Collapse
Affiliation(s)
- Valérie Long
- Research Center, Montreal Heart Institute, 5000 Bélanger, Montréal, Québec, Canada H1T 1C8
- Faculty of Pharmacy, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada H3T 1J4
| | - Gracia El Gebeily
- Research Center, Montreal Heart Institute, 5000 Bélanger, Montréal, Québec, Canada H1T 1C8
- Faculty of Pharmacy, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada H3T 1J4
| | - Élisabeth Leblanc
- Research Center, Montreal Heart Institute, 5000 Bélanger, Montréal, Québec, Canada H1T 1C8
- Faculty of Pharmacy, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada H3T 1J4
| | - Marwa Senhadji
- Research Center, Montreal Heart Institute, 5000 Bélanger, Montréal, Québec, Canada H1T 1C8
- Faculty of Pharmacy, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada H3T 1J4
| | - Céline Fiset
- Research Center, Montreal Heart Institute, 5000 Bélanger, Montréal, Québec, Canada H1T 1C8
- Faculty of Pharmacy, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada H3T 1J4
| |
Collapse
|
6
|
Shi X, He L, Wang Y, Wu Y, Lin D, Chen C, Yang M, Huang S. Mitochondrial dysfunction is a key link involved in the pathogenesis of sick sinus syndrome: a review. Front Cardiovasc Med 2024; 11:1488207. [PMID: 39534498 PMCID: PMC11554481 DOI: 10.3389/fcvm.2024.1488207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
Sick sinus syndrome (SSS) is a grave medical condition that can precipitate sudden death. The pathogenesis of SSS remains incompletely understood. Existing research postulates that the fundamental mechanism involves increased fibrosis of the sinoatrial node and its surrounding tissues, as well as disturbances in the coupled-clock system, comprising the membrane clock and the Ca2+ clock. Mitochondrial dysfunction exacerbates regional tissue fibrosis and disrupts the functioning of both the membrane and calcium clocks. This plays a crucial role in the underlying pathophysiology of SSS, including mitochondrial energy metabolism disorders, mitochondrial oxidative stress damage, calcium overload, and mitochondrial quality control disorders. Elucidating the mitochondrial mechanisms involved in the pathophysiology of SSS and further investigating the disease's mechanisms is of great significance.
Collapse
Affiliation(s)
- Xinxin Shi
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Liming He
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yucheng Wang
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yue Wu
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Dongming Lin
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Chao Chen
- Department of Cardiology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Ming Yang
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuwei Huang
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
7
|
Akerman EC, Read MJ, Bose SJ, Koschinski A, Capel RA, Chao YC, Folkmanaite M, Ayagama T, Broadbent SD, Ahamed R, Simon JN, Terrar DA, Zaccolo M, Burton RAB. Activation of IP 3R in atrial cardiomyocytes leads to generation of cytosolic cAMP. Am J Physiol Heart Circ Physiol 2024; 327:H830-H846. [PMID: 39093001 PMCID: PMC11482242 DOI: 10.1152/ajpheart.00152.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia. Excessive stimulation of the inositol (1,4,5)-trisphosphate (IP3) signaling pathway has been linked to AF through abnormal calcium handling. However, little is known about the mechanisms involved in this process. We expressed the fluorescence resonance energy transfer (FRET)-based cytosolic cyclic adenosine monophosphate (cAMP) sensor EPAC-SH187 in neonatal rat atrial myocytes (NRAMs) and neonatal rat ventricular myocytes (NRVMs). In NRAMs, the addition of the α1-agonist, phenylephrine (PE, 3 µM), resulted in a FRET change of 21.20 ± 7.43%, and the addition of membrane-permeant IP3 derivative 2,3,6-tri-O-butyryl-myo-IP3(1,4,5)-hexakis(acetoxymethyl)ester (IP3-AM, 20 μM) resulted in a peak of 20.31 ± 6.74%. These FRET changes imply an increase in cAMP. Prior application of IP3 receptor (IP3R) inhibitors 2-aminoethyl diphenylborinate (2-APB, 2.5 μM) or Xestospongin-C (0.3 μM) significantly inhibited the change in FRET in NRAMs in response to PE. Xestospongin-C (0.3 μM) significantly inhibited the change in FRET in NRAMs in response to IP3-AM. The FRET change in response to PE in NRVMs was not inhibited by 2-APB or Xestospongin-C. Finally, the localization of cAMP signals was tested by expressing the FRET-based cAMP sensor, AKAP79-CUTie, which targets the intracellular surface of the plasmalemma. We found in NRAMs that PE led to FRET change corresponding to an increase in cAMP that was inhibited by 2-APB and Xestospongin-C. These data support further investigation of the proarrhythmic nature and components of IP3-induced cAMP signaling to identify potential pharmacological targets.NEW & NOTEWORTHY This study shows that indirect activation of the IP3 pathway in atrial myocytes using phenylephrine and direct activation using IP3-AM leads to an increase in cAMP and is in part localized to the cell membrane. These changes can be pharmacologically inhibited using IP3R inhibitors. However, the cAMP rise in ventricular myocytes is independent of IP3R calcium release. Our data support further investigation into the proarrhythmic nature of IP3-induced cAMP signaling.
Collapse
Affiliation(s)
- Emily C Akerman
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Matthew J Read
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Samuel J Bose
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Andreas Koschinski
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Rebecca A Capel
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Ying-Chi Chao
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Milda Folkmanaite
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Thamali Ayagama
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | | | | | - Jillian N Simon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Derek A Terrar
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Rebecca A B Burton
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
8
|
Page DA, Ruben PC. Cannabidiol potentiates hyperpolarization-activated cyclic nucleotide-gated (HCN4) channels. J Gen Physiol 2024; 156:e202313505. [PMID: 38652080 PMCID: PMC11040500 DOI: 10.1085/jgp.202313505] [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: 11/10/2023] [Revised: 03/15/2024] [Accepted: 04/09/2024] [Indexed: 04/25/2024] Open
Abstract
Cannabidiol (CBD), the main non-psychotropic phytocannabinoid produced by the Cannabis sativa plant, blocks a variety of cardiac ion channels. We aimed to identify whether CBD regulated the cardiac pacemaker channel or the hyperpolarization-activated cyclic nucleotide-gated channel (HCN4). HCN4 channels are important for the generation of the action potential in the sinoatrial node of the heart and increased heart rate in response to β-adrenergic stimulation. HCN4 channels were expressed in HEK 293T cells, and the effect of CBD application was examined using a whole-cell patch clamp. We found that CBD depolarized the V1/2 of activation in holo-HCN4 channels, with an EC50 of 1.6 µM, without changing the current density. CBD also sped activation kinetics by approximately threefold. CBD potentiation of HCN4 channels occurred via binding to the closed state of the channel. We found that CBD's mechanism of action was distinct from cAMP, as CBD also potentiated apo-HCN4 channels. The addition of an exogenous PIP2 analog did not alter the ability of CBD to potentiate HCN4 channels, suggesting that CBD also acts using a unique mechanism from the known HCN4 potentiator PIP2. Lastly, to gain insight into CBD's mechanism of action, computational modeling and targeted mutagenesis were used to predict that CBD binds to a lipid-binding pocket at the C-terminus of the voltage sensor. CBD represents the first FDA-approved drug to potentiate HCN4 channels, and our findings suggest a novel starting point for drug development targeting HCN4 channels.
Collapse
Affiliation(s)
- Dana A. Page
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
| | - Peter C. Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
| |
Collapse
|
9
|
Yao ZJ, Jiang YP, Yuan D, Hong P, He MJ, Li FX, Xu SY, Lin HB, Zhang HF. Decreased connexin 40 expression of the sinoatrial node mediates ischemic stroke-induced arrhythmia in mice. Exp Neurol 2024; 376:114773. [PMID: 38599368 DOI: 10.1016/j.expneurol.2024.114773] [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: 12/19/2023] [Revised: 03/23/2024] [Accepted: 04/06/2024] [Indexed: 04/12/2024]
Abstract
BACKGROUND Arrhythmia is the most common cardiac complication after ischemic stroke. Connexin 40 is the staple component of gap junctions, which influences the propagation of cardiac electrical signals in the sinoatrial node. However, the role of connexin 40 in post-stroke arrhythmia remains unclear. METHODS In this study, a permanent middle cerebral artery occlusion model was used to simulate the occurrence of an ischemic stroke. Subsequently, an electrocardiogram was utilized to record and assess variations in electrocardiogram measures. In addition, optical tissue clearing and whole-mount immunofluorescence staining were used to confirm the anatomical localization of the sinoatrial node, and the sinoatrial node tissue was collected for RNA sequencing to screen for potential pathological mechanisms. Lastly, the rAAV9-Gja5 virus was injected with ultrasound guidance into the heart to increase Cx40 expression in the sinoatrial node. RESULTS We demonstrated that the mice suffering from a permanent middle cerebral artery occlusion displayed significant arrhythmia, including atrial fibrillation, premature ventricular contractions, atrioventricular block, and abnormal electrocardiogram parameters. Of note, we observed a decrease in connexin 40 expression within the sinoatrial node after the ischemic stroke via RNA sequencing and western blot. Furthermore, rAAV9-Gja5 treatment ameliorated the occurrence of arrhythmia following stroke. CONCLUSIONS In conclusion, decreased connexin 40 expression in the sinoatrial node contributed to the ischemic stroke-induced cardiac arrhythmia. Therefore, enhancing connexin 40 expression holds promise as a potential therapeutic approach for ischemic stroke-induced arrhythmia.
Collapse
Affiliation(s)
- Zhi-Jun Yao
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Yan-Pin Jiang
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China; Department of Anesthesiology, The First Hospital Affiliated to the Army Medical University, Chongqing 400038, China
| | - Dan Yuan
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Pu Hong
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Meng-Jiao He
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Feng-Xian Li
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Shi-Yuan Xu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Hong-Bin Lin
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| | - Hong-Fei Zhang
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| |
Collapse
|
10
|
Hennis K, Piantoni C, Biel M, Fenske S, Wahl-Schott C. Pacemaker Channels and the Chronotropic Response in Health and Disease. Circ Res 2024; 134:1348-1378. [PMID: 38723033 PMCID: PMC11081487 DOI: 10.1161/circresaha.123.323250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/13/2024]
Abstract
Loss or dysregulation of the normally precise control of heart rate via the autonomic nervous system plays a critical role during the development and progression of cardiovascular disease-including ischemic heart disease, heart failure, and arrhythmias. While the clinical significance of regulating changes in heart rate, known as the chronotropic effect, is undeniable, the mechanisms controlling these changes remain not fully understood. Heart rate acceleration and deceleration are mediated by increasing or decreasing the spontaneous firing rate of pacemaker cells in the sinoatrial node. During the transition from rest to activity, sympathetic neurons stimulate these cells by activating β-adrenergic receptors and increasing intracellular cyclic adenosine monophosphate. The same signal transduction pathway is targeted by positive chronotropic drugs such as norepinephrine and dobutamine, which are used in the treatment of cardiogenic shock and severe heart failure. The cyclic adenosine monophosphate-sensitive hyperpolarization-activated current (If) in pacemaker cells is passed by hyperpolarization-activated cyclic nucleotide-gated cation channels and is critical for generating the autonomous heartbeat. In addition, this current has been suggested to play a central role in the chronotropic effect. Recent studies demonstrate that cyclic adenosine monophosphate-dependent regulation of HCN4 (hyperpolarization-activated cyclic nucleotide-gated cation channel isoform 4) acts to stabilize the heart rate, particularly during rapid rate transitions induced by the autonomic nervous system. The mechanism is based on creating a balance between firing and recently discovered nonfiring pacemaker cells in the sinoatrial node. In this way, hyperpolarization-activated cyclic nucleotide-gated cation channels may protect the heart from sinoatrial node dysfunction, secondary arrhythmia of the atria, and potentially fatal tachyarrhythmia of the ventricles. Here, we review the latest findings on sinoatrial node automaticity and discuss the physiological and pathophysiological role of HCN pacemaker channels in the chronotropic response and beyond.
Collapse
Affiliation(s)
- Konstantin Hennis
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center Munich, Walter Brendel Centre of Experimental Medicine, Faculty of Medicine (K.H., C.P., C.W.-S.), Ludwig-Maximilians-Universität München, Germany
| | - Chiara Piantoni
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center Munich, Walter Brendel Centre of Experimental Medicine, Faculty of Medicine (K.H., C.P., C.W.-S.), Ludwig-Maximilians-Universität München, Germany
| | - Martin Biel
- Department of Pharmacy, Center for Drug Research (M.B., S.F.), Ludwig-Maximilians-Universität München, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Germany (M.B., S.F.)
| | - Stefanie Fenske
- Department of Pharmacy, Center for Drug Research (M.B., S.F.), Ludwig-Maximilians-Universität München, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Germany (M.B., S.F.)
| | - Christian Wahl-Schott
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center Munich, Walter Brendel Centre of Experimental Medicine, Faculty of Medicine (K.H., C.P., C.W.-S.), Ludwig-Maximilians-Universität München, Germany
| |
Collapse
|
11
|
Li T, Marashly Q, Kim JA, Li N, Chelu MG. Cardiac conduction diseases: understanding the molecular mechanisms to uncover targets for future treatments. Expert Opin Ther Targets 2024; 28:385-400. [PMID: 38700451 PMCID: PMC11395937 DOI: 10.1080/14728222.2024.2351501] [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: 11/18/2023] [Accepted: 05/01/2024] [Indexed: 05/05/2024]
Abstract
INTRODUCTION The cardiac conduction system (CCS) is crucial for maintaining adequate cardiac frequency at rest and modulation during exercise. Furthermore, the atrioventricular node and His-Purkinje system are essential for maintaining atrioventricular and interventricular synchrony and consequently maintaining an adequate cardiac output. AREAS COVERED In this review article, we examine the anatomy, physiology, and pathophysiology of the CCS. We then discuss in detail the most common genetic mutations and the molecular mechanisms of cardiac conduction disease (CCD) and provide our perspectives on future research and therapeutic opportunities in this field. EXPERT OPINION Significant advancement has been made in understanding the molecular mechanisms of CCD, including the recognition of the heterogeneous signaling at the subcellular levels of sinoatrial node, the involvement of inflammatory and autoimmune mechanisms, and the potential impact of epigenetic regulations on CCD. However, the current treatment of CCD manifested as bradycardia still relies primarily on cardiovascular implantable electronic devices (CIEDs). On the other hand, an If specific inhibitor was developed to treat inappropriate sinus tachycardia and sinus tachycardia in heart failure patients with reduced ejection fraction. More work is needed to translate current knowledge into pharmacologic or genetic interventions for the management of CCDs.
Collapse
Affiliation(s)
- Tingting Li
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Qussay Marashly
- Department of Cardiology, Montefiore Medical Center, New York, NY, USA
| | - Jitae A. Kim
- Division of CardiovasculMedicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Na Li
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Mihail G. Chelu
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine (Division of Cardiology), Baylor College of Medicine, Houston, TX, USA
- Baylor St. Luke’s Medical Center, Houston, Texas, USA
- Texas Heart Institute, Houston, Texas, USA
| |
Collapse
|
12
|
Maltsev AV, Stern MD, Lakatta EG, Maltsev VA. A novel conceptual model of heart rate autonomic modulation based on a small-world modular structure of the sinoatrial node. Front Physiol 2023; 14:1276023. [PMID: 38148905 PMCID: PMC10750401 DOI: 10.3389/fphys.2023.1276023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/27/2023] [Indexed: 12/28/2023] Open
Abstract
The present view on heartbeat initiation is that a primary pacemaker cell or a group of cells in the sinoatrial node (SAN) center paces the rest of the SAN and the atria. However, recent high-resolution imaging studies show a more complex paradigm of SAN function that emerges from heterogeneous signaling, mimicking brain cytoarchitecture and function. Here, we developed and tested a new conceptual numerical model of SAN organized similarly to brain networks featuring a modular structure with small-world topology. In our model, a lower rate module leads action potential (AP) firing in the basal state and during parasympathetic stimulation, whereas a higher rate module leads during β-adrenergic stimulation. Such a system reproduces the respective shift of the leading pacemaker site observed experimentally and a wide range of rate modulation and robust function while conserving energy. Since experimental studies found functional modules at different scales, from a few cells up to the highest scale of the superior and inferior SAN, the SAN appears to feature hierarchical modularity, i.e., within each module, there is a set of sub-modules, like in the brain, exhibiting greater robustness, adaptivity, and evolvability of network function. In this perspective, our model offers a new mainframe for interpreting new data on heterogeneous signaling in the SAN at different scales, providing new insights into cardiac pacemaker function and SAN-related cardiac arrhythmias in aging and disease.
Collapse
Affiliation(s)
| | | | | | - Victor A. Maltsev
- Intramural Research Program, National Institute on Aging, Baltimore, MD, United States
| |
Collapse
|
13
|
Wu Y, Wang Q, Granger J, Gaido OR, Aguilar EN, Ludwig A, Moroni A, Bianchet MA, Anderson ME. HCN channels sense temperature and determine heart rate responses to heat. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.02.556046. [PMID: 37693513 PMCID: PMC10491304 DOI: 10.1101/2023.09.02.556046] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Heart rate increases with heat, [1-3] constituting a fundamental physiological relationship in vertebrates. Each normal heartbeat is initiated by an action potential generated in a sinoatrial nodal pacemaker cell. Pacemaker cells are enriched with hyperpolarization activated cyclic nucleotide-gated ion channels (HCN) that deliver cell membrane depolarizing inward current that triggers action potentials. HCN channel current increases due to cAMP binding, a mechanism coupling adrenergic tone to physiological 'fight or flight' heart rate acceleration. However, the mechanism(s) for heart rate response to thermal energy is unknown. We used thermodynamical and homology computational modeling, site-directed mutagenesis and mouse models to identify a concise motif on the S4-S5 linker of the cardiac pacemaker HCN4 channels (M407/Y409) that determines HCN4 current (If) and cardiac pacemaker cell responses to heat. This motif is required for heat sensing in cardiac pacemaker cells and in isolated hearts. In contrast, the cyclic nucleotide binding domain is not required for heat induced HCN4 current increases. However, a loss of function M407/Y409 motif mutation prevented normal heat and cAMP responses, suggesting that heat sensing machinery is essential for operating the cAMP allosteric pathway and is central to HCN4 modulation. The M407/Y409 motif is conserved across all HCN family members suggesting that HCN channels participate broadly in coupling heat to changes in cell membrane excitability.
Collapse
Affiliation(s)
- Yuejin Wu
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Qinchuan Wang
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jonathan Granger
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Oscar Reyes Gaido
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Eric Nunez Aguilar
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andreas Ludwig
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Anna Moroni
- Department of Biosciences, University of Milan, via Celoria 26, 20133 Milan, Italy
| | - Mario A Bianchet
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mark E Anderson
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Medicine and Division of Biological Sciences, University of Chicago, Chicago, IL, USA
| |
Collapse
|
14
|
Ruan H, Mandla R, Ravi N, Galang G, Soe AW, Olgin JE, Lang D, Vedantham V. Cholecystokinin-A Signaling Regulates Automaticity of Pacemaker Cardiomyocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.24.525392. [PMID: 36747643 PMCID: PMC9900793 DOI: 10.1101/2023.01.24.525392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Aims The behavior of pacemaker cardiomyocytes (PCs) in the sinoatrial node (SAN) is modulated by neurohormonal and paracrine factors, many of which signal through G-protein coupled receptors (GPCRs). The aims of the present study are to catalog GPCRs that are differentially expressed in the mammalian SAN and to define the acute physiological consequences of activating the cholecystokinin-A signaling system in isolated PCs. Methods and Results Using bulk and single cell RNA sequencing datasets, we identify a set of GPCRs that are differentially expressed between SAN and right atrial tissue, including several whose roles in PCs and in the SAN have not been thoroughly characterized. Focusing on one such GPCR, Cholecystokinin-A receptor (CCK A R), we demonstrate expression of Cckar mRNA specifically in mouse PCs, and further demonstrate that subsets of SAN fibroblasts and neurons within the cardiac intrinsic nervous system express cholecystokinin, the ligand for CCK A R. Using mouse models, we find that while baseline SAN function is not dramatically affected by loss of CCK A R, the firing rate of individual PCs is slowed by exposure to sulfated cholecystokinin-8 (sCCK-8), the high affinity ligand for CCK A R. The effect of sCCK-8 on firing rate is mediated by reduction in the rate of spontaneous phase 4 depolarization of PCs and is mitigated by activation of beta-adrenergic signaling. Conclusions (1) PCs express many GPCRs whose specific roles in SAN function have not been characterized, (2) Activation of the the cholecystokinin-A signaling pathway regulates PC automaticity.
Collapse
|
15
|
Borges JI, Suster MS, Lymperopoulos A. Cardiac RGS Proteins in Human Heart Failure and Atrial Fibrillation: Focus on RGS4. Int J Mol Sci 2023; 24:6136. [PMID: 37047106 PMCID: PMC10147095 DOI: 10.3390/ijms24076136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 04/14/2023] Open
Abstract
The regulator of G protein signaling (RGS) proteins are crucial for the termination of G protein signals elicited by G protein-coupled receptors (GPCRs). This superfamily of cell membrane receptors, by far the largest and most versatile in mammals, including humans, play pivotal roles in the regulation of cardiac function and homeostasis. Perturbations in both the activation and termination of their G protein-mediated signaling underlie numerous heart pathologies, including heart failure (HF) and atrial fibrillation (AFib). Therefore, RGS proteins play important roles in the pathophysiology of these two devasting cardiac diseases, and several of them could be targeted therapeutically. Although close to 40 human RGS proteins have been identified, each RGS protein seems to interact only with a specific set of G protein subunits and GPCR types/subtypes in any given tissue or cell type. Numerous in vitro and in vivo studies in animal models, and also in diseased human heart tissue obtained from transplantations or tissue banks, have provided substantial evidence of the roles various cardiomyocyte RGS proteins play in cardiac normal homeostasis as well as pathophysiology. One RGS protein in particular, RGS4, has been reported in what are now decades-old studies to be selectively upregulated in human HF. It has also been implicated in protection against AFib via knockout mice studies. This review summarizes the current understanding of the functional roles of cardiac RGS proteins and their implications for the treatment of HF and AFib, with a specific focus on RGS4 for the aforementioned reasons but also because it can be targeted successfully with small organic molecule inhibitors.
Collapse
Affiliation(s)
| | | | - Anastasios Lymperopoulos
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Barry and Judy Silverrman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328-2018, USA
| |
Collapse
|
16
|
Del Calvo G, Baggio Lopez T, Lymperopoulos A. The therapeutic potential of targeting cardiac RGS4. Ther Adv Cardiovasc Dis 2023; 17:17539447231199350. [PMID: 37724539 PMCID: PMC10510358 DOI: 10.1177/17539447231199350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 08/16/2023] [Indexed: 09/21/2023] Open
Abstract
G protein-coupled receptors (GPCRs) play pivotal roles in regulation of cardiac function and homeostasis. To function properly, every cell needs these receptors to be stimulated only when a specific extracellular stimulus is present, and to be silenced the moment that stimulus is removed. The regulator of G protein signaling (RGS) proteins are crucial for the latter to occur at the cell membrane, where the GPCR normally resides. Perturbations in both activation and termination of G protein signaling underlie numerous heart pathologies. Although more than 30 mammalian RGS proteins have been identified, each RGS protein seems to interact only with a specific set of G protein subunits and GPCR types/subtypes in any given tissue or cell type, and this applies to the myocardium as well. A large number of studies have provided substantial evidence for the roles various RGS proteins expressed in cardiomyocytes play in cardiac physiology and heart disease pathophysiology. This review summarizes the current understanding of the functional roles of cardiac RGS proteins and their implications for the treatment of specific heart diseases, such as heart failure and atrial fibrillation. We focus on cardiac RGS4 in particular, since this isoform appears to be selectively (among the RGS protein family) upregulated in human heart failure and is also the target of ongoing drug discovery efforts for the treatment of a variety of diseases.
Collapse
Affiliation(s)
- Giselle Del Calvo
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Teresa Baggio Lopez
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Anastasios Lymperopoulos
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, 3200 South University Drive, HPD (Terry) Building/Room 1350, Fort Lauderdale, FL 33328-2018, USA
| |
Collapse
|
17
|
Ricci E, Bartolucci C, Severi S. The virtual sinoatrial node: What did computational models tell us about cardiac pacemaking? PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 177:55-79. [PMID: 36374743 DOI: 10.1016/j.pbiomolbio.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 10/17/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022]
Abstract
Since its discovery, the sinoatrial node (SAN) has represented a fascinating and complex matter of research. Despite over a century of discoveries, a full comprehension of pacemaking has still to be achieved. Experiments often produced conflicting evidence that was used either in support or against alternative theories, originating intense debates. In this context, mathematical descriptions of the phenomena underlying the heartbeat have grown in importance in the last decades since they helped in gaining insights where experimental evaluation could not reach. This review presents the most updated SAN computational models and discusses their contribution to our understanding of cardiac pacemaking. Electrophysiological, structural and pathological aspects - as well as the autonomic control over the SAN - are taken into consideration to reach a holistic view of SAN activity.
Collapse
Affiliation(s)
- Eugenio Ricci
- Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, Cesena (FC), Italy
| | - Chiara Bartolucci
- Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, Cesena (FC), Italy
| | - Stefano Severi
- Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, Cesena (FC), Italy.
| |
Collapse
|
18
|
Fan W, Sun X, Yang C, Wan J, Luo H, Liao B. Pacemaker activity and ion channels in the sinoatrial node cells: MicroRNAs and arrhythmia. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 177:151-167. [PMID: 36450332 DOI: 10.1016/j.pbiomolbio.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/13/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022]
Abstract
The primary pacemaking activity of the heart is determined by a spontaneous action potential (AP) within sinoatrial node (SAN) cells. This unique AP generation relies on two mechanisms: membrane clocks and calcium clocks. Nonhomologous arrhythmias are caused by several functional and structural changes in the myocardium. MicroRNAs (miRNAs) are essential regulators of gene expression in cardiomyocytes. These miRNAs play a vital role in regulating the stability of cardiac conduction and in the remodeling process that leads to arrhythmias. Although it remains unclear how miRNAs regulate the expression and function of ion channels in the heart, these regulatory mechanisms may support the development of emerging therapies. This study discusses the spread and generation of AP in the SAN as well as the regulation of miRNAs and individual ion channels. Arrhythmogenicity studies on ion channels will provide a research basis for miRNA modulation as a new therapeutic target.
Collapse
Affiliation(s)
- Wei Fan
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China
| | - Xuemei Sun
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China
| | - Chao Yang
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China
| | - Juyi Wan
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China.
| | - Hongli Luo
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China.
| | - Bin Liao
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China.
| |
Collapse
|
19
|
Qu JH, Telljohann R, Byshkov R, Lakatta EG. Characterization of diverse populations of sinoatrial node cells and their proliferation potential at single nucleus resolution. Heliyon 2022; 9:e12708. [PMID: 36632093 PMCID: PMC9826826 DOI: 10.1016/j.heliyon.2022.e12708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 11/10/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
Background Each heartbeat is initiated in the sinoatrial node (SAN), and although a recent study (GSE130710) using single nucleus RNA-seq had discovered different populations of cell types within SAN tissue, the distinct potential functions of these cell types have not been delineated. Methods To infer some special potential functions of different SAN cell clusters, we applied principal component analysis (PCA), t-distributed stochastic neighbor embedding (t-SNE) and uniform manifold approximation and projection (UMAP) to the GSE130710 dataset to reduce dimensions, followed by Pseudotime trajectory and AUCell analyses, ANOVA and Hurdle statistical models, and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichments to determine functional potential of cell types. Nuclear EdU immuno-labeling of SAN tissue confirmed cell type proliferation. Findings We identified elements of a coupled clock system known to drive SAN cell pacemaking within the GSE130710 sinus node myocyte cluster, which, surprisingly, manifested signals of suppressed fatty acid and nitrogen metabolism and reduced immune gene expression. Proliferation signaling was enriched in endocardial, epicardial, epithelial cells, and macrophages, in which, fatty acid and nitrogen metabolic signals were also suppressed, but immune signaling was enhanced. EdU labeling was rare in pacemaker cells but was robust in interstitial cells. Interpretation Pacemaker cells that initiate each heartbeat manifest suppressed fatty acid and nitrogen metabolism and limited immune signaling and proliferation potential. In contrast, other populations of SAN cells not directly involved in the initiation of heartbeats, manifest robust proliferation and immune potential, likely to ensure an environment required to sustain healthy SAN tissue pacemaker function.
Collapse
|
20
|
Maltsev VA, Stern MD. The paradigm shift: Heartbeat initiation without "the pacemaker cell". Front Physiol 2022; 13:1090162. [PMID: 36569749 PMCID: PMC9780451 DOI: 10.3389/fphys.2022.1090162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022] Open
Abstract
The current dogma about the heartbeat origin is based on "the pacemaker cell," a specialized cell residing in the sinoatrial node (SAN) that exhibits spontaneous diastolic depolarization triggering rhythmic action potentials (APs). Recent high-resolution imaging, however, demonstrated that Ca signals and APs in the SAN are heterogeneous, with many cells generating APs of different rates and rhythms or even remaining non-firing (dormant cells), i.e., generating only subthreshold signals. Here we numerically tested a hypothesis that a community of dormant cells can generate normal automaticity, i.e., "the pacemaker cell" is not required to initiate rhythmic cardiac impulses. Our model includes 1) non-excitable cells generating oscillatory local Ca releases and 2) an excitable cell lacking automaticity. While each cell in isolation was not "the pacemaker cell", the cell system generated rhythmic APs: The subthreshold signals of non-excitable cells were transformed into respective membrane potential oscillations via electrogenic Na/Ca exchange and further transferred and integrated (computed) by the excitable cells to reach its AP threshold, generating rhythmic pacemaking. Cardiac impulse is an emergent property of the SAN cellular network and can be initiated by cells lacking intrinsic automaticity. Cell heterogeneity, weak coupling, subthreshold signals, and their summation are critical properties of the new pacemaker mechanism, i.e., cardiac pacemaker can operate via a signaling process basically similar to that of "temporal summation" happening in a neuron with input from multiple presynaptic cells. The new mechanism, however, does not refute the classical pacemaker cell-based mechanism: both mechanisms can co-exist and interact within SAN tissue.
Collapse
|
21
|
Peters CH, Rickert C, Morotti S, Grandi E, Aronow KA, Beam KG, Proenza C. The funny current If is essential for the fight-or-flight response in cardiac pacemaker cells. J Gen Physiol 2022; 154:e202213193. [PMID: 36305844 PMCID: PMC9812006 DOI: 10.1085/jgp.202213193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/22/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
The sympathetic nervous system fight-or-flight response is characterized by a rapid increase in heart rate, which is mediated by an increase in the spontaneous action potential (AP) firing rate of pacemaker cells in the sinoatrial node. Sympathetic neurons stimulate sinoatrial myocytes (SAMs) by activating β adrenergic receptors (βARs) and increasing cAMP. The funny current (If) is among the cAMP-sensitive currents in SAMs. If is critical for pacemaker activity, however, its role in the fight-or-flight response remains controversial. In this study, we used AP waveform analysis, machine learning, and dynamic clamp experiments in acutely isolated SAMs from mice to quantitatively define the AP waveform changes and role of If in the fight-or-flight increase in AP firing rate. We found that while βAR stimulation significantly altered nearly all AP waveform parameters, the increase in firing rate was only correlated with changes in a subset of parameters (diastolic duration, late AP duration, and diastolic depolarization rate). Dynamic clamp injection of the βAR-sensitive component of If showed that it accounts for ∼41% of the fight-or-flight increase in AP firing rate and 60% of the decrease in the interval between APs. Thus, If is an essential contributor to the fight-or-flight increase in heart rate.
Collapse
Affiliation(s)
- Colin H. Peters
- Department of Physiology & Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Christian Rickert
- Department of Physiology & Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Stefano Morotti
- Department of Pharmacology, University of California, Davis, School of Medicine, Davis, CA
| | - Eleonora Grandi
- Department of Pharmacology, University of California, Davis, School of Medicine, Davis, CA
| | | | - Kurt G. Beam
- Department of Physiology & Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Catherine Proenza
- Department of Physiology & Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
22
|
Wirth AN, Tsutsui K, Maltsev VA, Lakatta EG. Adenosine reduces sinoatrial node cell action potential firing rate by uncoupling its membrane and calcium clocks. Front Physiol 2022; 13:977807. [PMID: 36505046 PMCID: PMC9730041 DOI: 10.3389/fphys.2022.977807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 11/01/2022] [Indexed: 11/25/2022] Open
Abstract
The spontaneous action potential (AP) firing rate of sinoatrial nodal cells (SANC) is regulated by a system of intracellular Ca2+ and membrane ion current clocks driven by Ca2+-calmodulin-activated adenylyl cyclase-protein kinase-A signaling. The mean AP-cycle length (APCL) and APCL variability inform on the effectiveness of clock coupling. Endogenous ATP metabolite adenosine binds to adenosine receptors (A1, A3) that couple to Gi protein-coupled receptors, reducing spontaneous AP firing rate via Gβγ signaling that activates IKAch,Ado. Adenosine also inhibits adenylyl cyclase activity via Gαi signaling, impacting cAMP-mediated protein kinase-A-dependent protein phosphorylation. We hypothesize that in addition to IKAch,Ado activation, adenosine impacts also Ca2+ via Gαi signaling and that both effects reduce AP firing rate by reducing the effectiveness of the Ca2+ and membrane clock coupling. To this end, we measured Ca2+ and membrane potential characteristics in enzymatically isolated single rabbit SANC. 10 µM adenosine substantially increased both the mean APCL (on average by 43%, n = 10) and AP beat-to-beat variability from 5.1 ± 1.7% to 7.2 ± 2.0% (n = 10) measured via membrane potential and 5.0 ± 2.2% to 10.6 ± 5.9% (n = 40) measured via Ca2+ (assessed as the coefficient of variability = SD/mean). These effects were mediated by hyperpolarization of the maximum diastolic membrane potential (membrane clock effect) and suppression of diastolic local Ca2+releases (LCRs) (Ca2+-clock effect): as LCR size distributions shifted to smaller values, the time of LCR occurrence during diastolic depolarization (LCR period) became prolonged, and the ensemble LCR signal became reduced. The tight linear relationship of coupling between LCR period to the APCL in the presence of adenosine "drifted" upward and leftward, i.e. for a given LCR period, APCL was prolonged, becoming non-linear indicating clock uncoupling. An extreme case of uncoupling occurred at higher adenosine concentrations (>100 µM): small stochastic LCRs failed to self-organize and synchronize to the membrane clock, thus creating a failed attempt to generate an AP resulting in arrhythmia and cessation of AP firing. Thus, the effects of adenosine to activate Gβγ and IKACh,Ado and to activate Gαi, suppressing adenylyl cyclase activity, both contribute to the adenosine-induced increase in the mean APCL and APCL variability by reducing the fidelity of clock coupling and AP firing rate.
Collapse
|
23
|
Ren L, Thai PN, Gopireddy RR, Timofeyev V, Ledford HA, Woltz RL, Park S, Puglisi JL, Moreno CM, Santana LF, Conti AC, Kotlikoff MI, Xiang YK, Yarov-Yarovoy V, Zaccolo M, Zhang XD, Yamoah EN, Navedo MF, Chiamvimonvat N. Adenylyl cyclase isoform 1 contributes to sinoatrial node automaticity via functional microdomains. JCI Insight 2022; 7:e162602. [PMID: 36509290 PMCID: PMC9746826 DOI: 10.1172/jci.insight.162602] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 10/05/2022] [Indexed: 11/22/2022] Open
Abstract
Sinoatrial node (SAN) cells are the heart's primary pacemaker. Their activity is tightly regulated by β-adrenergic receptor (β-AR) signaling. Adenylyl cyclase (AC) is a key enzyme in the β-AR pathway that catalyzes the production of cAMP. There are current gaps in our knowledge regarding the dominant AC isoforms and the specific roles of Ca2+-activated ACs in the SAN. The current study tests the hypothesis that distinct AC isoforms are preferentially expressed in the SAN and compartmentalize within microdomains to orchestrate heart rate regulation during β-AR signaling. In contrast to atrial and ventricular myocytes, SAN cells express a diverse repertoire of ACs, with ACI as the predominant Ca2+-activated isoform. Although ACI-KO (ACI-/-) mice exhibit normal cardiac systolic or diastolic function, they experience SAN dysfunction. Similarly, SAN-specific CRISPR/Cas9-mediated gene silencing of ACI results in sinus node dysfunction. Mechanistically, hyperpolarization-activated cyclic nucleotide-gated 4 (HCN4) channels form functional microdomains almost exclusively with ACI, while ryanodine receptor and L-type Ca2+ channels likely compartmentalize with ACI and other AC isoforms. In contrast, there were no significant differences in T-type Ca2+ and Na+ currents at baseline or after β-AR stimulation between WT and ACI-/- SAN cells. Due to its central characteristic feature as a Ca2+-activated isoform, ACI plays a unique role in sustaining the rise of local cAMP and heart rates during β-AR stimulation. The findings provide insights into the critical roles of the Ca2+-activated isoform of AC in sustaining SAN automaticity that is distinct from contractile cardiomyocytes.
Collapse
Affiliation(s)
- Lu Ren
- Department of Internal Medicine, Division of Cardiovascular Medicine, UCD, Davis, California, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Phung N. Thai
- Department of Internal Medicine, Division of Cardiovascular Medicine, UCD, Davis, California, USA
- Department of Veteran Affairs, Northern California Health Care System, Sacramento, California, USA
| | | | - Valeriy Timofeyev
- Department of Internal Medicine, Division of Cardiovascular Medicine, UCD, Davis, California, USA
| | - Hannah A. Ledford
- Department of Internal Medicine, Division of Cardiovascular Medicine, UCD, Davis, California, USA
| | - Ryan L. Woltz
- Department of Internal Medicine, Division of Cardiovascular Medicine, UCD, Davis, California, USA
- Department of Veteran Affairs, Northern California Health Care System, Sacramento, California, USA
| | - Seojin Park
- Department of Physiology and Cell Biology, University of Nevada, Reno, Reno, Nevada, USA
- Prestige Biopharma Korea, Myongjigukje 7-ro, Gangseo-gu, Busan, South Korea
| | - Jose L. Puglisi
- College of Medicine. California North State University, Sacramento, California, USA
| | - Claudia M. Moreno
- Department of Physiology and Membrane Biology, UCD, Davis, California, USA
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, Washington, USA
| | | | - Alana C. Conti
- Research & Development Service, John D. Dingell VA Medical Center, and
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan, USA
| | | | - Yang Kevin Xiang
- Department of Veteran Affairs, Northern California Health Care System, Sacramento, California, USA
- Department of Pharmacology, UCD, Davis, California, USA
| | | | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom
| | - Xiao-Dong Zhang
- Department of Internal Medicine, Division of Cardiovascular Medicine, UCD, Davis, California, USA
- Department of Veteran Affairs, Northern California Health Care System, Sacramento, California, USA
| | - Ebenezer N. Yamoah
- Department of Physiology and Cell Biology, University of Nevada, Reno, Reno, Nevada, USA
| | | | - Nipavan Chiamvimonvat
- Department of Internal Medicine, Division of Cardiovascular Medicine, UCD, Davis, California, USA
- Department of Veteran Affairs, Northern California Health Care System, Sacramento, California, USA
- Department of Pharmacology, UCD, Davis, California, USA
| |
Collapse
|
24
|
Campana C, Ricci E, Bartolucci C, Severi S, Sobie EA. Coupling and heterogeneity modulate pacemaking capability in healthy and diseased two-dimensional sinoatrial node tissue models. PLoS Comput Biol 2022; 18:e1010098. [PMID: 36409762 DOI: 10.1371/journal.pcbi.1010098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 12/14/2022] [Accepted: 11/04/2022] [Indexed: 11/22/2022] Open
Abstract
Both experimental and modeling studies have attempted to determine mechanisms by which a small anatomical region, such as the sinoatrial node (SAN), can robustly drive electrical activity in the human heart. However, despite many advances from prior research, important questions remain unanswered. This study aimed to investigate, through mathematical modeling, the roles of intercellular coupling and cellular heterogeneity in synchronization and pacemaking within the healthy and diseased SAN. In a multicellular computational model of a monolayer of either human or rabbit SAN cells, simulations revealed that heterogenous cells synchronize their discharge frequency into a unique beating rhythm across a wide range of heterogeneity and intercellular coupling values. However, an unanticipated behavior appeared under pathological conditions where perturbation of ionic currents led to reduced excitability. Under these conditions, an intermediate range of intercellular coupling (900-4000 MΩ) was beneficial to SAN automaticity, enabling a very small portion of tissue (3.4%) to drive propagation, with propagation failure occurring at both lower and higher resistances. This protective effect of intercellular coupling and heterogeneity, seen in both human and rabbit tissues, highlights the remarkable resilience of the SAN. Overall, the model presented in this work allowed insight into how spontaneous beating of the SAN tissue may be preserved in the face of perturbations that can cause individual cells to lose automaticity. The simulations suggest that certain degrees of gap junctional coupling protect the SAN from ionic perturbations that can be caused by drugs or mutations.
Collapse
Affiliation(s)
- Chiara Campana
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Eugenio Ricci
- Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, Cesena, Italy
| | - Chiara Bartolucci
- Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, Cesena, Italy
| | - Stefano Severi
- Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, Cesena, Italy
| | - Eric A Sobie
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| |
Collapse
|
25
|
Guarina L, Moghbel AN, Pourhosseinzadeh MS, Cudmore RH, Sato D, Clancy CE, Santana LF. Biological noise is a key determinant of the reproducibility and adaptability of cardiac pacemaking and EC coupling. J Gen Physiol 2022; 154:e202012613. [PMID: 35482009 PMCID: PMC9059386 DOI: 10.1085/jgp.202012613] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/16/2022] [Accepted: 04/07/2022] [Indexed: 12/23/2022] Open
Abstract
Each heartbeat begins with the generation of an action potential in pacemaking cells in the sinoatrial node. This signal triggers contraction of cardiac muscle through a process termed excitation-contraction (EC) coupling. EC coupling is initiated in dyadic structures of cardiac myocytes, where ryanodine receptors in the junctional sarcoplasmic reticulum come into close apposition with clusters of CaV1.2 channels in invaginations of the sarcolemma. Cooperative activation of CaV1.2 channels within these clusters causes a local increase in intracellular Ca2+ that activates the juxtaposed ryanodine receptors. A salient feature of healthy cardiac function is the reliable and precise beat-to-beat pacemaking and amplitude of Ca2+ transients during EC coupling. In this review, we discuss recent discoveries suggesting that the exquisite reproducibility of this system emerges, paradoxically, from high variability at subcellular, cellular, and network levels. This variability is attributable to stochastic fluctuations in ion channel trafficking, clustering, and gating, as well as dyadic structure, which increase intracellular Ca2+ variance during EC coupling. Although the effects of these large, local fluctuations in function and organization are sometimes negligible at the macroscopic level owing to spatial-temporal summation within and across cells in the tissue, recent work suggests that the "noisiness" of these intracellular Ca2+ events may either enhance or counterintuitively reduce variability in a context-dependent manner. Indeed, these noisy events may represent distinct regulatory features in the tuning of cardiac contractility. Collectively, these observations support the importance of incorporating experimentally determined values of Ca2+ variance in all EC coupling models. The high reproducibility of cardiac contraction is a paradoxical outcome of high Ca2+ signaling variability at subcellular, cellular, and network levels caused by stochastic fluctuations in multiple processes in time and space. This underlying stochasticity, which counterintuitively manifests as reliable, consistent Ca2+ transients during EC coupling, also allows for rapid changes in cardiac rhythmicity and contractility in health and disease.
Collapse
Affiliation(s)
- Laura Guarina
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, CA
| | - Ariana Neelufar Moghbel
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, CA
| | | | - Robert H. Cudmore
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, CA
| | - Daisuke Sato
- Department of Pharmacology, University of California Davis School of Medicine, Davis, CA
| | - Colleen E. Clancy
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, CA
| | - Luis Fernando Santana
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, CA
| |
Collapse
|
26
|
Maltsev AV, Stern MD, Maltsev VA. Disorder in Ca2+ release unit locations confers robustness but cuts flexibility of heart pacemaking. J Gen Physiol 2022; 154:e202113061. [PMID: 35943725 PMCID: PMC9366202 DOI: 10.1085/jgp.202113061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 05/04/2022] [Accepted: 06/21/2022] [Indexed: 12/12/2022] Open
Abstract
Excitation-contraction coupling kinetics is dictated by the action potential rate of sinoatrial-nodal cells. These cells generate local Ca releases (LCRs) that activate Na/Ca exchanger current, which accelerates diastolic depolarization and determines the pace. LCRs are generated by clusters of ryanodine receptors, Ca release units (CRUs), residing in the sarcoplasmic reticulum. While CRU distribution exhibits substantial heterogeneity, its functional importance remains unknown. Using numerical modeling, here we show that with a square lattice distribution of CRUs, Ca-induced-Ca-release propagation during diastolic depolarization is insufficient for pacemaking within a broad range of realistic ICaL densities. Allowing each CRU to deviate randomly from its lattice position allows sparks to propagate, as observed experimentally. As disorder increases, the CRU distribution exhibits larger empty spaces and simultaneously CRU clusters, as in Poisson clumping. Propagating within the clusters, Ca release becomes synchronized, increasing action potential rate and reviving pacemaker function of dormant/nonfiring cells. However, cells with fully disordered CRU positions could not reach low firing rates and their β-adrenergic-receptor stimulation effect was substantially decreased. Inclusion of Cav1.3, a low-voltage activation L-type Ca channel isoform into ICaL, strongly increases recruitment of CRUs to fire during diastolic depolarization, increasing robustness of pacemaking and complementing effects of CRU distribution. Thus, order/disorder in CRU locations along with Cav1.3 expression regulates pacemaker function via synchronization of CRU firing. Excessive CRU disorder and/or overexpression of Cav1.3 boosts pacemaker function in the basal state, but limits the rate range, which may contribute to heart rate range decline with age and disease.
Collapse
Affiliation(s)
- Anna V. Maltsev
- School of Mathematics, Queen Mary University of London, London, UK
| | - Michael D. Stern
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Victor A. Maltsev
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD
| |
Collapse
|
27
|
Liu J, Kasuya G, Zempo B, Nakajo K. Two HCN4 Channels Play Functional Roles in the Zebrafish Heart. Front Physiol 2022; 13:901571. [PMID: 35846012 PMCID: PMC9281569 DOI: 10.3389/fphys.2022.901571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
The HCN4 channel is essential for heart rate regulation in vertebrates by generating pacemaker potentials in the sinoatrial node. HCN4 channel abnormality may cause bradycardia and sick sinus syndrome, making it an important target for clinical research and drug discovery. The zebrafish is a popular animal model for cardiovascular research. They are potentially suitable for studying inherited heart diseases, including cardiac arrhythmia. However, it has not been determined how similar the ion channels that underlie cardiac automaticity are in zebrafish and humans. In the case of HCN4, humans have one gene, whereas zebrafish have two ortholog genes (DrHCN4 and DrHCN4L; ‘Dr’ referring to Danio rerio). However, it is not known whether the two HCN4 channels have different physiological functions and roles in heart rate regulation. In this study, we characterized the biophysical properties of the two zebrafish HCN4 channels in Xenopus oocytes and compared them to those of the human HCN4 channel. We found that they showed different gating properties: DrHCN4L currents showed faster activation kinetics and a more positively shifted G-V curve than did DrHCN4 and human HCN4 currents. We made chimeric channels of DrHCN4 and DrHCN4L and found that cytoplasmic domains were determinants for the faster activation and the positively shifted G-V relationship in DrHCN4L. The use of a dominant-negative HCN4 mutant confirmed that DrHCN4 and DrHCN4L can form a heteromultimeric channel in Xenopus oocytes. Next, we confirmed that both are sensitive to common HCN channel inhibitors/blockers including Cs+, ivabradine, and ZD7288. These HCN inhibitors successfully lowered zebrafish heart rate during early embryonic stages. Finally, we knocked down the HCN4 genes using antisense morpholino and found that knocking down either or both of the HCN4 channels caused a temporal decrease in heart rate and tended to cause pericardial edema. These findings suggest that both DrHCN4 and DrHCN4L play a significant role in zebrafish heart rate regulation.
Collapse
|
28
|
D'Souza A, Boink GJJ, Toyoda F, Mesirca P. Editorial: Cardiac Pacemaking in Health and Disease: From Genes to Function. Front Physiol 2022; 13:913506. [PMID: 35711314 PMCID: PMC9197676 DOI: 10.3389/fphys.2022.913506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Alicia D'Souza
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Gerard J J Boink
- Departments of Cardiology and Medical Biology, Amsterdam University Medical Centers, Location University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Cardiovascular Sciences, Research Program: Heart Failure and Arrhythmias, Amsterdam, Netherlands
| | - Futoshi Toyoda
- Department of Physiology, Shiga University of Medical Science, Otsu, Japan
| | - Pietro Mesirca
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France.,LabEx Ion Channels Science and Therapeutics, Montpellier, France
| |
Collapse
|
29
|
Nayir S, Lacour SP, Kucera JP. Active force generation contributes to the complexity of spontaneous activity and to the response to stretch of murine cardiomyocyte cultures. J Physiol 2022; 600:3287-3312. [PMID: 35679256 PMCID: PMC9541716 DOI: 10.1113/jp283083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/01/2022] [Indexed: 11/17/2022] Open
Abstract
Abstract Cardiomyocyte cultures exhibit spontaneous electrical and contractile activity, as in a natural cardiac pacemaker. In such preparations, beat rate variability exhibits features similar to those of heart rate variability in vivo. Mechanical deformations and forces feed back on the electrical properties of cardiomyocytes, but it is not fully elucidated how this mechano‐electrical interplay affects beating variability in such preparations. Using stretchable microelectrode arrays, we assessed the effects of the myosin inhibitor blebbistatin and the non‐selective stretch‐activated channel blocker streptomycin on beating variability and on the response of neonatal or fetal murine ventricular cell cultures against deformation. Spontaneous electrical activity was recorded without stretch and upon predefined deformation protocols (5% uniaxial and 2% equibiaxial strain, applied repeatedly for 1 min every 3 min). Without stretch, spontaneous activity originated from the edge of the preparations, and its site of origin switched frequently in a complex manner across the cultures. Blebbistatin did not change mean beat rate, but it decreased the spatial complexity of spontaneous activity. In contrast, streptomycin did not exert any manifest effects. During the deformation protocols, beat rate increased transiently upon stretch but, paradoxically, also upon release. Blebbistatin attenuated the response to stretch, whereas this response was not affected by streptomycin. Therefore, our data support the notion that in a spontaneously firing network of cardiomyocytes, active force generation, rather than stretch‐activated channels, is involved mechanistically in the complexity of the spatiotemporal patterns of spontaneous activity and in the stretch‐induced acceleration of beating.
![]() Key points Monolayer cultures of cardiac cells exhibit spontaneous electrical and contractile activity, as in a natural cardiac pacemaker. Beating variability in these preparations recapitulates the power‐law behaviour of heart rate variability in vivo. However, the effects of mechano‐electrical feedback on beating variability are not yet fully understood. Using stretchable microelectrode arrays, we examined the effects of the contraction uncoupler blebbistatin and the non‐specific stretch‐activated channel blocker streptomycin on beating variability and on stretch‐induced changes of beat rate. Without stretch, blebbistatin decreased the spatial complexity of beating variability, whereas streptomycin had no effects. Both stretch and release increased beat rate transiently; blebbistatin attenuated the increase of beat rate upon stretch, whereas streptomycin had no effects. Active force generation contributes to the complexity of spatiotemporal patterns of beating variability and to the increase of beat rate upon mechanical deformation. Our study contributes to the understanding of how mechano‐electrical feedback influences heart rate variability.
Collapse
Affiliation(s)
- Seyma Nayir
- Department of Physiology, University of Bern, Bern, Switzerland
| | | | - Jan P Kucera
- Department of Physiology, University of Bern, Bern, Switzerland
| |
Collapse
|
30
|
Oniani T, Vinnenberg L, Chaudhary R, Schreiber JA, Riske K, Williams B, Pape HC, White JA, Junker A, Seebohm G, Meuth SG, Hundehege P, Budde T, Zobeiri M. Effects of Axonal Demyelination, Inflammatory Cytokines and Divalent Cation Chelators on Thalamic HCN Channels and Oscillatory Bursting. Int J Mol Sci 2022; 23:ijms23116285. [PMID: 35682964 PMCID: PMC9181513 DOI: 10.3390/ijms23116285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/31/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) is a demyelinating disease of the central nervous system that is characterized by the progressive loss of oligodendrocytes and myelin and is associated with thalamic dysfunction. Cuprizone (CPZ)-induced general demyelination in rodents is a valuable model for studying different aspects of MS pathology. CPZ feeding is associated with the altered distribution and expression of different ion channels along neuronal somata and axons. However, it is largely unknown whether the copper chelator CPZ directly influences ion channels. Therefore, we assessed the effects of different divalent cations (copper; zinc) and trace metal chelators (EDTA; Tricine; the water-soluble derivative of CPZ, BiMPi) on hyperpolarization-activated cyclic nucleotide-gated (HCN) channels that are major mediators of thalamic function and pathology. In addition, alterations of HCN channels induced by CPZ treatment and MS-related proinflammatory cytokines (IL-1β; IL-6; INF-α; INF-β) were characterized in C57Bl/6J mice. Thus, the hyperpolarization-activated inward current (Ih) was recorded in thalamocortical (TC) neurons and heterologous expression systems (mHCN2 expressing HEK cells; hHCN4 expressing oocytes). A number of electrophysiological characteristics of Ih (potential of half-maximal activation (V0.5); current density; activation kinetics) were unchanged following the extracellular application of trace metals and divalent cation chelators to native neurons, cell cultures or oocytes. Mice were fed a diet containing 0.2% CPZ for 35 days, resulting in general demyelination in the brain. Withdrawal of CPZ from the diet resulted in rapid remyelination, the effects of which were assessed at three time points after stopping CPZ feeding (Day1, Day7, Day25). In TC neurons, Ih was decreased on Day1 and Day25 and revealed a transient increased availability on Day7. In addition, we challenged naive TC neurons with INF-α and IL-1β. It was found that Ih parameters were differentially altered by the application of the two cytokines to thalamic cells, while IL-1β increased the availability of HCN channels (depolarized V0.5; increased current density) and the excitability of TC neurons (depolarized resting membrane potential (RMP); increased the number of action potentials (APs); produced a larger voltage sag; promoted higher input resistance; increased the number of burst spikes; hyperpolarized the AP threshold), INF-α mediated contrary effects. The effect of cytokine modulation on thalamic bursting was further assessed in horizontal slices and a computational model of slow thalamic oscillations. Here, IL-1β and INF-α increased and reduced oscillatory bursting, respectively. We conclude that HCN channels are not directly modulated by trace metals and divalent cation chelators but are subject to modulation by different MS-related cytokines.
Collapse
Affiliation(s)
- Tengiz Oniani
- Institute of Physiology I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149 Münster, Germany; (T.O.); (R.C.); (H.-C.P.); (M.Z.)
| | - Laura Vinnenberg
- Department of Neurology with Institute of Translational Neurology, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany; (L.V.); (P.H.)
| | - Rahul Chaudhary
- Institute of Physiology I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149 Münster, Germany; (T.O.); (R.C.); (H.-C.P.); (M.Z.)
| | - Julian A. Schreiber
- Institute of Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universität, Corren-Str. 48, D-48149 Münster, Germany;
- Cellular Electrophysiology and Molecular Biology, Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Robert-Koch-Str. 45, D-48149 Münster, Germany;
| | - Kathrin Riske
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität, Waldeyer-Str. 15, D-48149 Münster, Germany; (K.R.); (A.J.)
| | - Brandon Williams
- Center for Systems Neuroscience, Neurophotonics Center, Department of Biomedical Engineering, Boston University, 610 Commonwealth Ave., Boston, MA 02215, USA; (B.W.); (J.A.W.)
| | - Hans-Christian Pape
- Institute of Physiology I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149 Münster, Germany; (T.O.); (R.C.); (H.-C.P.); (M.Z.)
| | - John A. White
- Center for Systems Neuroscience, Neurophotonics Center, Department of Biomedical Engineering, Boston University, 610 Commonwealth Ave., Boston, MA 02215, USA; (B.W.); (J.A.W.)
| | - Anna Junker
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität, Waldeyer-Str. 15, D-48149 Münster, Germany; (K.R.); (A.J.)
| | - Guiscard Seebohm
- Cellular Electrophysiology and Molecular Biology, Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Robert-Koch-Str. 45, D-48149 Münster, Germany;
| | - Sven G. Meuth
- Neurology Clinic, University Clinic Düsseldorf, Moorenstraße 5, D-40225 Düsseldorf, Germany;
| | - Petra Hundehege
- Department of Neurology with Institute of Translational Neurology, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany; (L.V.); (P.H.)
| | - Thomas Budde
- Institute of Physiology I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149 Münster, Germany; (T.O.); (R.C.); (H.-C.P.); (M.Z.)
- Correspondence:
| | - Mehrnoush Zobeiri
- Institute of Physiology I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149 Münster, Germany; (T.O.); (R.C.); (H.-C.P.); (M.Z.)
| |
Collapse
|
31
|
Chen K, Wang S, Sun Z. In Vivo Cardiac-specific Expression of Adenylyl Cyclase 4 Gene Protects against Klotho Deficiency-induced Heart Failure. Transl Res 2022; 244:101-113. [PMID: 35114419 PMCID: PMC9119924 DOI: 10.1016/j.trsl.2022.01.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/10/2022] [Accepted: 01/26/2022] [Indexed: 01/10/2023]
Abstract
Klotho is an aging-suppressor gene. Klotho gene deficiency causes heart failure in Klotho-hypomorphic mutant (KL (-/-)) mice. RNA-seq and western blot analysis showed that adenylyl cyclase type IV (AC4) mRNA and protein expression was largely decreased in cardiomyocytes of KL (-/-) mice. The objective of this study was to investigate whether in vivo cardiac-specific expression of AC4 gene protects against Klotho deficiency-induced heart failure. Interestingly, in vivo AAV-based cardiac-specific AC4 gene expression increased left ventricular fractional shortening, ejection fraction, stroke volume, and left ventricular end-diastolic volume in KL (-/-) mice, suggesting that cardiac-specific AC4 gene expression improves Klotho deficiency-induced heart dysfunction. Cardiac-specific AC4 gene expression also decreased Klotho deficiency-induced cardiac hypertrophy. Cardiac-specific AC4 gene expression alleviated Klotho deficiency-induced cardiac fibrosis and calcification. Furthermore, cardiac-specific AC4 gene expression attenuated mitochondrial dysfunction, superoxide accumulation and cardiomyocyte apoptotic cell death. Thus, downregulation of AC4 may contribute to Klotho deficiency-induced heart failure. Mechanistically, AAV2/9-αMHC-AC4 increased cardiomyocytic cAMP levels and thus regulated the PKA-PLN-SERCA2 signal pathway, which is critical in modulating calcium flux and mitochondrial function. In conclusion, cardiac-specific AC4 gene expression protects against Klotho deficiency-induced heart failure through increasing cardiomyocytic cAMP levels, which alleviates cAMP-dependent mitochondrial dysfunction, superoxide accumulation and apoptotic cell death. AC4 regulates superoxide levels via the cAMP-PKA pathway. AC4 could be a potential therapeutic target for heart failure associated with Klotho deficiency. Heart failure is the major cause of mortality in patients with chronic kidney disease (CKD). A decrease in Klotho levels is linked to CKD.
Collapse
Affiliation(s)
- Kai Chen
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America; Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Shirley Wang
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America; Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Zhongjie Sun
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America; Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America.
| |
Collapse
|
32
|
Paradigm shift: new concepts for HCN4 function in cardiac pacemaking. Pflugers Arch 2022; 474:649-663. [PMID: 35556164 PMCID: PMC9192375 DOI: 10.1007/s00424-022-02698-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/29/2022] [Indexed: 11/05/2022]
Abstract
Hyperpolarization-activated cyclic nucleotide–gated (HCN) channels are the molecular correlate of the If current and are critically involved in controlling neuronal excitability and the autonomous rhythm of the heart. The HCN4 isoform is the main HCN channel subtype expressed in the sinoatrial node (SAN), a tissue composed of specialized pacemaker cells responsible for generating the intrinsic heartbeat. More than 40 years ago, the If current was first discovered in rabbit SAN tissue. Along with this discovery, a theory was proposed that cyclic adenosine monophosphate–dependent modulation of If mediates heart rate regulation by the autonomic nervous system—a process called chronotropic effect. However, up to the present day, this classical theory could not be reliably validated. Recently, new concepts emerged confirming that HCN4 channels indeed play an important role in heart rate regulation. However, the cellular mechanism by which HCN4 controls heart rate turned out to be completely different than originally postulated. Here, we review the latest findings regarding the physiological role of HCN4 in the SAN. We describe a newly discovered mechanism underlying heart rate regulation by HCN4 at the tissue and single cell levels, and we discuss these observations in the context of results from previously studied HCN4 mouse models.
Collapse
|
33
|
Hennis K, Rötzer RD, Rilling J, Wu Y, Thalhammer SB, Biel M, Wahl-Schott C, Fenske S. In vivo and ex vivo electrophysiological study of the mouse heart to characterize the cardiac conduction system, including atrial and ventricular vulnerability. Nat Protoc 2022; 17:1189-1222. [PMID: 35314849 DOI: 10.1038/s41596-021-00678-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/09/2021] [Indexed: 01/05/2023]
Abstract
The mouse is a common and cost-effective animal model for basic research, and the number of genetically engineered mouse models with cardiac phenotype is increasing. In vivo electrophysiological study in mice is similar to that performed in humans. It is indispensable for acquiring intracardiac electrocardiogram recordings and determining baseline cardiac cycle intervals. Furthermore, the use of programmed electrical stimulation enables determination of parameters such as sinoatrial conduction time, sinus node recovery time, atrioventricular-nodal conduction properties, Wenckebach periodicity, refractory periods and arrhythmia vulnerability. This protocol describes specific procedures for determining these parameters that were adapted from analogous human protocols for use in mice. We include details of ex vivo electrophysiological study, which provides detailed insights into intrinsic cardiac electrophysiology without external influences from humoral and neural factors. In addition, we describe a heart preparation with intact innervation by the vagus nerve that can be used as an ex vivo model for vagal control of the cardiac conduction system. Data acquisition for in vivo and ex vivo electrophysiological study takes ~1 h per mouse, depending on the number of stimulation protocols applied during the procedure. The technique yields highly reliable results and can be used for phenotyping of cardiac disease models, elucidating disease mechanisms and confirming functional improvements in gene therapy approaches as well as for drug and toxicity testing.
Collapse
Affiliation(s)
- Konstantin Hennis
- Center for Drug Research, Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - René D Rötzer
- Center for Drug Research, Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Julia Rilling
- Center for Drug Research, Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Yakun Wu
- Center for Drug Research, Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Stefan B Thalhammer
- Center for Drug Research, Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Martin Biel
- Center for Drug Research, Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | | | - Stefanie Fenske
- Center for Drug Research, Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany.
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
34
|
Maltsev AV, Stern MD, Lakatta EG, Maltsev VA. Functional Heterogeneity of Cell Populations Increases Robustness of Pacemaker Function in a Numerical Model of the Sinoatrial Node Tissue. Front Physiol 2022; 13:845634. [PMID: 35574456 PMCID: PMC9091312 DOI: 10.3389/fphys.2022.845634] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/15/2022] [Indexed: 11/19/2022] Open
Abstract
Each heartbeat is initiated by specialized pacemaker cells operating within the sinoatrial node (SAN). While individual cells within SAN tissue exhibit substantial heterogeneity of their electrophysiological parameters and Ca cycling, the role of this heterogeneity for cardiac pacemaker function remains mainly unknown. Here we investigated the problem numerically in a 25 × 25 square grid of connected coupled-clock Maltsev-Lakatta cell models. The tissue models were populated by cells with different degree of heterogeneity of the two key model parameters, maximum L-type Ca current conductance (gCaL) and sarcoplasmic reticulum Ca pumping rate (Pup). Our simulations showed that in the areas of Pup-gCaL parametric space at the edge of the system stability, where action potential (AP) firing is absent or dysrhythmic in SAN tissue models populated with identical cells, rhythmic AP firing can be rescued by populating the tissues with heterogeneous cells. This robust SAN function is synergistic with respect to heterogeneity in gCaL and Pup and can be further strengthened by clustering of cells with similar properties. The effect of cell heterogeneity is not due to a simple summation of activity of intrinsically firing cells naturally present in heterogeneous SAN; rather AP firing cells locally and critically interact with non-firing/dormant cells. When firing cells prevail, they recruit many dormant cells to fire, strongly enhancing overall SAN function; and vice versa, prevailing dormant cells suppress AP firing in cells with intrinsic automaticity and halt SAN function. The transitions between firing and non-firing states of the system are sharp, resembling phase transitions in statistical physics. Furthermore, robust function of heterogeneous SAN tissue requires weak cell coupling, a known property of the central area of SAN where cardiac impulse emerges; stronger cell coupling reduces AP firing rate and ultimately halts SAN automaticity at the edge of stability.
Collapse
|
35
|
Jiménez A, Lu Y, Jambhekar A, Lahav G. Principles, mechanisms and functions of entrainment in biological oscillators. Interface Focus 2022; 12:20210088. [PMID: 35450280 PMCID: PMC9010850 DOI: 10.1098/rsfs.2021.0088] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/07/2022] [Indexed: 12/12/2022] Open
Abstract
Entrainment is a phenomenon in which two oscillators interact with each other, typically through physical or chemical means, to synchronize their oscillations. This phenomenon occurs in biology to coordinate processes from the molecular to organismal scale. Biological oscillators can be entrained within a single cell, between cells or to an external input. Using six illustrative examples of entrainable biological oscillators, we discuss the distinctions between entrainment and synchrony and explore features that contribute to a system's propensity to entrain. Entrainment can either enhance or reduce the heterogeneity of oscillations within a cell population, and we provide examples and mechanisms of each case. Finally, we discuss the known functions of entrainment and discuss potential functions from an evolutionary perspective.
Collapse
Affiliation(s)
- Alba Jiménez
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA
| | - Ying Lu
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA
| | - Ashwini Jambhekar
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA
- Ludwig Center at Harvard, Boston, MA 02115, USA
| | - Galit Lahav
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA
- Ludwig Center at Harvard, Boston, MA 02115, USA
| |
Collapse
|
36
|
Hu W, Clark RB, Giles WR, Kondo C, Zhang H. Frequency-Dependent Properties of the Hyperpolarization-Activated Cation Current, I f, in Adult Mouse Heart Primary Pacemaker Myocytes. Int J Mol Sci 2022; 23:4299. [PMID: 35457119 PMCID: PMC9024942 DOI: 10.3390/ijms23084299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/05/2022] [Accepted: 04/11/2022] [Indexed: 02/04/2023] Open
Abstract
A number of distinct electrophysiological mechanisms that modulate the myogenic spontaneous pacemaker activity in the sinoatrial node (SAN) of the mammalian heart have been investigated extensively. There is agreement that several (3 or 4) different transmembrane ionic current changes (referred to as the voltage clock) are involved; and that the resulting net current interacts with direct and indirect effects of changes in intracellular Ca2+ (the calcium clock). However, significant uncertainties, and important knowledge gaps, remain concerning the functional roles in SAN spontaneous pacing of many of the individual ion channel- or exchanger-mediated transmembrane current changes. We report results from patch clamp studies and mathematical modeling of the hyperpolarization-activated current, If, in the generation/modulation of the diastolic depolarization, or pacemaker potential, produced by individual myocytes that were enzymatically isolated from the adult mouse sinoatrial node (SAN). Amphotericin-mediated patch microelectrode recordings at 35 °C were made under control conditions and in the presence of 5 or 10 nM isoproterenol (ISO). These sets of results were complemented and integrated with mathematical modeling of the current changes that take place in the range of membrane potentials (-70 to -50 mV), which corresponds to the 'pacemaker depolarization' in the adult mouse SAN. Our results reveal a very small, but functionally important, approximately steady-state or time-independent current generated by residual activation of If channels that are expressed in these pacemaker myocytes. Recordings of the pacemaker depolarization and action potential, combined with measurements of changes in If, and the well-known increases in the L-type Ca2+ current, ICaL, demonstrated that ICaL activation, is essential for myogenic pacing. Moreover, after being enhanced (approximately 3-fold) by 5 or 10 nM ISO, ICaL contributes significantly to the positive chronotropic effect. Our mathematical model has been developed in an attempt to better understand the underlying mechanisms for the pacemaker depolarization and action potential in adult mouse SAN myocytes. After being updated with our new experimental data describing If, our simulations reveal a novel functional component of If in adult mouse SAN. Computational work carried out with this model also confirms that in the presence of ISO the residual activation of If and opening of ICaL channels combine to generate a net current change during the slow diastolic depolarization phase that is essential for the observed accelerated pacemaking rate of these SAN myocytes.
Collapse
Affiliation(s)
- Wei Hu
- Biological Physics Group, Department of Physics and Astronomy, The University of Manchester, Manchester M13 9PL, UK;
| | - Robert B. Clark
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (R.B.C.); (W.R.G.); (C.K.)
| | - Wayne R. Giles
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (R.B.C.); (W.R.G.); (C.K.)
| | - Colleen Kondo
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (R.B.C.); (W.R.G.); (C.K.)
| | - Henggui Zhang
- Biological Physics Group, Department of Physics and Astronomy, The University of Manchester, Manchester M13 9PL, UK;
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646099, China
| |
Collapse
|
37
|
Louradour J, Bortolotti O, Torre E, Bidaud I, Lamb N, Fernandez A, Le Guennec JY, Mangoni ME, Mesirca P. L-Type Cav1.3 Calcium Channels Are Required for Beta-Adrenergic Triggered Automaticity in Dormant Mouse Sinoatrial Pacemaker Cells. Cells 2022; 11:cells11071114. [PMID: 35406677 PMCID: PMC8997967 DOI: 10.3390/cells11071114] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/18/2022] [Accepted: 03/23/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Sinoatrial node cells (SANC) automaticity is generated by functional association between the activity of plasmalemmal ion channels and local diastolic intracellular Ca2+ release (LCR) from ryanodine receptors. Strikingly, most isolated SANC exhibit a “dormant” state, whereas only a fraction shows regular firing as observed in intact SAN. Recent studies showed that β-adrenergic stimulation can initiate spontaneous firing in dormant SANC, though this mechanism is not entirely understood. Methods: To investigate the role of L-type Cav1.3 Ca2+ channels in the adrenergic regulation of automaticity in dormant SANC, we used a knock-in mouse strain in which the sensitivity of L-type Cav1.2 α1 subunits to dihydropyridines (DHPs) was inactivated (Cav1.2DHP−/−), enabling the selective pharmacological inhibition of Cav1.3 by DHPs. Results: In dormant SANC, β-adrenergic stimulation with isoproterenol (ISO) induced spontaneous action potentials (AP) and Ca2+ transients, which were completely arrested with concomitant perfusion of the DHP nifedipine. In spontaneously firing SANC at baseline, Cav1.3 inhibition completely reversed the effect of β-adrenergic stimulation on AP and the frequency of Ca2+ transients. Confocal calcium imaging of SANC showed that the β-adrenergic-induced synchronization of LCRs is regulated by the activity of Cav1.3 channels. Conclusions: Our study shows a novel role of Cav1.3 channels in initiating and maintaining automaticity in dormant SANC upon β-adrenergic stimulation.
Collapse
Affiliation(s)
- Julien Louradour
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34090 Montpellier, France; (J.L.); (O.B.); (E.T.); (I.B.)
- LabEx Ion Channels Science and Therapeutics (ICST), 34090 Montpellier, France
- PhyMedExp, Université de Montpellier, INSERM U1046, UMR CNRS, 34090 Montpellier, France;
| | - Olivier Bortolotti
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34090 Montpellier, France; (J.L.); (O.B.); (E.T.); (I.B.)
- LabEx Ion Channels Science and Therapeutics (ICST), 34090 Montpellier, France
| | - Eleonora Torre
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34090 Montpellier, France; (J.L.); (O.B.); (E.T.); (I.B.)
- LabEx Ion Channels Science and Therapeutics (ICST), 34090 Montpellier, France
| | - Isabelle Bidaud
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34090 Montpellier, France; (J.L.); (O.B.); (E.T.); (I.B.)
- LabEx Ion Channels Science and Therapeutics (ICST), 34090 Montpellier, France
| | - Ned Lamb
- Mammalian Stem Cell Biology Group, Institute of Human Genetics, Université de Montpellier, CNRS, 34090 Montpellier, France; (N.L.); (A.F.)
| | - Anne Fernandez
- Mammalian Stem Cell Biology Group, Institute of Human Genetics, Université de Montpellier, CNRS, 34090 Montpellier, France; (N.L.); (A.F.)
| | - Jean-Yves Le Guennec
- PhyMedExp, Université de Montpellier, INSERM U1046, UMR CNRS, 34090 Montpellier, France;
| | - Matteo E. Mangoni
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34090 Montpellier, France; (J.L.); (O.B.); (E.T.); (I.B.)
- LabEx Ion Channels Science and Therapeutics (ICST), 34090 Montpellier, France
- Correspondence: (M.E.M.); (P.M.)
| | - Pietro Mesirca
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34090 Montpellier, France; (J.L.); (O.B.); (E.T.); (I.B.)
- LabEx Ion Channels Science and Therapeutics (ICST), 34090 Montpellier, France
- Correspondence: (M.E.M.); (P.M.)
| |
Collapse
|
38
|
Tibbo AJ, Mika D, Dobi S, Ling J, McFall A, Tejeda GS, Blair C, MacLeod R, MacQuaide N, Gök C, Fuller W, Smith BO, Smith GL, Vandecasteele G, Brand T, Baillie GS. Phosphodiesterase type 4 anchoring regulates cAMP signaling to Popeye domain-containing proteins. J Mol Cell Cardiol 2022; 165:86-102. [PMID: 34999055 PMCID: PMC8986152 DOI: 10.1016/j.yjmcc.2022.01.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/16/2021] [Accepted: 01/03/2022] [Indexed: 12/04/2022]
Abstract
Cyclic AMP is a ubiquitous second messenger used to transduce intracellular signals from a variety of Gs-coupled receptors. Compartmentalisation of protein intermediates within the cAMP signaling pathway underpins receptor-specific responses. The cAMP effector proteins protein-kinase A and EPAC are found in complexes that also contain phosphodiesterases whose presence ensures a coordinated cellular response to receptor activation events. Popeye domain containing (POPDC) proteins are the most recent class of cAMP effectors to be identified and have crucial roles in cardiac pacemaking and conduction. We report the first observation that POPDC proteins exist in complexes with members of the PDE4 family in cardiac myocytes. We show that POPDC1 preferentially binds the PDE4A sub-family via a specificity motif in the PDE4 UCR1 region and that PDE4s bind to the Popeye domain of POPDC1 in a region known to be susceptible to a mutation that causes human disease. Using a cell-permeable disruptor peptide that displaces the POPDC1-PDE4 complex we show that PDE4 activity localized to POPDC1 modulates cycle length of spontaneous Ca2+ transients firing in intact mouse sinoatrial nodes. POPDC1 forms a complex with type 4 phosphodiesterases (PDE4s) in cardiac myocytes. POPDC1 binds PDE4 enzymes in the Upstream Conserved Region 1 (UCR1) domain. The PDE4 binding motif within the Popeye domain lies in a region that harbours a mutation, which underpins human disease. Disruption of the POPDC1-PDE4 complex modulates the cycle length of spontaneous Ca2+ transients in the sinoatrial node. Disruption of the POPDC1-PDE4 complex causes a significant prolongation of the action potential repolarization phase.
Collapse
Affiliation(s)
- Amy J Tibbo
- College of Veterinary, Medical and Life Sciences, University of Glasgow, Glasgow G128QQ, UK
| | - Delphine Mika
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, 92296 Châtenay-Malabry, France
| | - Sara Dobi
- College of Veterinary, Medical and Life Sciences, University of Glasgow, Glasgow G128QQ, UK
| | - Jiayue Ling
- College of Veterinary, Medical and Life Sciences, University of Glasgow, Glasgow G128QQ, UK
| | - Aisling McFall
- College of Veterinary, Medical and Life Sciences, University of Glasgow, Glasgow G128QQ, UK
| | - Gonzalo S Tejeda
- College of Veterinary, Medical and Life Sciences, University of Glasgow, Glasgow G128QQ, UK
| | - Connor Blair
- College of Veterinary, Medical and Life Sciences, University of Glasgow, Glasgow G128QQ, UK
| | - Ruth MacLeod
- College of Veterinary, Medical and Life Sciences, University of Glasgow, Glasgow G128QQ, UK
| | - Niall MacQuaide
- School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Caglar Gök
- College of Veterinary, Medical and Life Sciences, University of Glasgow, Glasgow G128QQ, UK
| | - William Fuller
- College of Veterinary, Medical and Life Sciences, University of Glasgow, Glasgow G128QQ, UK
| | - Brian O Smith
- College of Veterinary, Medical and Life Sciences, University of Glasgow, Glasgow G128QQ, UK
| | - Godfrey L Smith
- College of Veterinary, Medical and Life Sciences, University of Glasgow, Glasgow G128QQ, UK
| | - Grégoire Vandecasteele
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, 92296 Châtenay-Malabry, France
| | - Thomas Brand
- National Heart and Lung Institute, Imperial College, W12 0NN, London
| | - George S Baillie
- College of Veterinary, Medical and Life Sciences, University of Glasgow, Glasgow G128QQ, UK.
| |
Collapse
|
39
|
Al Kury LT, Chacar S, Alefishat E, Khraibi AA, Nader M. Structural and Electrical Remodeling of the Sinoatrial Node in Diabetes: New Dimensions and Perspectives. Front Endocrinol (Lausanne) 2022; 13:946313. [PMID: 35872997 PMCID: PMC9302195 DOI: 10.3389/fendo.2022.946313] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 06/14/2022] [Indexed: 11/14/2022] Open
Abstract
The sinoatrial node (SAN) is composed of highly specialized cells that mandate the spontaneous beating of the heart through self-generation of an action potential (AP). Despite this automaticity, the SAN is under the modulation of the autonomic nervous system (ANS). In diabetes mellitus (DM), heart rate variability (HRV) manifests as a hallmark of diabetic cardiomyopathy. This is paralleled by an impaired regulation of the ANS, and by a pathological remodeling of the pacemaker structure and function. The direct effect of diabetes on the molecular signatures underscoring this pathology remains ill-defined. The recent focus on the electrical currents of the SAN in diabetes revealed a repressed firing rate of the AP and an elongation of its tracing, along with conduction abnormalities and contractile failure. These changes are blamed on the decreased expression of ion transporters and cell-cell communication ports at the SAN (i.e., HCN4, calcium and potassium channels, connexins 40, 45, and 46) which further promotes arrhythmias. Molecular analysis crystallized the RGS4 (regulator of potassium currents), mitochondrial thioredoxin-2 (reactive oxygen species; ROS scavenger), and the calcium-dependent calmodulin kinase II (CaMKII) as metabolic culprits of relaying the pathological remodeling of the SAN cells (SANCs) structure and function. A special attention is given to the oxidation of CaMKII and the generation of ROS that induce cell damage and apoptosis of diabetic SANCs. Consequently, the diabetic SAN contains a reduced number of cells with significant infiltration of fibrotic tissues that further delay the conduction of the AP between the SANCs. Failure of a genuine generation of AP and conduction of their derivative waves to the neighboring atrial myocardium may also occur as a result of the anti-diabetic regiment (both acute and/or chronic treatments). All together, these changes pose a challenge in the field of cardiology and call for further investigations to understand the etiology of the structural/functional remodeling of the SANCs in diabetes. Such an understanding may lead to more adequate therapies that can optimize glycemic control and improve health-related outcomes in patients with diabetes.
Collapse
Affiliation(s)
- Lina T. Al Kury
- Department of Health Sciences, College of Natural and Health Sciences, Zayed University, Abu Dhabi, United Arab Emirates
- *Correspondence: Lina T. Al Kury, ; Moni Nader,
| | - Stephanie Chacar
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Eman Alefishat
- Department of Pharmacology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman, Jordan
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Ali A. Khraibi
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Moni Nader
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- *Correspondence: Lina T. Al Kury, ; Moni Nader,
| |
Collapse
|
40
|
Dixon RE, Navedo MF, Binder MD, Santana LF. Mechanisms and Physiological Implications of Cooperative Gating of Ion Channels Clusters. Physiol Rev 2021; 102:1159-1210. [PMID: 34927454 DOI: 10.1152/physrev.00022.2021] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Ion channels play a central role in the regulation of nearly every cellular process. Dating back to the classic 1952 Hodgkin-Huxley model of the generation of the action potential, ion channels have always been thought of as independent agents. A myriad of recent experimental findings exploiting advances in electrophysiology, structural biology, and imaging techniques, however, have posed a serious challenge to this long-held axiom as several classes of ion channels appear to open and close in a coordinated, cooperative manner. Ion channel cooperativity ranges from variable-sized oligomeric cooperative gating in voltage-gated, dihydropyridine-sensitive Cav1.2 and Cav1.3 channels to obligatory dimeric assembly and gating of voltage-gated Nav1.5 channels. Potassium channels, transient receptor potential channels, hyperpolarization cyclic nucleotide-activated channels, ryanodine receptors (RyRs), and inositol trisphosphate receptors (IP3Rs) have also been shown to gate cooperatively. The implications of cooperative gating of these ion channels range from fine tuning excitation-contraction coupling in muscle cells to regulating cardiac function and vascular tone, to modulation of action potential and conduction velocity in neurons and cardiac cells, and to control of pace-making activity in the heart. In this review, we discuss the mechanisms leading to cooperative gating of ion channels, their physiological consequences and how alterations in cooperative gating of ion channels may induce a range of clinically significant pathologies.
Collapse
Affiliation(s)
- Rose Ellen Dixon
- Department of Physiology and Membrane Biology, University of California, Davis, CA, United States
| | - Manuel F Navedo
- Department of Pharmacology, University of California, Davis, CA, United States
| | - Marc D Binder
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States
| | - L Fernando Santana
- Department of Physiology and Membrane Biology, University of California, Davis, CA, United States
| |
Collapse
|
41
|
Gruscheski L, Brand T. The Role of POPDC Proteins in Cardiac Pacemaking and Conduction. J Cardiovasc Dev Dis 2021; 8:160. [PMID: 34940515 PMCID: PMC8706714 DOI: 10.3390/jcdd8120160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/17/2021] [Accepted: 11/20/2021] [Indexed: 11/17/2022] Open
Abstract
The Popeye domain-containing (POPDC) gene family, consisting of Popdc1 (also known as Bves), Popdc2, and Popdc3, encodes transmembrane proteins abundantly expressed in striated muscle. POPDC proteins have recently been identified as cAMP effector proteins and have been proposed to be part of the protein network involved in cAMP signaling. However, their exact biochemical activity is presently poorly understood. Loss-of-function mutations in animal models causes abnormalities in skeletal muscle regeneration, conduction, and heart rate adaptation after stress. Likewise, patients carrying missense or nonsense mutations in POPDC genes have been associated with cardiac arrhythmias and limb-girdle muscular dystrophy. In this review, we introduce the POPDC protein family, and describe their structure function, and role in cAMP signaling. Furthermore, the pathological phenotypes observed in zebrafish and mouse models and the clinical and molecular pathologies in patients carrying POPDC mutations are described.
Collapse
Affiliation(s)
| | - Thomas Brand
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK;
| |
Collapse
|
42
|
Yang D, Morrell CH, Lyashkov AE, Tagirova Sirenko S, Zahanich I, Yaniv Y, Vinogradova TM, Ziman BD, Maltsev VA, Lakatta EG. Ca 2+ and Membrane Potential Transitions During Action Potentials Are Self-Similar to Each Other and to Variability of AP Firing Intervals Across the Broad Physiologic Range of AP Intervals During Autonomic Receptor Stimulation. Front Physiol 2021; 12:612770. [PMID: 34566668 PMCID: PMC8456031 DOI: 10.3389/fphys.2021.612770] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 06/02/2021] [Indexed: 12/02/2022] Open
Abstract
Ca2+ and V m transitions occurring throughout action potential (AP) cycles in sinoatrial nodal (SAN) cells are cues that (1) not only regulate activation states of molecules operating within criticality (Ca2+ domain) and limit-cycle (V m domain) mechanisms of a coupled-clock system that underlies SAN cell automaticity, (2) but are also regulated by the activation states of the clock molecules they regulate. In other terms, these cues are both causes and effects of clock molecular activation (recursion). Recently, we demonstrated that Ca2+ and V m transitions during AP cycles in single SAN cells isolated from mice, guinea pigs, rabbits, and humans are self-similar (obey a power law) and are also self-similar to trans-species AP firing intervals (APFIs) of these cells in vitro, to heart rate in vivo, and to body mass. Neurotransmitter stimulation of β-adrenergic receptor or cholinergic receptor-initiated signaling in SAN cells modulates their AP firing rate and rhythm by impacting on the degree to which SAN clocks couple to each other, creating the broad physiologic range of SAN cell mean APFIs and firing interval variabilities. Here we show that Ca2+ and V m domain kinetic transitions (time to AP ignition in diastole and 90% AP recovery) occurring within given AP, the mean APFIs, and APFI variabilities within the time series of APs in 230 individual SAN cells are self-similar (obey power laws). In other terms, these long-range correlations inform on self-similar distributions of order among SAN cells across the entire broad physiologic range of SAN APFIs, regardless of whether autonomic receptors of these cells are stimulated or not and regardless of the type (adrenergic or cholinergic) of autonomic receptor stimulation. These long-range correlations among distributions of Ca2+ and V m kinetic functions that regulate SAN cell clock coupling during each AP cycle in different individual, isolated SAN cells not in contact with each other. Our numerical model simulations further extended our perspectives to the molecular scale and demonstrated that many ion currents also behave self-similar across autonomic states. Thus, to ensure rapid flexibility of AP firing rates in response to different types and degrees of autonomic input, nature "did not reinvent molecular wheels within the coupled-clock system of pacemaker cells," but differentially engaged or scaled the kinetics of gears that regulate the rate and rhythm at which the "wheels spin" in a given autonomic input context.
Collapse
Affiliation(s)
- Dongmei Yang
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Christopher H. Morrell
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
- Department of Mathematics and Statistics, Loyola University Maryland, Baltimore, MD, United States
| | - Alexey E. Lyashkov
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Syevda Tagirova Sirenko
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Ihor Zahanich
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Yael Yaniv
- Biomedical Engineering Faculty, Technion–Israel Institute of Technology, Haifa, Israel
| | - Tatiana M. Vinogradova
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Bruce D. Ziman
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Victor A. Maltsev
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Edward G. Lakatta
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
43
|
Shenfu Injection: A Famous Chinese Prescription That Promotes HCN4 Activity in Bone Marrow Mesenchymal Stem Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:9912844. [PMID: 34457032 PMCID: PMC8387162 DOI: 10.1155/2021/9912844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 07/27/2021] [Accepted: 08/08/2021] [Indexed: 11/30/2022]
Abstract
We investigated the effects of Shenfu Injection (SFI) on HCN4 activity in bone marrow mesenchymal stem cells (BMSCs). The sample of BMSCs was divided into six groups: a control group, a high-dose SFI group (0.25 ml/ml), a middle-dose SFI group (0.1 ml/ml), a low-dose SFI group (0.05 ml/ml), an adenovirus-encoded control vector group, and an adenovirus-encoded HCN4 group. Cell ultrastructure was observed using a transmission electron microscope. Quantitative reverse transcription PCR (RT-qPCR) was performed to detect HCN4 expression, and HCN4 activity was detected using the whole-cell patch clamp technique. An enzyme-linked immunosorbent assay was performed to detect cAMP content. Application of flow cytometry confirmed that the isolated cells showed BMSC-like phenotypes. Differentiation of BMSCs in both the SFI and the adenovirus-encoding HCN4 groups occurred according to the cellular ultrastructure. Application of the whole-cell patch clamp technique revealed that SFI could activate the inward pacing current of BMSCs in a concentration-dependent manner. The RT-qPCR results showed that HCN4 expression was significantly higher in the high-dose SFI group than in the medium- and low-dose groups, whereas the cAMP content in the overexpressed HCN4 group decreased significantly; this content in the high-dose SFI group increased significantly. In conclusion, SFI promotes HCN4 activity in BMSCs, which could explain its treatment effect when administered to patients with cardiovascular diseases.
Collapse
|
44
|
Turner D, Kang C, Mesirca P, Hong J, Mangoni ME, Glukhov AV, Sah R. Electrophysiological and Molecular Mechanisms of Sinoatrial Node Mechanosensitivity. Front Cardiovasc Med 2021; 8:662410. [PMID: 34434970 PMCID: PMC8382116 DOI: 10.3389/fcvm.2021.662410] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/24/2021] [Indexed: 01/01/2023] Open
Abstract
The understanding of the electrophysiological mechanisms that underlie mechanosensitivity of the sinoatrial node (SAN), the primary pacemaker of the heart, has been evolving over the past century. The heart is constantly exposed to a dynamic mechanical environment; as such, the SAN has numerous canonical and emerging mechanosensitive ion channels and signaling pathways that govern its ability to respond to both fast (within second or on beat-to-beat manner) and slow (minutes) timescales. This review summarizes the effects of mechanical loading on the SAN activity and reviews putative candidates, including fast mechanoactivated channels (Piezo, TREK, and BK) and slow mechanoresponsive ion channels [including volume-regulated chloride channels and transient receptor potential (TRP)], as well as the components of mechanochemical signal transduction, which may contribute to SAN mechanosensitivity. Furthermore, we examine the structural foundation for both mechano-electrical and mechanochemical signal transduction and discuss the role of specialized membrane nanodomains, namely, caveolae, in mechanical regulation of both membrane and calcium clock components of the so-called coupled-clock pacemaker system responsible for SAN automaticity. Finally, we emphasize how these mechanically activated changes contribute to the pathophysiology of SAN dysfunction and discuss controversial areas necessitating future investigations. Though the exact mechanisms of SAN mechanosensitivity are currently unknown, identification of such components, their impact into SAN pacemaking, and pathological remodeling may provide new therapeutic targets for the treatment of SAN dysfunction and associated rhythm abnormalities.
Collapse
Affiliation(s)
- Daniel Turner
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States
| | - Chen Kang
- Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Pietro Mesirca
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Juan Hong
- Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Matteo E Mangoni
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Alexey V Glukhov
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States
| | - Rajan Sah
- Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
45
|
Zarei Ghobadi M, Emamzadeh R, Teymoori-Rad M, Mozhgani SH. Decoding pathogenesis factors involved in the progression of ATLL or HAM/TSP after infection by HTLV-1 through a systems virology study. Virol J 2021; 18:175. [PMID: 34446027 PMCID: PMC8393454 DOI: 10.1186/s12985-021-01643-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/18/2021] [Indexed: 12/28/2022] Open
Abstract
Background Human T-cell Leukemia Virus type-1 (HTLV-1) is a retrovirus that causes two diseases including Adult T-cell Leukemia/Lymphoma (ATLL cancer) and HTLV-1 Associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP, a neurodegenerative disease) after a long latency period as an asymptomatic carrier (AC). There are no obvious explanations about how each of the mentioned diseases develops in the AC carriers. Finding the discriminative molecular factors and pathways may clarify the destiny of the infection. Methods To shed light on the involved molecular players and activated pathways in each state, differentially co-expressed modules (DiffCoEx) algorithm was employed to identify the highly correlated genes which were co-expressed differently between normal and ACs, ACs and ATLL, as well as ACs and HAM/TSP samples. Through differential pathway analysis, the dysregulated pathways and the specific disease-genes-pathways were figured out. Moreover, the common genes between the member of DiffCoEx and differentially expressed genes were found and the specific genes in ATLL and HAM/TSP were introduced as possible biomarkers. Results The dysregulated genes in the ATLL were mostly enriched in immune and cancer-related pathways while the ones in the HAM/TSP were enriched in immune, inflammation, and neurological pathways. The differential pathway analysis clarified the differences between the gene players in the common activated pathways. Eventually, the final analysis revealed the involvement of specific dysregulated genes including KIRREL2, RAB36, and KANK1 in HAM/TSP as well as LTB4R2, HCN4, FZD9, GRIK5, CREB3L4, TACR2, FRMD1, LHB, FGF3, TEAD3, GRIN2D, GNRH2, PRLH, GPR156, and CRHR2 in ATLL. Conclusion The identified potential prognostic biomarkers and therapeutic targets are proposed as the most important platers in developing ATLL or HAM/TSP. Moreover, the proposed signaling network clarifies the differences between the functional players in the activated pathways in ACs, ATLL, and HAM/TSP. Supplementary Information The online version contains supplementary material available at 10.1186/s12985-021-01643-8.
Collapse
Affiliation(s)
- Mohadeseh Zarei Ghobadi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Rahman Emamzadeh
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Majid Teymoori-Rad
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sayed-Hamidreza Mozhgani
- Department of Microbiology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.,Non‑Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
46
|
Kim MS, Monfredi O, Maltseva LA, Lakatta EG, Maltsev VA. β-Adrenergic Stimulation Synchronizes a Broad Spectrum of Action Potential Firing Rates of Cardiac Pacemaker Cells toward a Higher Population Average. Cells 2021; 10:2124. [PMID: 34440893 PMCID: PMC8391682 DOI: 10.3390/cells10082124] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/10/2021] [Accepted: 08/14/2021] [Indexed: 01/28/2023] Open
Abstract
The heartbeat is initiated by pacemaker cells residing in the sinoatrial node (SAN). SAN cells generate spontaneous action potentials (APs), i.e., normal automaticity. The sympathetic nervous system increases the heart rate commensurate with the cardiac output demand via stimulation of SAN β-adrenergic receptors (βAR). While SAN cells reportedly represent a highly heterogeneous cell population, the current dogma is that, in response to βAR stimulation, all cells increase their spontaneous AP firing rate in a similar fashion. The aim of the present study was to investigate the cell-to-cell variability in the responses of a large population of SAN cells. We measured the βAR responses among 166 single SAN cells isolated from 33 guinea pig hearts. In contrast to the current dogma, the SAN cell responses to βAR stimulation substantially varied. In each cell, changes in the AP cycle length were highly correlated (R2 = 0.97) with the AP cycle length before βAR stimulation. While, as expected, on average, the cells increased their pacemaker rate, greater responses were observed in cells with slower basal rates, and vice versa: cells with higher basal rates showed smaller responses, no responses, or even decreased their rate. Thus, βAR stimulation synchronized the operation of the SAN cell population toward a higher average rate, rather than uniformly shifting the rate in each cell, creating a new paradigm of βAR-driven fight-or-flight responses among individual pacemaker cells.
Collapse
Affiliation(s)
| | | | | | | | - Victor A. Maltsev
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD 21224, USA; (M.S.K.); (O.M.); (L.A.M.); (E.G.L.)
| |
Collapse
|
47
|
Yip D, Accili E. Kinetic modelling of voltage-dependent gating in funny channels. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 166:182-188. [PMID: 34310984 DOI: 10.1016/j.pbiomolbio.2021.07.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 07/12/2021] [Accepted: 07/20/2021] [Indexed: 10/20/2022]
Affiliation(s)
- Delbert Yip
- Department of Cellular and Physiological Sciences, University of British Columbia, Health Sciences Mall, V6T 1Z3, 2350, Canada
| | - Eric Accili
- Department of Cellular and Physiological Sciences, University of British Columbia, Health Sciences Mall, V6T 1Z3, 2350, Canada.
| |
Collapse
|
48
|
Mika D, Fischmeister R. Cyclic nucleotide signaling and pacemaker activity. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 166:29-38. [PMID: 34298001 DOI: 10.1016/j.pbiomolbio.2021.07.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 06/21/2021] [Accepted: 07/13/2021] [Indexed: 01/01/2023]
Abstract
The sinoatrial node (SAN) is the natural pacemaker of the heart, producing the electrical impulse that initiates every heart beat. Its activity is tightly controlled by the autonomic nervous system, and by circulating and locally released factors. Neurohumoral regulation of heart rate plays a crucial role in the integration of vital functions and influences behavior and ability to respond to changing environmental conditions. At the cellular level, modulation of SAN activity occurs through intracellular signaling pathways involving cyclic nucleotides: cyclic AMP (cAMP) and cyclic GMP (cGMP). In this Review, dedicated to Professor Dario DiFrancesco and his accomplishements in the field of cardiac pacemaking, we summarize all findings on the role of cyclic nucleotides signaling in regulating the key actors of cardiac automatism, and we provide an up-to-date review on cAMP- and cGMP-phosphodiesterases (PDEs), compellingly involved in this modulation.
Collapse
Affiliation(s)
- Delphine Mika
- Université Paris-Saclay, Inserm, UMR-S, 1180, Châtenay-Malabry, France.
| | | |
Collapse
|
49
|
Peters CH, Liu PW, Morotti S, Gantz SC, Grandi E, Bean BP, Proenza C. Bidirectional flow of the funny current (I f) during the pacemaking cycle in murine sinoatrial node myocytes. Proc Natl Acad Sci U S A 2021; 118:e2104668118. [PMID: 34260402 PMCID: PMC8285948 DOI: 10.1073/pnas.2104668118] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Sinoatrial node myocytes (SAMs) act as cardiac pacemaker cells by firing spontaneous action potentials (APs) that initiate each heartbeat. The funny current (If) is critical for the generation of these spontaneous APs; however, its precise role during the pacemaking cycle remains unresolved. Here, we used the AP-clamp technique to quantify If during the cardiac cycle in mouse SAMs. We found that If is persistently active throughout the sinoatrial AP, with surprisingly little voltage-dependent gating. As a consequence, it carries both inward and outward current around its reversal potential of -30 mV. Despite operating at only 2 to 5% of its maximal conductance, If carries a substantial fraction of both depolarizing and repolarizing net charge movement during the firing cycle. We also show that β-adrenergic receptor stimulation increases the percentage of net depolarizing charge moved by If, consistent with a contribution of If to the fight-or-flight increase in heart rate. These properties were confirmed by heterologously expressed HCN4 channels and by mathematical models of If Modeling further suggested that the slow rates of activation and deactivation of the HCN4 isoform underlie the persistent activity of If during the sinoatrial AP. These results establish a new conceptual framework for the role of If in pacemaking, in which it operates at a very small fraction of maximal activation but nevertheless drives membrane potential oscillations in SAMs by providing substantial driving force in both inward and outward directions.
Collapse
Affiliation(s)
- Colin H Peters
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Pin W Liu
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115
| | - Stefano Morotti
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Stephanie C Gantz
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Eleonora Grandi
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Bruce P Bean
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115
| | - Catherine Proenza
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045;
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
50
|
Abstract
The physiological heart function is controlled by a well-orchestrated interplay of different ion channels conducting Na+, Ca2+ and K+. Cardiac K+ channels are key players of cardiac repolarization counteracting depolarizating Na+ and Ca2+ currents. In contrast to Na+ and Ca2+, K+ is conducted by many different channels that differ in activation/deactivation kinetics as well as in their contribution to different phases of the action potential. Together with modulatory subunits these K+ channel α-subunits provide a wide range of repolarizing currents with specific characteristics. Moreover, due to expression differences, K+ channels strongly influence the time course of the action potentials in different heart regions. On the other hand, the variety of different K+ channels increase the number of possible disease-causing mutations. Up to now, a plethora of gain- as well as loss-of-function mutations in K+ channel forming or modulating proteins are known that cause severe congenital cardiac diseases like the long-QT-syndrome, the short-QT-syndrome, the Brugada syndrome and/or different types of atrial tachyarrhythmias. In this chapter we provide a comprehensive overview of different K+ channels in cardiac physiology and pathophysiology.
Collapse
|