1
|
Chelu A, Cartwright EJ, Dobrzynski H. Empowering artificial intelligence in characterizing the human primary pacemaker of the heart at single cell resolution. Sci Rep 2024; 14:14041. [PMID: 38890395 PMCID: PMC11189420 DOI: 10.1038/s41598-024-63542-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 05/29/2024] [Indexed: 06/20/2024] Open
Abstract
The sinus node (SN) serves as the primary pacemaker of the heart and is the first component of the cardiac conduction system. Due to its anatomical properties and sample scarcity, the cellular composition of the human SN has been historically challenging to study. Here, we employed a novel deep learning deconvolution method, namely Bulk2space, to characterise the cellular heterogeneity of the human SN using existing single-cell datasets of non-human species. As a proof of principle, we used Bulk2Space to profile the cells of the bulk human right atrium using publicly available mouse scRNA-Seq data as a reference. 18 human cell populations were identified, with cardiac myocytes being the most abundant. Each identified cell population correlated to its published experimental counterpart. Subsequently, we applied the deconvolution to the bulk transcriptome of the human SN and identified 11 cell populations, including a population of pacemaker cardiomyocytes expressing pacemaking ion channels (HCN1, HCN4, CACNA1D) and transcription factors (SHOX2 and TBX3). The connective tissue of the SN was characterised by adipocyte and fibroblast populations, as well as key immune cells. Our work unravelled the unique single cell composition of the human SN by leveraging the power of a novel machine learning method.
Collapse
Affiliation(s)
- Alexandru Chelu
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PL, UK.
| | - Elizabeth J Cartwright
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PL, UK
| | - Halina Dobrzynski
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PL, UK
- Department of Anatomy, Jagiellonian University Medical College, 31-008, Kraków, Poland
| |
Collapse
|
2
|
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
|
3
|
Li H, Tang Q, Yang T, Wang Z, Li D, Wang L, Li L, Chen Y, Huang H, Zhang Y, Chen Y. Segregation of morphogenetic regulatory function of Shox2 from its cell fate guardian role in sinoatrial node development. Commun Biol 2024; 7:385. [PMID: 38553636 PMCID: PMC10980793 DOI: 10.1038/s42003-024-06039-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 03/11/2024] [Indexed: 04/02/2024] Open
Abstract
Shox2 plays a vital role in the morphogenesis and physiological function of the sinoatrial node (SAN), the primary cardiac pacemaker, manifested by the formation of a hypoplastic SAN and failed differentiation of pacemaker cells in Shox2 mutants. Shox2 and Nkx2-5 are co-expressed in the developing SAN and regulate the fate of the pacemaker cells through a Shox2-Nkx2-5 antagonistic mechanism. Here we show that simultaneous inactivation of Nkx2-5 in the SAN of Shox2 mutants (dKO) rescued the pacemaking cell fate but not the hypoplastic defects, indicating uncoupling of SAN cell fate determination and morphogenesis. Single-cell RNA-seq revealed that the presumptive SAN cells of Shox2-/- mutants failed to activate pacemaking program but remained in a progenitor state preceding working myocardium, while both wildtype and dKO SAN cells displayed normal pacemaking cell fate with similar cellular state. Shox2 thus acts as a safeguard but not a determinant to ensure the pacemaking cell fate through the Shox2-Nkx2-5 antagonistic mechanism, which is segregated from its morphogenetic regulatory function in SAN development.
Collapse
Affiliation(s)
- Hua Li
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, Fujian Province, 350122, PR China.
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA.
- Southern Center for Biomedical Research and Fujian Key Laboratory of Developmental and Neural Biology, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian Province, 350108, PR China.
| | - Qinghuang Tang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY, 14214, USA
| | - Tianfang Yang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Zhengsen Wang
- Southern Center for Biomedical Research and Fujian Key Laboratory of Developmental and Neural Biology, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian Province, 350108, PR China
| | - Dainan Li
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Linyan Wang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
- Department of Stomatology, Chengdu Second People's Hospital, Chengdu, Sichuan Province, 610021, PR China
| | - Liwen Li
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
- Department of Biological Sciences, College of Arts and Sciences, University at Buffalo, Buffalo, NY, 14260, USA
| | - Yaoyi Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Hai Huang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Yanding Zhang
- Southern Center for Biomedical Research and Fujian Key Laboratory of Developmental and Neural Biology, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian Province, 350108, PR China
| | - YiPing Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA.
| |
Collapse
|
4
|
Engel JL, Zhang X, Lu DR, Vila OF, Arias V, Lee J, Hale C, Hsu YH, Li CM, Wu RS, Vedantham V, Ang YS. Single Cell Multi-Omics of an iPSC Model of Human Sinoatrial Node Development Reveals Genetic Determinants of Heart Rate and Arrhythmia Susceptibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.01.547335. [PMID: 37425707 PMCID: PMC10327193 DOI: 10.1101/2023.07.01.547335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Cellular heterogeneity within the sinoatrial node (SAN) is functionally important but has been difficult to model in vitro , presenting a major obstacle to studies of heart rate regulation and arrhythmias. Here we describe a scalable method to derive sinoatrial node pacemaker cardiomyocytes (PCs) from human induced pluripotent stem cells that recapitulates differentiation into distinct PC subtypes, including SAN Head, SAN Tail, transitional zone cells, and sinus venosus myocardium. Single cell (sc) RNA-sequencing, sc-ATAC-sequencing, and trajectory analyses were used to define epigenetic and transcriptomic signatures of each cell type, and to identify novel transcriptional pathways important for PC subtype differentiation. Integration of our multi-omics datasets with genome wide association studies uncovered cell type-specific regulatory elements that associated with heart rate regulation and susceptibility to atrial fibrillation. Taken together, these datasets validate a novel, robust, and realistic in vitro platform that will enable deeper mechanistic exploration of human cardiac automaticity and arrhythmia.
Collapse
|
5
|
Zheng H, Peri L, Ward GK, Sanders KM, Ward SM. Cardiac PDGFRα + interstitial cells generate spontaneous inward currents that contribute to excitability in the heart. FASEB J 2023; 37:e22929. [PMID: 37086093 PMCID: PMC10402933 DOI: 10.1096/fj.202201712r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 03/06/2023] [Accepted: 04/05/2023] [Indexed: 04/23/2023]
Abstract
The cell types and conductance that contribute to normal cardiac functions remain under investigation. We used mice that express an enhanced green fluorescent protein (eGFP)-histone 2B fusion protein driven off the cell-specific endogenous promoter for Pdgfra to investigate the distribution and functional role of PDGFRα+ cells in the heart. Cardiac PDGFRα+ cells were widely distributed within the endomysium of atria, ventricle, and sino-atrial node (SAN) tissues. PDGFRα+ cells formed a discrete network of cells, lying in close apposition to neighboring cardiac myocytes in mouse and Cynomolgus monkey (Macaca fascicularis) hearts. Expression of eGFP in nuclei allowed unequivocal identification of these cells following enzymatic dispersion of muscle tissues. FACS purification of PDGFRα+ cells from the SAN and analysis of gene transcripts by qPCR revealed that they were a distinct population of cells that expressed gap junction transcripts, Gja1 and Gjc1. Cardiac PDGFRα+ cells generated spontaneous transient inward currents (STICs) and spontaneous transient depolarizations (STDs) that reversed at 0 mV. Reversal potential was maintained when ECl = -40 mV. [Na+ ]o replacement and FTY720 abolished STICs, suggesting they were due to a non-selective cation conductance (NSCC) carried by TRPM7. PDGFRα+ cells also express β2 -adrenoceptor gene transcripts, Adrb2. Zinterol, a selective β2 -receptor agonist, increased the amplitude and frequency of STICs, suggesting these cells could contribute to adrenergic regulation of cardiac excitability. PDGFRα+ cells in cardiac muscles generate inward currents via an NSCC. STICs generated by these cells may contribute to the integrated membrane potentials of cardiac muscles, possibly affecting the frequency of pacemaker activity.
Collapse
Affiliation(s)
- Haifeng Zheng
- Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - Lauren Peri
- Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - Grace K. Ward
- Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - Kenton M. Sanders
- Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - Sean M. Ward
- Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| |
Collapse
|
6
|
Ock S, Choi SW, Choi SH, Kang H, Kim SJ, Lee WS, Kim J. Insulin signaling is critical for sinoatrial node maintenance and function. Exp Mol Med 2023; 55:965-973. [PMID: 37121973 PMCID: PMC10238478 DOI: 10.1038/s12276-023-00988-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 02/12/2023] [Accepted: 02/12/2023] [Indexed: 05/02/2023] Open
Abstract
Insulin and insulin-like growth factor 1 (IGF-1) signaling regulate cellular growth and glucose metabolism in the myocardium. However, their physiological role in the cells of the cardiac conduction system has never been explored. Therefore, we sought to determine the spatiotemporal function of insulin/IGF-1 receptors in the sinoatrial node (SAN). We generated cardiac conduction cell-specific inducible IGF-1 receptor (IGF-1R) knockout (KO) (CSIGF1RKO), insulin receptor (IR) KO (CSIRKO), and IR/IGF-1R double-KO (CSDIRKO) mice and evaluated their phenotypes. Telemetric electrocardiography revealed regular sinus rhythm in CSIGF1RKO mice, indicating that IGF-1R is dispensable for normal pacemaking. In contrast, CSIRKO and CSDIRKO mice exhibited profound sinus bradycardia. CSDIRKO mice showed typical sinus node dysfunction characterized by junctional rhythm and sinus pauses on electrocardiography. Interestingly, the lack of an insulin receptor in the SAN cells of CSIRKO and CSDIRKO mice caused sinus nodal fibrosis. Mechanistically, hyperpolarization-activated cyclic nucleotide-gated channel 4 (HCN4) protein expression significantly decreased in the CSIRKO and CSDIRKO mice relative to the controls. A patch-clamp study of the SAN cells of CSIRKO mice revealed a significant decrease in the funny current, which is responsible for spontaneous diastolic depolarization in the SAN. This result suggested that insulin receptor loss reduces the heart rate via downregulation of the HCN4 channel. Additionally, HCN1 expression was decreased in CSDIRKO mice, explaining their sinus node dysfunction. Our results reveal a previously unrecognized role of insulin/IGF-1 signaling in sinus node structural maintenance and pacemaker function.
Collapse
Affiliation(s)
- Sangmi Ock
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Seong Woo Choi
- Departments of Physiology and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Korea
- Department of Physiology, Dongguk University College of Medicine, Gyeongju, Korea
| | - Seung Hee Choi
- Division of Endocrinology and Metabolism, Departments of Internal Medicine and Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Daegu, Korea
| | - Hyun Kang
- Department of Anesthesiology, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Sung Joon Kim
- Departments of Physiology and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Korea
| | - Wang-Soo Lee
- Division of Cardiology, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Korea.
| | - Jaetaek Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Korea.
| |
Collapse
|
7
|
Arbel Ganon L, Davoodi M, Alexandrovich A, Yaniv Y. Synergy between Membrane Currents Prevents Severe Bradycardia in Mouse Sinoatrial Node Tissue. Int J Mol Sci 2023; 24:ijms24065786. [PMID: 36982861 PMCID: PMC10051777 DOI: 10.3390/ijms24065786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/04/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
Bradycardia is initiated by the sinoatrial node (SAN), which is regulated by a coupled-clock system. Due to the clock coupling, reduction in the 'funny' current (If), which affects SAN automaticity, can be compensated, thus preventing severe bradycardia. We hypothesize that this fail-safe system is an inherent feature of SAN pacemaker cells and is driven by synergy between If and other ion channels. This work aimed to characterize the connection between membrane currents and their underlying mechanisms in SAN cells. SAN tissues were isolated from C57BL mice and Ca2+ signaling was measured in pacemaker cells within them. A computational model of SAN cells was used to understand the interactions between cell components. Beat interval (BI) was prolonged by 54 ± 18% (N = 16) and 30 ± 9% (N = 21) in response to If blockade, by ivabradine, or sodium current (INa) blockade, by tetrodotoxin, respectively. Combined drug application had a synergistic effect, manifested by a BI prolonged by 143 ± 25% (N = 18). A prolongation in the local Ca2+ release period, which reports on the level of crosstalk within the coupled-clock system, was measured and correlated with the prolongation in BI. The computational model predicted that INa increases in response to If blockade and that this connection is mediated by changes in T and L-type Ca2+ channels.
Collapse
Affiliation(s)
- Limor Arbel Ganon
- Laboratory of Bioelectric and Bioenergetic Systems, Faculty of Biomedical Engineering, Technion-IIT, Haifa 3200003, Israel
| | - Moran Davoodi
- Laboratory of Bioelectric and Bioenergetic Systems, Faculty of Biomedical Engineering, Technion-IIT, Haifa 3200003, Israel
| | - Alexandra Alexandrovich
- Laboratory of Bioelectric and Bioenergetic Systems, Faculty of Biomedical Engineering, Technion-IIT, Haifa 3200003, Israel
| | - Yael Yaniv
- Laboratory of Bioelectric and Bioenergetic Systems, Faculty of Biomedical Engineering, Technion-IIT, Haifa 3200003, Israel
| |
Collapse
|
8
|
Manoj P, Kim JA, Kim S, Li T, Sewani M, Chelu MG, Li N. Sinus node dysfunction: current understanding and future directions. Am J Physiol Heart Circ Physiol 2023; 324:H259-H278. [PMID: 36563014 PMCID: PMC9886352 DOI: 10.1152/ajpheart.00618.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
The sinoatrial node (SAN) is the primary pacemaker of the heart. Normal SAN function is crucial in maintaining proper cardiac rhythm and contraction. Sinus node dysfunction (SND) is due to abnormalities within the SAN, which can affect the heartbeat frequency, regularity, and the propagation of electrical pulses through the cardiac conduction system. As a result, SND often increases the risk of cardiac arrhythmias. SND is most commonly seen as a disease of the elderly given the role of degenerative fibrosis as well as other age-dependent changes in its pathogenesis. Despite the prevalence of SND, current treatment is limited to pacemaker implantation, which is associated with substantial medical costs and complications. Emerging evidence has identified various genetic abnormalities that can cause SND, shedding light on the molecular underpinnings of SND. Identification of these molecular mechanisms and pathways implicated in the pathogenesis of SND is hoped to identify novel therapeutic targets for the development of more effective therapies for this disease. In this review article, we examine the anatomy of the SAN and the pathophysiology and epidemiology of SND. We then discuss in detail the most common genetic mutations correlated with SND and provide our perspectives on future research and therapeutic opportunities in this field.
Collapse
Affiliation(s)
- Pavan Manoj
- School of Public Health, Texas A&M University, College Station, Texas
| | - Jitae A Kim
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Stephanie Kim
- Department of BioSciences, Rice University, Houston, Texas
| | - Tingting Li
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Maham Sewani
- Department of BioSciences, Rice University, Houston, Texas
| | - Mihail G Chelu
- Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Na Li
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
9
|
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
|
10
|
Ding Y, Lang D, Yan J, Bu H, Li H, Jiao K, Yang J, Ni H, Morotti S, Le T, Clark KJ, Port J, Ekker SC, Cao H, Zhang Y, Wang J, Grandi E, Li Z, Shi Y, Li Y, Glukhov AV, Xu X. A phenotype-based forward genetic screen identifies Dnajb6 as a sick sinus syndrome gene. eLife 2022; 11:e77327. [PMID: 36255053 PMCID: PMC9642998 DOI: 10.7554/elife.77327] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 10/17/2022] [Indexed: 11/13/2022] Open
Abstract
Previously we showed the generation of a protein trap library made with the gene-break transposon (GBT) in zebrafish (Danio rerio) that could be used to facilitate novel functional genome annotation towards understanding molecular underpinnings of human diseases (Ichino et al, 2020). Here, we report a significant application of this library for discovering essential genes for heart rhythm disorders such as sick sinus syndrome (SSS). SSS is a group of heart rhythm disorders caused by malfunction of the sinus node, the heart's primary pacemaker. Partially owing to its aging-associated phenotypic manifestation and low expressivity, molecular mechanisms of SSS remain difficult to decipher. From 609 GBT lines screened, we generated a collection of 35 zebrafish insertional cardiac (ZIC) mutants in which each mutant traps a gene with cardiac expression. We further employed electrocardiographic measurements to screen these 35 ZIC lines and identified three GBT mutants with SSS-like phenotypes. More detailed functional studies on one of the arrhythmogenic mutants, GBT411, in both zebrafish and mouse models unveiled Dnajb6 as a novel SSS causative gene with a unique expression pattern within the subpopulation of sinus node pacemaker cells that partially overlaps with the expression of hyperpolarization activated cyclic nucleotide gated channel 4 (HCN4), supporting heterogeneity of the cardiac pacemaker cells.
Collapse
Affiliation(s)
- Yonghe Ding
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo ClinicRochesterUnited States
- The Affiliated Hospital of Qingdao University & The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao UniversityQingdaoChina
| | - Di Lang
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-MadisonMadisonUnited States
- Department of Medicine, University of California, San FranciscoSan FranciscoUnited States
| | - Jianhua Yan
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo ClinicRochesterUnited States
- Division of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School Of MedicineShanghaiChina
| | - Haisong Bu
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo ClinicRochesterUnited States
- Department of Cardiothoracic Surgery, Xiangya Hospital, Central South UniversityChangshaChina
| | - Hongsong Li
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo ClinicRochesterUnited States
- Department of Cardiovascular Medicine, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health ScienceShanghaiChina
| | - Kunli Jiao
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo ClinicRochesterUnited States
- Division of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School Of MedicineShanghaiChina
| | - Jingchun Yang
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo ClinicRochesterUnited States
| | - Haibo Ni
- Department of Pharmacology, University of California, DavisDavisUnited States
| | - Stefano Morotti
- Department of Pharmacology, University of California, DavisDavisUnited States
| | - Tai Le
- Department of Biomedical Engineering, University of California, IrvineIrvineUnited States
| | - Karl J Clark
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo ClinicRochesterUnited States
| | - Jenna Port
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-MadisonMadisonUnited States
| | - Stephen C Ekker
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo ClinicRochesterUnited States
| | - Hung Cao
- Department of Biomedical Engineering, University of California, IrvineIrvineUnited States
- Department of Electrical Engineering and Computer Science, University of California, IrvineIrvineUnited States
| | - Yuji Zhang
- Department of Epidemiology and Public Health, University of Maryland School of MedicineBaltimoreUnited States
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at HoustonHoustonUnited States
| | - Eleonora Grandi
- Department of Pharmacology, University of California, DavisDavisUnited States
| | - Zhiqiang Li
- The Affiliated Hospital of Qingdao University & The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao UniversityQingdaoChina
| | - Yongyong Shi
- The Affiliated Hospital of Qingdao University & The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao UniversityQingdaoChina
| | - Yigang Li
- Division of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School Of MedicineShanghaiChina
| | - Alexey V Glukhov
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-MadisonMadisonUnited States
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo ClinicRochesterUnited States
| |
Collapse
|
11
|
Wang H, Wu T, Huang Z, Huang J, Geng Z, Cui B, Yan Y, Zhang Y, Wang Y. Channel HCN4 mutation R666Q associated with sporadic arrhythmia decreases channel electrophysiological function and increases protein degradation. J Biol Chem 2022; 298:102599. [PMID: 36244448 PMCID: PMC9663530 DOI: 10.1016/j.jbc.2022.102599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 10/06/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022] Open
Abstract
Mutations in the hyperpolarization-activated nucleotide-gated channel 4 (HCN4) are known to be associated with arrhythmias in which QT prolongation (delayed ventricular repolarization) is rare. Here, we identified a HCN4 mutation, HCN4-R666Q, in two sporadic arrhythmia patients with sinus bradycardia, QT prolongation, and short bursts of ventricular tachycardia. To determine the functional effect of the mutation, we conducted clinical, genetic, and functional analyses using whole-cell voltage-clamp, qPCR, Western blot, confocal microscopy, and co-immunoprecipitation. The mean current density of HEK293T cells transfected with HCN4-R666Q was lower in 24 to 36 h after transfection and was much lower in 36 to 48 h after transfection relative to cells transfected with wildtype HCN4. Additionally, we determined that the HCN4-R666Q mutant was more susceptible to ubiquitin-proteasome system–mediated protein degradation than wildtype HCN4. This decreased current density for HCN4-R666Q could be partly rescued by treatment with a proteasome inhibitor. Therefore, we conclude that HCN4-R666Q had an effect on HCN4 function in two aspects, including decreasing the current density of the channel as a biophysical effect and weakening its protein stability. Our findings provide new insights into the pathogenesis of the HCN4-R666Q mutation.
Collapse
|
12
|
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: 10] [Impact Index Per Article: 5.0] [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
|
13
|
Electro-anatomical computational cardiology in humans and experimental animal models. TRANSLATIONAL RESEARCH IN ANATOMY 2022. [DOI: 10.1016/j.tria.2022.100162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
14
|
A comparative review on heart ion channels, action potentials and electrocardiogram in rodents and human: extrapolation of experimental insights to clinic. Lab Anim Res 2021; 37:25. [PMID: 34496976 PMCID: PMC8424989 DOI: 10.1186/s42826-021-00102-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/26/2021] [Indexed: 11/10/2022] Open
Abstract
Electrocardiogram (ECG) is a non-invasive valuable diagnostic tool that is used in clinics for investigation and monitoring of heart electrical rhythm/conduction, ischemia/injury of heart, electrolyte disturbances and agents/drugs induced cardiac toxicity. Nowadays using animal models to study heart diseases such as electrical and mechanical disturbance is common. In addition, given to ethical consideration and availability, the use of small rodents has been a top priority for cardiovascular researchers. However, extrapolation of experimental findings from the lab to the clinic needs sufficient basic knowledge of similarities and differences between heart action potential and ECG of rodents and humans in normal and disease conditions. This review compares types of human action potentials, the dominant ion currents during action potential phases, alteration in ion channels activities in channelopathies-induced arrhythmias and the ECG appearance of mouse, rat, guinea pig, rabbit and human. Also, it briefly discusses the responsiveness and alterations in ECG following some interventions such as cardiac injury and arrhythmia induction. Overall, it provides a roadmap for researchers in selecting the best animal model/species whose studies results can be translated into clinical practice. In addition, this study will also be useful to biologists, physiologists, pharmacologists, veterinarians and physicians working in the fields of comparative physiology, pharmacology, toxicology and diseases.
Collapse
|
15
|
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: 1.0] [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
|
16
|
Naumova N, Iop L. Bioengineering the Cardiac Conduction System: Advances in Cellular, Gene, and Tissue Engineering for Heart Rhythm Regeneration. Front Bioeng Biotechnol 2021; 9:673477. [PMID: 34409019 PMCID: PMC8365186 DOI: 10.3389/fbioe.2021.673477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 06/24/2021] [Indexed: 01/01/2023] Open
Abstract
Heart rhythm disturbances caused by different etiologies may affect pediatric and adult patients with life-threatening consequences. When pharmacological therapy is ineffective in treating the disturbances, the implantation of electronic devices to control and/or restore normal heart pacing is a unique clinical management option. Although these artificial devices are life-saving, they display many limitations; not least, they do not have any capability to adapt to somatic growth or respond to neuroautonomic physiological changes. A biological pacemaker could offer a new clinical solution for restoring heart rhythms in the conditions of disorder in the cardiac conduction system. Several experimental approaches, such as cell-based, gene-based approaches, and the combination of both, for the generation of biological pacemakers are currently established and widely studied. Pacemaker bioengineering is also emerging as a technology to regenerate nodal tissues. This review analyzes and summarizes the strategies applied so far for the development of biological pacemakers, and discusses current translational challenges toward the first-in-human clinical application.
Collapse
Affiliation(s)
| | - Laura Iop
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padua, Padua, Italy
| |
Collapse
|
17
|
Zheng X, Wang F, Hu X, Li H, Guan Z, Zhang Y, Hu X. PDGFRα-Signaling Is Dispensable for the Development of the Sinoatrial Node After Its Fate Commitment. Front Cell Dev Biol 2021; 9:647165. [PMID: 34178981 PMCID: PMC8222823 DOI: 10.3389/fcell.2021.647165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 05/18/2021] [Indexed: 11/13/2022] Open
Abstract
Palate-derived growth factor receptor α (Pdgfrα) signaling has been reported to play important roles in the cardiac development. A previous study utilizing Pdgfrα conventional knockout mice reported hypoplasia of the sinus venous myocardium including the sinoatrial node (SAN) accompanied by increased expression of Nkx2.5. This mouse line embryos die by E11.5 due to embryonic lethality, rendering them difficult to investigate the details. To elucidate the underlying mechanism, in this study, we revisited this observation by generation of specific ablation of Pdgfrα in the SAN by Shox2-Cre at E9.5, using a Shox2-Cre;Pdgfrα flox/flox conditional mouse line. Surprisingly, we found that resultant homozygous mutant mice did not exhibit any malformation in SAN morphology as compared to their wild-type littermates. Further analysis revealed the normal cardiac function in adult mutant mice assessed by the record of heart rate and electrocardiogram and unaltered expression of Nkx2.5 in the E13.5 SAN of Pdgfrα conditional knockout mice. Our results unambiguously demonstrate that Pdgfrα is dispensable for SAN development after its fate commitment in mice.
Collapse
Affiliation(s)
- Xi Zheng
- Fujian Key Laboratory of Developmental and Neural Biology & Southern Center for Biomedical Research, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Fengjiao Wang
- Fujian Key Laboratory of Developmental and Neural Biology & Southern Center for Biomedical Research, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Xiaoxiao Hu
- Fujian Key Laboratory of Developmental and Neural Biology & Southern Center for Biomedical Research, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Hua Li
- Key Laboratory of Stem Cell Engineering Societ and Regenerative Medicine, School of Basic Medical Science, Fujian Medical University, Fuzhou, China
| | - Zhen Guan
- Fujian Key Laboratory of Developmental and Neural Biology & Southern Center for Biomedical Research, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Yanding Zhang
- Fujian Key Laboratory of Developmental and Neural Biology & Southern Center for Biomedical Research, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Xuefeng Hu
- Fujian Key Laboratory of Developmental and Neural Biology & Southern Center for Biomedical Research, College of Life Sciences, Fujian Normal University, Fuzhou, China
| |
Collapse
|
18
|
Saadeh K, Chadda KR, Ahmad S, Valli H, Nanthakumar N, Fazmin IT, Edling CE, Huang CLH, Jeevaratnam K. Molecular basis of ventricular arrhythmogenicity in a Pgc-1α deficient murine model. Mol Genet Metab Rep 2021; 27:100753. [PMID: 33898262 PMCID: PMC8059080 DOI: 10.1016/j.ymgmr.2021.100753] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 04/03/2021] [Indexed: 11/17/2022] Open
Abstract
Mitochondrial dysfunction underlying metabolic disorders such as obesity and diabetes mellitus is strongly associated with cardiac arrhythmias. Murine Pgc-1α-/- hearts replicate disrupted mitochondrial function and model the associated pro-arrhythmic electrophysiological abnormalities. Quantitative PCR, western blotting and histological analysis were used to investigate the molecular basis of the electrophysiological changes associated with mitochondrial dysfunction. qPCR analysis implicated downregulation of genes related to Na+-K+ ATPase activity (Atp1b1), surface Ca2+ entry (Cacna1c), action potential repolarisation (Kcnn1), autonomic function (Adra1d, Adcy4, Pde4d, Prkar2a), and morphological properties (Myh6, Tbx3) in murine Pgc-1α-/- ventricles. Western blotting revealed reduced NaV1.5 but normal Cx43 expression. Histological analysis revealed increased tissue fibrosis in the Pgc-1α-/- ventricles. These present findings identify altered transcription amongst a strategically selected set of genes established as encoding proteins involved in cardiac electrophysiological activation and therefore potentially involved in alterations in ventricular activation and Ca2+ homeostasis in arrhythmic substrate associated with Pgc-1α deficiency. They complement and complete previous studies examining such expression characteristics in the atria and ventricles of Pgc-1 deficient murine hearts.
Collapse
Affiliation(s)
- Khalil Saadeh
- Faculty of Health and Medical Sciences, University of Surrey, GU2 7AL Guildford, United Kingdom
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Karan R. Chadda
- Faculty of Health and Medical Sciences, University of Surrey, GU2 7AL Guildford, United Kingdom
| | - Shiraz Ahmad
- Faculty of Health and Medical Sciences, University of Surrey, GU2 7AL Guildford, United Kingdom
- Physiological Laboratory and Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Haseeb Valli
- Physiological Laboratory and Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Nakulan Nanthakumar
- Faculty of Health and Medical Sciences, University of Surrey, GU2 7AL Guildford, United Kingdom
- Bristol Medical School. University of Bristol, Bristol, United Kingdom
| | - Ibrahim T. Fazmin
- Faculty of Health and Medical Sciences, University of Surrey, GU2 7AL Guildford, United Kingdom
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Charlotte E. Edling
- Faculty of Health and Medical Sciences, University of Surrey, GU2 7AL Guildford, United Kingdom
| | - Christopher L.-H. Huang
- Faculty of Health and Medical Sciences, University of Surrey, GU2 7AL Guildford, United Kingdom
- Physiological Laboratory and Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Kamalan Jeevaratnam
- Faculty of Health and Medical Sciences, University of Surrey, GU2 7AL Guildford, United Kingdom
| |
Collapse
|
19
|
Hennis K, Rötzer RD, Piantoni C, Biel M, Wahl-Schott C, Fenske S. Speeding Up the Heart? Traditional and New Perspectives on HCN4 Function. Front Physiol 2021; 12:669029. [PMID: 34122140 PMCID: PMC8191466 DOI: 10.3389/fphys.2021.669029] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/19/2021] [Indexed: 01/20/2023] Open
Abstract
The sinoatrial node (SAN) is the primary pacemaker of the heart and is responsible for generating the intrinsic heartbeat. Within the SAN, spontaneously active pacemaker cells initiate the electrical activity that causes the contraction of all cardiomyocytes. The firing rate of pacemaker cells depends on the slow diastolic depolarization (SDD) and determines the intrinsic heart rate (HR). To adapt cardiac output to varying physical demands, HR is regulated by the autonomic nervous system (ANS). The sympathetic and parasympathetic branches of the ANS innervate the SAN and regulate the firing rate of pacemaker cells by accelerating or decelerating SDD-a process well-known as the chronotropic effect. Although this process is of fundamental physiological relevance, it is still incompletely understood how it is mediated at the subcellular level. Over the past 20 years, most of the work to resolve the underlying cellular mechanisms has made use of genetically engineered mouse models. In this review, we focus on the findings from these mouse studies regarding the cellular mechanisms involved in the generation and regulation of the heartbeat, with particular focus on the highly debated role of the hyperpolarization-activated cyclic nucleotide-gated cation channel HCN4 in mediating the chronotropic effect. By focusing on experimental data obtained in mice and humans, but not in other species, we outline how findings obtained in mice relate to human physiology and pathophysiology and provide specific information on how dysfunction or loss of HCN4 channels leads to human SAN disease.
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
| | - Chiara Piantoni
- Institute for Neurophysiology, Hannover Medical School, Hanover, 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
| | - Christian Wahl-Schott
- Institute for Neurophysiology, Hannover Medical School, Hanover, 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
|
20
|
Lang D, Glukhov AV. Cellular and Molecular Mechanisms of Functional Hierarchy of Pacemaker Clusters in the Sinoatrial Node: New Insights into Sick Sinus Syndrome. J Cardiovasc Dev Dis 2021; 8:jcdd8040043. [PMID: 33924321 PMCID: PMC8069964 DOI: 10.3390/jcdd8040043] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 12/17/2022] Open
Abstract
The sinoatrial node (SAN), the primary pacemaker of the heart, consists of a heterogeneous population of specialized cardiac myocytes that can spontaneously produce action potentials, generating the rhythm of the heart and coordinating heart contractions. Spontaneous beating can be observed from very early embryonic stage and under a series of genetic programing, the complex heterogeneous SAN cells are formed with specific biomarker proteins and generate robust automaticity. The SAN is capable to adjust its pacemaking rate in response to environmental and autonomic changes to regulate the heart's performance and maintain physiological needs of the body. Importantly, the origin of the action potential in the SAN is not static, but rather dynamically changes according to the prevailing conditions. Changes in the heart rate are associated with a shift of the leading pacemaker location within the SAN and accompanied by alterations in P wave morphology and PQ interval on ECG. Pacemaker shift occurs in response to different interventions: neurohormonal modulation, cardiac glycosides, pharmacological agents, mechanical stretch, a change in temperature, and a change in extracellular electrolyte concentrations. It was linked with the presence of distinct anatomically and functionally defined intranodal pacemaker clusters that are responsible for the generation of the heart rhythm at different rates. Recent studies indicate that on the cellular level, different pacemaker clusters rely on a complex interplay between the calcium (referred to local subsarcolemmal Ca2+ releases generated by the sarcoplasmic reticulum via ryanodine receptors) and voltage (referred to sarcolemmal electrogenic proteins) components of so-called "coupled clock pacemaker system" that is used to describe a complex mechanism of SAN pacemaking. In this review, we examine the structural, functional, and molecular evidence for hierarchical pacemaker clustering within the SAN. We also demonstrate the unique molecular signatures of intranodal pacemaker clusters, highlighting their importance for physiological rhythm regulation as well as their role in the development of SAN dysfunction, also known as sick sinus syndrome.
Collapse
|
21
|
Hennis K, Biel M, Wahl-Schott C, Fenske S. Beyond pacemaking: HCN channels in sinoatrial node function. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 166:51-60. [PMID: 33753086 DOI: 10.1016/j.pbiomolbio.2021.03.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/04/2021] [Accepted: 03/16/2021] [Indexed: 01/16/2023]
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are key proteins involved in the initiation and regulation of the heartbeat. Pacemaker cells within the sinoatrial node generate the electrical impulse that underlies the contraction of all atrial and ventricular cardiomyocytes. To generate a stable heart rhythm, it is necessary that the spontaneous activity of pacemaker cells is synchronized. Entrainment processes in the sinoatrial node create synchrony and also mediate heart rate regulation. In the past years it has become clear that the role of HCN channels goes beyond just pacemaking and that the channels play pivotal roles in these entrainment processes that coordinate and balance sinoatrial node network activity. Here, we review the role of HCN channels in the central pacemaker process and highlight new aspects of the contribution of HCN channels to stabilizing the electrical activity of the sinoatrial node network, especially during heart rate regulation by the autonomic nervous system.
Collapse
Affiliation(s)
- Konstantin Hennis
- Center for Drug Research, Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377, Munich, Germany
| | - Martin Biel
- Center for Drug Research, Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377, Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80802, Munich, Germany
| | - Christian Wahl-Schott
- Hannover Medical School, Institute for Neurophysiology, 30625, Hannover, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80802, Munich, Germany.
| | - Stefanie Fenske
- Center for Drug Research, Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377, Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80802, Munich, Germany.
| |
Collapse
|
22
|
RNAseq shows an all-pervasive day-night rhythm in the transcriptome of the pacemaker of the heart. Sci Rep 2021; 11:3565. [PMID: 33574422 PMCID: PMC7878777 DOI: 10.1038/s41598-021-82202-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 01/01/2021] [Indexed: 12/12/2022] Open
Abstract
Physiological systems vary in a day-night manner anticipating increased demand at a particular time. Heart is no exception. Cardiac output is primarily determined by heart rate and unsurprisingly this varies in a day-night manner and is higher during the day in the human (anticipating increased day-time demand). Although this is attributed to a day-night rhythm in post-translational ion channel regulation in the heart's pacemaker, the sinus node, by the autonomic nervous system, we investigated whether there is a day-night rhythm in transcription. RNAseq revealed that ~ 44% of the sinus node transcriptome (7134 of 16,387 transcripts) has a significant day-night rhythm. The data revealed the oscillating components of an intrinsic circadian clock. Presumably this clock (or perhaps the master circadian clock in the suprachiasmatic nucleus) is responsible for the rhythm observed in the transcriptional machinery, which in turn is responsible for the rhythm observed in the transcriptome. For example, there is a rhythm in transcripts responsible for the two principal pacemaker mechanisms (membrane and Ca2+ clocks), transcripts responsible for receptors and signalling pathways known to control pacemaking, transcripts from genes identified by GWAS as determinants of resting heart rate, and transcripts from genes responsible for familial and acquired sick sinus syndrome.
Collapse
|
23
|
MacDonald EA, Madl J, Greiner J, Ramadan AF, Wells SM, Torrente AG, Kohl P, Rog-Zielinska EA, Quinn TA. Sinoatrial Node Structure, Mechanics, Electrophysiology and the Chronotropic Response to Stretch in Rabbit and Mouse. Front Physiol 2020; 11:809. [PMID: 32774307 PMCID: PMC7388775 DOI: 10.3389/fphys.2020.00809] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 06/18/2020] [Indexed: 12/12/2022] Open
Abstract
The rhythmic electrical activity of the heart's natural pacemaker, the sinoatrial node (SAN), determines cardiac beating rate (BR). SAN electrical activity is tightly controlled by multiple factors, including tissue stretch, which may contribute to adaptation of BR to changes in venous return. In most animals, including human, there is a robust increase in BR when the SAN is stretched. However, the chronotropic response to sustained stretch differs in mouse SAN, where it causes variable responses, including decreased BR. The reasons for this species difference are unclear. They are thought to relate to dissimilarities in SAN electrophysiology (particularly action potential morphology) between mouse and other species and to how these interact with subcellular stretch-activated mechanisms. Furthermore, species-related differences in structural and mechanical properties of the SAN may influence the chronotropic response to SAN stretch. Here we assess (i) how the BR response to sustained stretch of rabbit and mouse isolated SAN relates to tissue stiffness, (ii) whether structural differences could account for observed differences in BR responsiveness to stretch, and (iii) whether pharmacological modification of mouse SAN electrophysiology alters stretch-induced chronotropy. We found disparities in the relationship between SAN stiffness and the magnitude of the chronotropic response to stretch between rabbit and mouse along with differences in SAN collagen structure, alignment, and changes with stretch. We further observed that pharmacological modification to prolong mouse SAN action potential plateau duration rectified the direction of BR changes during sustained stretch, resulting in a positive chronotropic response akin to that of other species. Overall, our results suggest that structural, mechanical, and background electrophysiological properties of the SAN influence the chronotropic response to stretch. Improved insight into the biophysical determinants of stretch effects on SAN pacemaking is essential for a comprehensive understanding of SAN regulation with important implications for studies of SAN physiology and its dysfunction, such as in the aging and fibrotic heart.
Collapse
Affiliation(s)
- Eilidh A MacDonald
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada
| | - Josef Madl
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg - Bad Krozingen, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Joachim Greiner
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg - Bad Krozingen, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ahmed F Ramadan
- Department of Electrical and Computer Engineering, Dalhousie University, Halifax, NS, Canada
| | - Sarah M Wells
- School of Biomedical Engineering, Dalhousie University, Halifax, NS, Canada
| | - Angelo G Torrente
- Department of Physiology, Institut de Génomique Fonctionnelle, Montpellier, France
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg - Bad Krozingen, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Eva A Rog-Zielinska
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg - Bad Krozingen, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - T Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada.,Department of Electrical and Computer Engineering, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
24
|
Wang F, Zhao H, Yin L, Tang Y, Wang X, Zhao Q, Wang T, Huang C. Transcription Factor TBX18 Reprograms Vascular Smooth Muscle Cells of Ascending Aorta to Pacemaker-Like Cells. DNA Cell Biol 2019; 38:1470-1479. [PMID: 31633376 DOI: 10.1089/dna.2019.4940] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) of ascending aorta and TBX18+ sinus node both originated from the second heart field. The study explored whether ascending aortic smooth muscle cells in vitro could be reprogrammed into pacemaker-like cells with human TBX18. In the study, VSMCs were infected with TBX18, and then cocultured with neonatal rat ventricular cardiomyocytes (NRVMs) in vitro. By overexpressing TBX18, the transfected VSMCs expressed high levels of hyperpolarization-activated cyclic nucleotide-gated channel 4 (HCN4), insulin gene enhancer binding protein 1, and human dwarf homeobox gene SHOX2, cardiac troponin I, and low level of connexin 43. In addition, funny current (If) was recorded by patch clamp appeared the time and voltage dependence in TBX18 group, which the amplitude of If density was from -5.164 ± 0.662 pA/pF to -0.765 ± 0.358 pA/pF (n = 14). Furthermore, TBX18-transfected VSMCs coupled with NRVMs showed typical action potential of pacemaker-like cells and the beating rate was faster (178.00 ± 7.55 bpm, p < 0.05) compared with other groups. In conclusion, our study indicated that transcription factor TBX18 could reprogram VSMCs into pacemaker-like cells in vitro.
Collapse
Affiliation(s)
- Fengyuan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Hongyi Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Lin Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Qingyan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Teng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| |
Collapse
|
25
|
Dong R, Mu-u-min R, Reith AJM, O’Shea C, He S, Duan K, Kou K, Grassam-Rowe A, Tan X, Pavlovic D, Ou X, Lei M. A Protocol for Dual Calcium-Voltage Optical Mapping in Murine Sinoatrial Preparation With Optogenetic Pacing. Front Physiol 2019; 10:954. [PMID: 31456689 PMCID: PMC6698704 DOI: 10.3389/fphys.2019.00954] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 07/09/2019] [Indexed: 01/08/2023] Open
Abstract
Among the animal models for studying the molecular basis of atrial and sinoatrial node (SAN) biology and disease, the mouse is a widely used species due to its feasibility for genetic modifications in genes encoding ion channels or calcium handling and signaling proteins in the heart. It is therefore highly valuable to develop robust methodologies for studying SAN and atrial electrophysiological function in this species. Here, we describe a protocol for performing dual calcium-voltage optical mapping on mouse sinoatrial preparation (SAP), in combination with an optogenetic approach, for studying SAP membrane potential, intracellular Ca2+ transients, and pacemaker activity. The protocol includes the details for preparing the intact SAP, robust tissue dual-dye loading, light-programmed pacing, and high-resolution optical mapping. Our protocol provides an example of use of the combination of optogenetic and optical mapping techniques for investigating SAP membrane potential and intracellular Ca2+ transients and pacemaker activity with high temporal and spatial resolution in specific cardiac tissues. Thus, our protocol provides a useful tool for studying SAP physiology and pathophysiology in mice.
Collapse
Affiliation(s)
- Ruirui Dong
- 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, China
| | - Razik Mu-u-min
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | | | - Christopher O’Shea
- Institute for Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Shicheng He
- 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, China
| | - Kaizhong Duan
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Kun Kou
- 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, China
| | | | - Xiaoqiu Tan
- 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, China
| | - Davor Pavlovic
- Institute for Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Xianhong Ou
- 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, China
| | - Ming Lei
- 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, China
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
26
|
Li H, Li D, Wang Y, Huang Z, Xu J, Yang T, Wang L, Tang Q, Cai CL, Huang H, Zhang Y, Chen Y. Nkx2-5 defines a subpopulation of pacemaker cells and is essential for the physiological function of the sinoatrial node in mice. Development 2019; 146:dev.178145. [PMID: 31320323 DOI: 10.1242/dev.178145] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 07/04/2019] [Indexed: 02/05/2023]
Abstract
The sinoatrial node (SAN), the primary cardiac pacemaker, consists of a head domain and a junction/tail domain that exhibit different functional properties. However, the underlying molecular mechanism defining these two pacemaker domains remains elusive. Nkx2-5 is a key transcription factor essential for the formation of the working myocardium, but it was generally thought to be detrimental to SAN development. However, Nkx2-5 is expressed in the developing SAN junction, suggesting a role for Nkx2-5 in SAN junction development and function. In this study, we present unambiguous evidence that SAN junction cells exhibit unique action potential configurations intermediate to those manifested by the SAN head and the surrounding atrial cells, suggesting a specific role for the junction cells in impulse generation and in SAN-atrial exit conduction. Single-cell RNA-seq analyses support this concept. Although Nkx2-5 inactivation in the SAN junction did not cause a malformed SAN at birth, the mutant mice manifested sinus node dysfunction. Thus, Nkx2-5 defines a population of pacemaker cells in the transitional zone. Despite Nkx2-5 being dispensable for SAN morphogenesis during embryogenesis, its deletion hampers atrial activation by the pacemaker.
Collapse
Affiliation(s)
- Hua Li
- Southern Center for Biomedical Research and Fujian Key Laboratory of Developmental and Neural Biology, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian Province 350108, PR China.,Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Dainan Li
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Yuzhi Wang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Zhen Huang
- Southern Center for Biomedical Research and Fujian Key Laboratory of Developmental and Neural Biology, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian Province 350108, PR China
| | - Jue Xu
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA.,West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan Province 610041, PR China
| | - Tianfang Yang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Linyan Wang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA.,State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province 610041, PR China
| | - Qinghuang Tang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Chen-Leng Cai
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hai Huang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Yanding Zhang
- Southern Center for Biomedical Research and Fujian Key Laboratory of Developmental and Neural Biology, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian Province 350108, PR China
| | - YiPing Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| |
Collapse
|
27
|
Linscheid N, Logantha SJRJ, Poulsen PC, Zhang S, Schrölkamp M, Egerod KL, Thompson JJ, Kitmitto A, Galli G, Humphries MJ, Zhang H, Pers TH, Olsen JV, Boyett M, Lundby A. Quantitative proteomics and single-nucleus transcriptomics of the sinus node elucidates the foundation of cardiac pacemaking. Nat Commun 2019; 10:2889. [PMID: 31253831 PMCID: PMC6599035 DOI: 10.1038/s41467-019-10709-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 05/28/2019] [Indexed: 12/13/2022] Open
Abstract
The sinus node is a collection of highly specialised cells constituting the heart’s pacemaker. The molecular underpinnings of its pacemaking abilities are debated. Using high-resolution mass spectrometry, we here quantify >7,000 proteins from sinus node and neighbouring atrial muscle. Abundances of 575 proteins differ between the two tissues. By performing single-nucleus RNA sequencing of sinus node biopsies, we attribute measured protein abundances to specific cell types. The data reveal significant differences in ion channels responsible for the membrane clock, but not in Ca2+ clock proteins, suggesting that the membrane clock underpins pacemaking. Consistently, incorporation of ion channel expression differences into a biophysically-detailed atrial action potential model result in pacemaking and a sinus node-like action potential. Combining our quantitative proteomics data with computational modeling, we estimate ion channel copy numbers for sinus node myocytes. Our findings provide detailed insights into the unique molecular make-up of the cardiac pacemaker. The sinus node generates rhythmic heartbeat but the molecular basis of pacemaking is still under debate. Here, the authors combine quantitative proteomics and single-nucleus transcriptomics to characterize the molecular composition of the sinus node and provide insights into the underpinnings of pacemaking.
Collapse
Affiliation(s)
- Nora Linscheid
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, København, 2200, Denmark
| | | | - Pi Camilla Poulsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, København, 2200, Denmark
| | - Shanzhuo Zhang
- School of Computer Science and Technology, Harbin Institute of Technology, Haerbin Shi, 150006, China
| | - Maren Schrölkamp
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, København, 2200, Denmark
| | - Kristoffer Lihme Egerod
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, København, 2200, Denmark
| | - Jonatan James Thompson
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, København, 2200, Denmark
| | - Ashraf Kitmitto
- Division of Cardiovascular Sciences, University of Manchester, Manchester, M13 9NT, UK
| | - Gina Galli
- Division of Cardiovascular Sciences, University of Manchester, Manchester, M13 9NT, UK
| | - Martin J Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, M13 9PT, UK
| | - Henggui Zhang
- Biological Physics Group, School of Physics & Astronomy, University of Manchester, Manchester, M13 9PL, UK
| | - Tune H Pers
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, København, 2200, Denmark
| | - Jesper Velgaard Olsen
- The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, København, 2200, Denmark
| | - Mark Boyett
- Division of Cardiovascular Sciences, University of Manchester, Manchester, M13 9NT, UK.
| | - Alicia Lundby
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, København, 2200, Denmark. .,The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, København, 2200, Denmark.
| |
Collapse
|
28
|
Natriuretic Peptide Receptor-C Protects Against Angiotensin II-Mediated Sinoatrial Node Disease in Mice. JACC Basic Transl Sci 2018; 3:824-843. [PMID: 30623142 PMCID: PMC6314975 DOI: 10.1016/j.jacbts.2018.08.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 08/15/2018] [Accepted: 08/16/2018] [Indexed: 12/19/2022]
Abstract
SAN disease is prevalent in hypertension and heart failure and can be induced by chronic Ang II treatment in mice. Ang II caused SAN disease in mice in association with impaired electrical conduction, reduction in the hyperpolarization-activated current (If) in SAN myocytes, and increased SAN fibrosis. Ang II-induced SAN disease was worsened in mice lacking NPR-C in association with enhanced SAN fibrosis. Mice co-treated with Ang II and an NPR-C agonist (cANF) were protected from SAN disease. NPR-C may represent a new target to protect against Ang II-induced SAN disease.
Sinoatrial node (SAN) disease mechanisms are poorly understood, and therapeutic options are limited. Natriuretic peptide(s) (NP) are cardioprotective hormones whose effects can be mediated partly by the NP receptor C (NPR-C). We investigated the role of NPR-C in angiotensin II (Ang II)-mediated SAN disease in mice. Ang II caused SAN disease due to impaired electrical activity in SAN myocytes and increased SAN fibrosis. Strikingly, Ang II treatment in NPR-C−/− mice worsened SAN disease, whereas co-treatment of wild-type mice with Ang II and a selective NPR-C agonist (cANF) prevented SAN dysfunction. NPR-C may represent a new target to protect against the development of Ang II-induced SAN disease.
Collapse
Key Words
- AP, action potential
- Ang II, angiotensin II
- CV, conduction velocity
- DD, diastolic depolarization
- Gmax, maximum conductance
- HR, heart rate
- ICa,L, L-type calcium current
- ICa,T, T-type calcium current
- INCX, sodium–calcium exchanger current
- IV, current voltage relationship
- If, hyperpolarization-activated current
- NP, natriuretic peptide
- NPR, natriuretic peptide receptor
- NPR-C, natriuretic peptide receptor C
- SAN, sinoatrial node
- SBP, systolic blood pressure
- V1/2(act), voltage for 50% channel activation
- cSNRT, corrected sinoatrial node recovery time
- fibrosis
- hypertension
- ion currents
- natriuretic peptide
- sinoatrial node
Collapse
|
29
|
Zhang J, Yang M, Yang AK, Wang X, Tang YH, Zhao QY, Wang T, Chen YT, Huang CX. Insulin gene enhancer binding protein 1 induces adipose tissue‑derived stem cells to differentiate into pacemaker‑like cells. Int J Mol Med 2018; 43:879-889. [PMID: 30483766 PMCID: PMC6317671 DOI: 10.3892/ijmm.2018.4002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 11/20/2018] [Indexed: 01/22/2023] Open
Abstract
Hybrid approaches combining gene- and cell-based therapies to make biological pacemakers are a promising therapeutic avenue for bradyarrhythmia. The present study aimed to direct adipose tissue-derived stem cells (ADSCs) to differentiate specifically into cardiac pacemaker cells by overexpressing a single transcription factor, insulin gene enhancer binding protein 1 (ISL-1). In the present study, the ADSCs were transfected with ISL‑1 or mCherry fluorescent protein lentiviral vectors and co-cultured with neonatal rat ventricular cardiomyocytes (NRVMs) in vitro for 5-7 days. The feasibility of regulating the differentiation of ADSCs into pacemaker-like cells by overexpressing ISL-1 was evaluated by observation of cell morphology and beating rate, reverse transcription-quantitative polymerase chain reaction analysis, western blotting, immunofluorescence and analysis of electrophysiological activity. In conclusion, these data indicated that the overexpression of ISL-1 in ADSCs may enhance the pacemaker phenotype and automaticity in vitro, features which were significantly increased following co‑culture induction.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Mei Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - An-Kang Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yan-Hong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qing-Yan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Teng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yu-Ting Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Cong-Xin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
30
|
Prando V, Da Broi F, Franzoso M, Plazzo AP, Pianca N, Francolini M, Basso C, Kay MW, Zaglia T, Mongillo M. Dynamics of neuroeffector coupling at cardiac sympathetic synapses. J Physiol 2018; 596:2055-2075. [PMID: 29524231 PMCID: PMC5983210 DOI: 10.1113/jp275693] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 02/28/2018] [Indexed: 01/16/2023] Open
Abstract
KEY POINTS The present study demonstrates, by in vitro and in vivo analyses, the novel concept that signal transmission between sympathetic neurons and the heart, underlying the physiological regulation of cardiac function, operates in a quasi-synaptic fashion. This is a result of the direct coupling between neurotransmitter releasing sites and effector cardiomyocyte membranes. ABSTRACT Cardiac sympathetic neurons (SNs) finely tune the rate and strength of heart contractions to match blood demand, both at rest and during acute stress, through the release of noradrenaline (NE). Junctional sites at the interface between the two cell types have been observed, although whether direct neurocardiac coupling has a role in heart physiology has not been clearly demonstrated to date. We investigated the dynamics of SN/cardiomyocyte intercellular signalling, both by fluorescence resonance energy transfer-based imaging of cAMP in co-cultures, as a readout of cardiac β-adrenergic receptor activation, and in vivo, using optogenetics in transgenic mice with SN-specific expression of Channelrhodopsin-2. We demonstrate that SNs and cardiomyocytes interact at specific sites in the human and rodent heart, as well as in co-cultures. Accordingly, neuronal activation elicited intracellular cAMP increases only in directly contacted myocytes and cell-cell coupling utilized a junctional extracellular signalling domain with an elevated NE concentration. In the living mouse, optogenetic activation of cardiac SNs innervating the sino-atrial node resulted in an instantaneous chronotropic effect, which shortened the heartbeat interval with single beat precision. Remarkably, inhibition of the optogenetically elicited chronotropic responses required a high dose of propranolol (20-50 mg kg-1 ), suggesting that sympathetic neurotransmission in the heart occurs at a locally elevated NE concentration. Our in vitro and in vivo data suggest that the control of cardiac function by SNs occurs via direct intercellular coupling as a result of the establishment of a specific junctional site.
Collapse
Affiliation(s)
- Valentina Prando
- Venetian Institute of Molecular MedicinePadovaItaly
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | | | - Mauro Franzoso
- Venetian Institute of Molecular MedicinePadovaItaly
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | | | - Nicola Pianca
- Venetian Institute of Molecular MedicinePadovaItaly
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | | | - Cristina Basso
- Department of Cardiac, Thoracic and Vascular SceincesUniversity of PadovaPadovaItaly
| | - Matthew W. Kay
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDCUSA
| | - Tania Zaglia
- Venetian Institute of Molecular MedicinePadovaItaly
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
- Department of Cardiac, Thoracic and Vascular SceincesUniversity of PadovaPadovaItaly
| | - Marco Mongillo
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
- University of MilanoMilanoItaly
- CNR Institute of NeurosciencePadovaItaly
| |
Collapse
|
31
|
Mäki-Marttunen T, Lines GT, Edwards AG, Tveito A, Dale AM, Einevoll GT, Andreassen OA. Pleiotropic effects of schizophrenia-associated genetic variants in neuron firing and cardiac pacemaking revealed by computational modeling. Transl Psychiatry 2017; 7:5. [PMID: 30446648 PMCID: PMC5802468 DOI: 10.1038/s41398-017-0007-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 07/07/2017] [Accepted: 07/14/2017] [Indexed: 01/01/2023] Open
Abstract
Schizophrenia patients have an increased risk of cardiac dysfunction. A possible factor underlying this comorbidity are the common variants in the large set of genes that have recently been discovered in genome-wide association studies (GWASs) as risk genes of schizophrenia. Many of these genes control the cell electrogenesis and calcium homeostasis. We applied biophysically detailed models of layer V pyramidal cells and sinoatrial node cells to study the contribution of schizophrenia-associated genes on cellular excitability. By including data from functional genomics literature to simulate the effects of common variants of these genes, we showed that variants of voltage-gated Na+ channel or hyperpolarization-activated cation channel-encoding genes cause qualitatively similar effects on layer V pyramidal cell and sinoatrial node cell excitability. By contrast, variants of Ca2+ channel or transporter-encoding genes mostly have opposite effects on cellular excitability in the two cell types. We also show that the variants may crucially affect the propagation of the cardiac action potential in the sinus node. These results may help explain some of the cardiac comorbidity in schizophrenia, and may facilitate generation of effective antipsychotic medications without cardiac side-effects such as arrhythmia.
Collapse
Affiliation(s)
- Tuomo Mäki-Marttunen
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway. .,Simula Research Laboratory and Center for Cardiological Innovation, Oslo, Norway.
| | - Glenn T. Lines
- Simula Research Laboratory and Center for Cardiological Innovation, Oslo, Norway
| | - Andrew G. Edwards
- Simula Research Laboratory and Center for Cardiological Innovation, Oslo, Norway
| | - Aslak Tveito
- Simula Research Laboratory and Center for Cardiological Innovation, Oslo, Norway
| | - Anders M. Dale
- 0000 0001 2107 4242grid.266100.3Multimodal Imaging Laboratory, UC San Diego, La Jolla, CA USA ,0000 0001 2107 4242grid.266100.3Department of Neurosciences, University of California San Diego, La Jolla, CA USA ,0000 0001 2107 4242grid.266100.3Department of Radiology, University of California, San Diego, La Jolla, CA USA
| | - Gaute T. Einevoll
- 0000 0004 0607 975Xgrid.19477.3cDepartment of Mathematical Sciences and Technology, Norwegian University of Life Sciences, Ås, Norway ,0000 0004 1936 8921grid.5510.1Department of Physics, University of Oslo, Oslo, Norway
| | - Ole A. Andreassen
- 0000 0004 1936 8921grid.5510.1NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway ,0000 0004 0389 8485grid.55325.34Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
32
|
Mitrofanova LB, Gorshkov AN, Konovalov PV, Krylova JS. Telocytes in the human sinoatrial node. J Cell Mol Med 2017; 22:521-532. [PMID: 29148209 PMCID: PMC5742677 DOI: 10.1111/jcmm.13340] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 07/05/2017] [Indexed: 12/25/2022] Open
Abstract
The sinoatrial node (SAN) is composed mostly of pacemaker, transitional and Purkinje‐like cells. Pacemaker cells, especially in the centre of the SAN, are surrounded by dense fibrous tissue and do not have any contact with transitional cells. We hypothesize that the SAN contains telocytes that have contacts with pacemaker cells and contractile myocardium. Immunohistochemistry using antibodies against HCN4 and antibody combinations against CD34 and HCN4 was carried out on 12 specimens. Confocal laser scanning microscopy (CLSM) with two mixtures of primary antibodies, namely CD34/S100 and vimentin/S100, was performed in three cases. In two cases, CLSM was carried out with CD117 antibody. Specimens for electron microscopy and immunocytochemistry with HCN4 immunogold labelling were taken from another three patients. In our study, we found cells with the immunophenotype of telocytes in the SAN. There were twice as many of these cells in the centre of the SAN as in the periphery (20.3 ± 4.8 versus 10.8 ± 4.4 per high‐power field). They had close contact with pacemaker cells and contractile cardiomyocytes and expressed HCN4. The ultrastructural characteristics of these cells are identical to those of telocytes observed earlier in other organs. Our study provides evidence that telocytes are present in the SAN.
Collapse
Affiliation(s)
- Lubov B Mitrofanova
- Department of Pathology, Federal Almazov North-West Medical Research Centre, Saint Petersburg, Russia
| | - Andrey N Gorshkov
- Department of Pathology, Federal Almazov North-West Medical Research Centre, Saint Petersburg, Russia.,Laboratory of Intracellular Signaling and Transport, Research Institute of Influenza of the Ministry of Healthcare of the Russian Federation, Saint Petersburg, Russia
| | - Petr V Konovalov
- Department of Pathology, Federal Almazov North-West Medical Research Centre, Saint Petersburg, Russia
| | - Julia S Krylova
- Laboratory of Cell Biology, Department of Pathology at the FSBI 'The Ott Research Institute of Obstetrics, Gynaecology and Reproductology', Saint Petersburg, Russia
| |
Collapse
|
33
|
Hassinen M, Haverinen J, Vornanen M. Small functional If current in sinoatrial pacemaker cells of the brown trout ( Salmo trutta fario) heart despite strong expression of HCN channel transcripts. Am J Physiol Regul Integr Comp Physiol 2017; 313:R711-R722. [PMID: 28855177 DOI: 10.1152/ajpregu.00227.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 08/21/2017] [Accepted: 08/24/2017] [Indexed: 01/26/2023]
Abstract
Funny current (If), formed by hyperpolarization-activated cyclic nucleotide-gated channels (HCN channels), is supposed to be crucial for the membrane clock regulating the cardiac pacemaker mechanism. We examined the presence and activity of HCN channels in the brown trout (Salmo trutta fario) sinoatrial (SA) pacemaker cells and their putative role in heart rate (fH) regulation. Six HCN transcripts (HCN1, HCN2a, HCN2ba, HCN2bb, HCN3, and HCN4) were expressed in the brown trout heart. The total HCN transcript abundance was 4.0 and 4.9 times higher in SA pacemaker tissue than in atrium and ventricle, respectively. In the SA pacemaker, HCN3 and HCN4 were the main isoforms representing 35.8 ± 2.7 and 25.0 ± 1.5%, respectively, of the total HCN transcripts. Only a small If with a mean current density of -1.2 ± 0.37 pA/pF at -140 mV was found in 4 pacemaker cells out of 16 spontaneously beating cells examined, despite the optimization of recording conditions for If activity. If was not found in any of the 24 atrial myocytes and 21 ventricular myocytes examined. HCN4 coexpressed with the MinK-related peptide 1 (MiRP1) β-subunit in CHO cells generated large If currents. In contrast, HCN3 (+MiRP1) failed to produce If in the same expression system. Cs+ (2 mM), which blocked 84 ± 12% of the native If, reversibly reduced fH 19.2 ± 3.6% of the excised multicellular pacemaker tissue from 53 ± 5 to 44 ± 5 beats/min (P < 0.05). However, this effect was probably due to the reduction of IKr, which was also inhibited (63.5 ± 4.6%) by Cs+ These results strongly suggest that fH regulation in the brown trout heart is largely independent on If.
Collapse
Affiliation(s)
- Minna Hassinen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland
| | - Jaakko Haverinen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland
| | - Matti Vornanen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland
| |
Collapse
|
34
|
Phosphodiesterases 3 and 4 Differentially Regulate the Funny Current, I f, in Mouse Sinoatrial Node Myocytes. J Cardiovasc Dev Dis 2017; 4. [PMID: 28868308 PMCID: PMC5573264 DOI: 10.3390/jcdd4030010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cardiac pacemaking, at rest and during the sympathetic fight-or-flight response, depends on cAMP (3',5'-cyclic adenosine monophosphate) signaling in sinoatrial node myocytes (SAMs). The cardiac "funny current" (If) is among the cAMP-sensitive effectors that drive pacemaking in SAMs. If is produced by hyperpolarization-activated, cyclic nucleotide-sensitive (HCN) channels. Voltage-dependent gating of HCN channels is potentiated by cAMP, which acts either by binding directly to the channels or by activating the cAMP-dependent protein kinase (PKA), which phosphorylates them. PKA activity is required for signaling between β adrenergic receptors (βARs) and HCN channels in SAMs but the mechanism that constrains cAMP signaling to a PKA-dependent pathway is unknown. Phosphodiesterases (PDEs) hydrolyze cAMP and form cAMP signaling domains in other types of cardiomyocytes. Here we examine the role of PDEs in regulation of If in SAMs. If was recorded in whole-cell voltage-clamp experiments from acutely-isolated mouse SAMs in the absence or presence of PDE and PKA inhibitors, and before and after βAR stimulation. General PDE inhibition caused a PKA-independent depolarizing shift in the midpoint activation voltage (V1/2) of If at rest and removed the requirement for PKA in βAR-to-HCN signaling. PDE4 inhibition produced a similar PKA-independent depolarizing shift in the V1/2 of If at rest, but did not remove the requirement for PKA in βAR-to-HCN signaling. PDE3 inhibition produced PKA-dependent changes in If both at rest and in response to βAR stimulation. Our results suggest that PDE3 and PDE4 isoforms create distinct cAMP signaling domains that differentially constrain access of cAMP to HCN channels and establish the requirement for PKA in signaling between βARs and HCN channels in SAMs.
Collapse
|
35
|
Du J, Deng S, Pu D, Liu Y, Xiao J, She Q. Age-dependent down-regulation of hyperpolarization-activated cyclic nucleotide-gated channel 4 causes deterioration of canine sinoatrial node function. Acta Biochim Biophys Sin (Shanghai) 2017; 49:400-408. [PMID: 28369243 DOI: 10.1093/abbs/gmx026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Indexed: 12/19/2022] Open
Abstract
The activity of pacemaker cells in the sinoatrial node (SAN) is an indicator of normal sinus rhythm. Clinical studies have revealed that the dysfunction of the SAN progressively increases with aging. In this study, we determined the changes in hyperpolarization-activated cyclic nucleotide-gated channel 4 (HCN4) expression and the relationship between aging and canine SAN dysfunction. The results of cardiac electrophysiological determination revealed that the intrinsic heart rate decreased from 168 ± 11 beats min-1 in young canines to 120 ± 9 beats min-1 in adults and to 88 ± 9 beats min-1 in aged canines. The sinus node recovery time (SNRT) increased from 412 ± 32 ms in young canines to 620 ± 56 ms in adults and to 838 ± 120 ms in aged canines. Corrected SNRT (CSNRT) increased from 55 ± 12 ms in young canines to 117 ± 27 ms in adults and to 171 ± 37 ms in aged canines. These results indicated that SAN function deteriorated with aging in the canine heart. However, histological staining illustrated that fibrosis was not significantly increased with aging in canine SAN. Real-time polymerase chain reaction indicated that the expression of HCN4 mRNA was downregulated in the elderly canine SAN. Similarly, we also verified that HCN4 protein expression within the SAN declined with aging via immunofluorescence staining and western blot analysis. Taken together, our data show that electrical remodeling, related to the down-regulation of HCN4, is responsible for the gradually increased incidence of SAN dysfunction with aging. Our results provide further evidence for explaining the mechanisms of age-related deterioration in the SAN.
Collapse
Affiliation(s)
- Jianlin Du
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Songbai Deng
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Di Pu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yajie Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jun Xiao
- Department of Cardiology, Chongqing Medical Emergency Center, Chongqing 400014, China
| | - Qiang She
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
36
|
Wen Y, Li B. The conduction system and expressions of hyperpolarization-activated cyclic nucleotide-gated cation channel 4 and connexin43 expressions in the hearts of fetal day 13 mice. Biotech Histochem 2017; 92:86-91. [PMID: 28296544 DOI: 10.1080/10520295.2016.1255994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
We investigated the development of the sinus node of the heart conduction system by localizing hyperpolarization-activated cyclic nucleotide-gated cation channel 4 (HCN4) and connexin43 (Cx43) in the hearts of fetal day 13 mice. Horizontal serial sections of day 13 whole fetuses were stained by hematoxylin and eosin and immunofluorescence to identify myocardial cells that express HCN4, hyperpolarization-activated cyclic nucleotide-gated cation channel 2 (HCN2) and Cx43. Expression levels of HCN4 and Cx43 were determined by quantitative RT-PCR in both fetal day 13 and adult mice. We found that both Cx43 and HCN4 expressions were located on the cell membranes in the hearts of fetal day 13 mice, but Cx43 was distributed throughout the myocardial cells. HCN4 expression was concentrated mainly in the left dorsal epicardium of the right atrium where Cx43 expression was low or absent. Quantitative RT-PCR demonstrated that HCN4 expression was significantly higher and HCN2 expression was significantly lower in fetal day 13 mice than in adults. We found no statistically significant difference in Cx43 expression between fetal day 13 mice and adults. HCN4 stained myocardial cells in the left dorsal epicardium of the right atrium are the origin of the sinus node and the remainder of the heart conduction system.
Collapse
Affiliation(s)
- Y Wen
- a Department of Histology and Embryology , College of Basic Medical Sciences
| | - B Li
- b Department of Sports Medicine, Shengjing Hospital , China Medical University , Shenyang , China
| |
Collapse
|
37
|
Sharpe EJ, Larson ED, Proenza C. Cyclic AMP reverses the effects of aging on pacemaker activity and If in sinoatrial node myocytes. J Gen Physiol 2017; 149:237-247. [PMID: 28057842 PMCID: PMC5299620 DOI: 10.1085/jgp.201611674] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 11/09/2016] [Accepted: 11/22/2016] [Indexed: 12/19/2022] Open
Abstract
Aging reduces pacemaker activity and shifts the voltage dependence of activation of the funny current, If, in sinoatrial node myocytes. Sharpe et al. find that these effects of aging can be reversed by application of exogenous cAMP but not by stimulation of endogenous cAMP. Aerobic capacity decreases with age, in part because of an age-dependent decline in maximum heart rate (mHR) and a reduction in the intrinsic pacemaker activity of the sinoatrial node of the heart. Isolated sinoatrial node myocytes (SAMs) from aged mice have slower spontaneous action potential (AP) firing rates and a hyperpolarizing shift in the voltage dependence of activation of the “funny current,” If. Cyclic AMP (cAMP) is a critical modulator of both AP firing rate and If in SAMs. Here, we test the ability of endogenous and exogenous cAMP to overcome age-dependent changes in acutely isolated murine SAMs. We found that maximal stimulation of endogenous cAMP with 3-isobutyl-1-methylxanthine (IBMX) and forskolin significantly increased AP firing rate and depolarized the voltage dependence of activation of If in SAMs from both young and aged mice. However, these changes were insufficient to overcome the deficits in aged SAMs, and significant age-dependent differences in AP firing rate and If persisted in the presence of IBMX and forskolin. In contrast, the effects of aging on SAMs were completely abolished by a high concentration of exogenous cAMP, which restored AP firing rate and If activation to youthful levels in cells from aged animals. Interestingly, the age-dependent differences in AP firing rates and If were similar in whole-cell and perforated-patch recordings, and the hyperpolarizing shift in If persisted in excised inside-out patches, suggesting a limited role for cAMP in causing these changes. Collectively, the data indicate that aging does not impose an absolute limit on pacemaker activity and that it does not act by simply reducing the concentration of freely diffusible cAMP in SAMs.
Collapse
Affiliation(s)
- Emily J Sharpe
- Department of Physiology and Biophysics, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045
| | - Eric D Larson
- Department of Physiology and Biophysics, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045
| | - 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
|
38
|
Abstract
Sino-atrial node (SAN) dysfunctions and associated complications constitute important causes of morbidity in patients with cardiac diseases. The development of novel pharmacological therapies to cure these patients relies on the thorough understanding of both normal physiology and pathophysiology of the SAN. Among the studies of cardiac pacemaking, the mouse SAN is widely used due to its feasibility for modifications in the expression of different genes that encode SAN ion channels or calcium handling proteins. Emerging evidence from electrophysiological and histological studies has also proved the representativeness and similarity of the mouse SAN structure and functions to larger mammals, including the presence of specialized conduction pathways from the SAN to the atrium and a complex pacemakers' hierarchy within the SAN. Recently, the technique of optical mapping has greatly facilitated the exploration and investigation of the origin of excitation and conduction within and from the mouse SAN, which in turn has extended the understanding of the SAN and benefited clinical treatments of SAN dysfunction associated diseases. In this manuscript, we have described in detail how to perform the optical mapping of the mouse SAN from the intact, Langendorff-perfused heart and from the isolated atrial preparation. This protocol is a useful tool to enhance the understanding of mouse SAN physiology and pathophysiology.
Collapse
Affiliation(s)
- Di Lang
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health
| | - Alexey V Glukhov
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health;
| |
Collapse
|
39
|
Development of the cardiac pacemaker. Cell Mol Life Sci 2016; 74:1247-1259. [PMID: 27770149 DOI: 10.1007/s00018-016-2400-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 10/16/2016] [Accepted: 10/17/2016] [Indexed: 01/11/2023]
Abstract
The sinoatrial node (SAN) is the dominant pacemaker of the heart. Abnormalities in SAN formation and function can cause sinus arrhythmia, including sick sinus syndrome and sudden death. A better understanding of genes and signaling pathways that regulate SAN development and function is essential to develop more effective treatment to sinus arrhythmia, including biological pacemakers. In this review, we briefly summarize the key processes of SAN morphogenesis during development, and focus on the transcriptional network that drives SAN development.
Collapse
|
40
|
Moghtadaei M, Jansen HJ, Mackasey M, Rafferty SA, Bogachev O, Sapp JL, Howlett SE, Rose RA. The impacts of age and frailty on heart rate and sinoatrial node function. J Physiol 2016; 594:7105-7126. [PMID: 27598221 DOI: 10.1113/jp272979] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 08/24/2016] [Indexed: 12/19/2022] Open
Abstract
KEY POINTS Sinoatrial node (SAN) function declines with age; however, not all individuals age at the same rate and health status can vary from fit to frail. Frailty was quantified in young and aged mice using a non-invasive frailty index so that the impacts of age and frailty on heart rate and SAN function could be assessed. SAN function was impaired in aged mice due to alterations in electrical conduction, changes in SAN action potential morphology and fibrosis in the SAN. Changes in SAN function, electrical conduction, action potential morphology and fibrosis were correlated with, and graded by, frailty. This study shows that mice of the same chronological age have quantifiable differences in health status that impact heart rate and SAN function and that these differences in health status can be identified using our frailty index. ABSTRACT Sinoatrial node (SAN) dysfunction increases with age, although not all older adults are affected in the same way. This is because people age at different rates and individuals of the same chronological age vary in health status from very fit to very frail. Our objective was to determine the impacts of age and frailty on heart rate (HR) and SAN function using a new model of frailty in ageing mice. Frailty, which was quantified in young and aged mice using a frailty index (FI), was greater in aged vs. young mice. Intracardiac electrophysiology demonstrated that HR was reduced whereas SAN recovery time (SNRT) was prolonged in aged mice; however, both parameters showed heteroscedasticity suggesting differences in health status among mice of similar chronological age. Consistent with this, HR and corrected SNRT were correlated with, and graded by, FI score. Optical mapping of the SAN demonstrated that conduction velocity (CV) was reduced in aged hearts in association with reductions in diastolic depolarization (DD) slope and action potential (AP) duration. In agreement with in vivo results, SAN CV, DD slope and AP durations all correlated with FI score. Finally, SAN dysfunction in aged mice was associated with increased interstitial fibrosis and alterations in expression of matrix metalloproteinases, which also correlated with frailty. These findings demonstrate that age-related SAN dysfunction occurs in association with electrical and structural remodelling and that frailty is a critical determinant of health status of similarly aged animals that correlates with changes in HR and SAN function.
Collapse
Affiliation(s)
- Motahareh Moghtadaei
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Hailey J Jansen
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Martin Mackasey
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Sara A Rafferty
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Oleg Bogachev
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - John L Sapp
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada.,Division of Cardiology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Susan E Howlett
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Robert A Rose
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada.,School of Biomedical Engineering, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
41
|
Logantha SJRJ, Stokke MK, Atkinson AJ, Kharche SR, Parveen S, Saeed Y, Sjaastad I, Sejersted OM, Dobrzynski H. Ca(2+)-Clock-Dependent Pacemaking in the Sinus Node Is Impaired in Mice with a Cardiac Specific Reduction in SERCA2 Abundance. Front Physiol 2016; 7:197. [PMID: 27313537 PMCID: PMC4889599 DOI: 10.3389/fphys.2016.00197] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 05/17/2016] [Indexed: 12/23/2022] Open
Abstract
Background: The sarcoplasmic reticulum Ca2+-ATPase (SERCA2) pump is an important component of the Ca2+-clock pacemaker mechanism that provides robustness and flexibility to sinus node pacemaking. We have developed transgenic mice with reduced cardiac SERCA2 abundance (Serca2 KO) as a model for investigating SERCA2's role in sinus node pacemaking. Methods and Results: In Serca2 KO mice, ventricular SERCA2a protein content measured by Western blotting was 75% (P < 0.05) lower than that in control mice (Serca2 FF) tissue. Immunofluorescent labeling of SERCA2a in ventricular, atrial, sinus node periphery and center tissue sections revealed 46, 45, 55, and 34% (all P < 0.05 vs. Serca2 FF) lower labeling, respectively and a mosaic pattern of expression. With telemetric ECG surveillance, we observed no difference in basal heart rate, but the PR-interval was prolonged in Serca2 KO mice: 49 ± 1 vs. 40 ± 1 ms (P < 0.001) in Serca2 FF. During exercise, heart rate in Serca2 KO mice was elevated to 667 ± 22 bpm, considerably less than 780 ± 17 bpm (P < 0.01) in Serca2 FF. In isolated sinus node preparations, 2 mM Cs+ caused bradycardia that was equally pronounced in Serca2 KO and Serca2 FF (32 ± 4% vs. 29 ± 5%), indicating no change in the pacemaker current, If. Disabling the Ca2+-clock with 2 μM ryanodine induced bradycardia that was less pronounced in Serca2 KO preparations (9 ± 1% vs. 20 ± 3% in Serca2 FF; P < 0.05), suggesting a disrupted Ca2+-clock. Mathematical modeling was used to dissect the effects of membrane- and Ca2+-clock components on Serca2 KO mouse heart rate and sinus node action potential. Computer modeling predicted a slowing of heart rate with SERCA2 downregulation and the heart rate slowing was pronounced at >70% reduction in SERCA2 activity. Conclusions:Serca2 KO mice show a disrupted Ca2+-clock-dependent pacemaker mechanism contributing to impaired sinus node and atrioventricular node function.
Collapse
Affiliation(s)
| | - Mathis K Stokke
- Institute for Experimental Medical Research, Oslo University Hospital and University of OsloOslo, Norway; Center for Heart Failure Research, University of OsloOslo, Norway; Clinic for Internal Medicine, Lovisenberg Deaconess Hospital ASOslo, Norway
| | - Andrew J Atkinson
- Institute of Cardiovascular Sciences, University of Manchester Manchester, UK
| | - Sanjay R Kharche
- Institute of Cardiovascular Sciences, University of Manchester Manchester, UK
| | - Sajida Parveen
- Institute of Cardiovascular Sciences, University of Manchester Manchester, UK
| | - Yawer Saeed
- Institute of Cardiovascular Sciences, University of Manchester Manchester, UK
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of OsloOslo, Norway; Center for Heart Failure Research, University of OsloOslo, Norway
| | - Ole M Sejersted
- Institute for Experimental Medical Research, Oslo University Hospital and University of OsloOslo, Norway; Center for Heart Failure Research, University of OsloOslo, Norway
| | - Halina Dobrzynski
- Institute of Cardiovascular Sciences, University of Manchester Manchester, UK
| |
Collapse
|
42
|
Innervation of sinoatrial nodal cells in the rabbit. Ann Anat 2016; 205:113-21. [DOI: 10.1016/j.aanat.2016.03.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 02/12/2016] [Accepted: 03/21/2016] [Indexed: 11/23/2022]
|
43
|
Stephenson RS, Agger P, Lunkenheimer PP, Zhao J, Smerup M, Niederer P, Anderson RH, Jarvis JC. The functional architecture of skeletal compared to cardiac musculature: Myocyte orientation, lamellar unit morphology, and the helical ventricular myocardial band. Clin Anat 2015; 29:316-32. [PMID: 26478993 DOI: 10.1002/ca.22661] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 10/13/2015] [Accepted: 10/15/2015] [Indexed: 11/06/2022]
Abstract
How the cardiomyocytes are aggregated within the heart walls remains contentious. We still do not fully understand how the end-to-end longitudinal myocytic chains are arranged, nor the true extent and shape of the lamellar units they aggregate to form. In this article, we show that an understanding of the complex arrangement of cardiac musculature requires knowledge of three-dimensional myocyte orientation (helical and intrusion angle), and appreciation of myocyte packing within the connective tissue matrix. We show how visualization and segmentation of high-resolution three-dimensional image data can accurately identify the morphology and orientation of the myocytic chains, and the lamellar units. Some maintain that the ventricles can be unwrapped in the form of a "helical ventricular myocardial band," that is, as a compartmentalized band with selective regional innervation and deformation, and a defined origin and insertion like most skeletal muscles. In contrast to the simpler interpretation of the helical ventricular myocardial band, we provide insight as to how the complex myocytic chains, the heterogeneous lamellar units, and connective tissue matrix form an interconnected meshwork, which facilitates the complex internal deformations of the ventricular wall. We highlight the dangers of disregarding the intruding cardiomyocytes. Preparation of the band destroys intruding myocytic chains, and thus disregards the functional implications of the antagonistic auxotonic forces they produce. We conclude that the ventricular myocardium is not analogous to skeletal muscle, but is a complex three-dimensional meshwork, with a heterogeneous branching lamellar architecture.
Collapse
Affiliation(s)
- Robert S Stephenson
- Research Institute for Sport and Exercise Science, Liverpool John Moores University, Liverpool, United Kingdom
| | - Peter Agger
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Paul P Lunkenheimer
- Department of Experimental Thoracic and Cardiovascular Surgery, University Hospital Munster, Munster, DE, Germany
| | - Jichao Zhao
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Morten Smerup
- Department of Cardiothoracic & Vascular Surgery, Aarhus University, Aarhus, Denmark
| | - Peter Niederer
- Institute for Biomedical Engineering, University of Zurich, Zurich, CH, Switzerland
| | - Robert H Anderson
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,Division of Biomedical Sciences, University College London, London, United Kingdom
| | - Jonathan C Jarvis
- Research Institute for Sport and Exercise Science, Liverpool John Moores University, Liverpool, United Kingdom
| |
Collapse
|
44
|
Li N, Csepe TA, Hansen BJ, Dobrzynski H, Higgins RSD, Kilic A, Mohler PJ, Janssen PML, Rosen MR, Biesiadecki BJ, Fedorov VV. Molecular Mapping of Sinoatrial Node HCN Channel Expression in the Human Heart. Circ Arrhythm Electrophysiol 2015; 8:1219-27. [PMID: 26304511 DOI: 10.1161/circep.115.003070] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 08/04/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND The hyperpolarization-activated current, If, plays an important role in sinoatrial node (SAN) pacemaking. Surprisingly, the distribution of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels in human SAN has only been investigated at the mRNA level. Our aim was to define the expression pattern of HCN proteins in human SAN and different atrial regions. METHODS AND RESULTS Entire SAN complexes were isolated from failing (n=5) and nonfailing (n=9) human hearts cardioplegically arrested in the operating room. Three-dimensional intramural SAN structure was identified as the fibrotic compact region around the SAN artery with Connexin 43-negative pacemaker cardiomyocytes visualized in Masson's trichrome and immunostained cryosections. SAN protein was precisely isolated from the adjacent frozen SAN tissue blocks using a 16G biopsy needle. The purity of the SAN protein was confirmed by Connexin 43 immunoblot. All 3 HCN isoform proteins were detected in SAN. HCN1 was predominantly distributed in the human SAN with a 125.1±40.2 (n=12) expression ratio of SAN to right atrium. HCN2 and HCN4 expression levels were higher in SAN than in atria, with SAN to right atrium ratios of 6.1±0.9 and 4.6±0.6 (n=12), respectively. CONCLUSIONS This is the first study to conduct precise 3D molecular mapping of the human SAN by isolating pure pacemaker SAN tissue. All 3 cardiac HCN isoforms had higher expression in the SAN than in the atria. HCN1 was almost exclusively expressed in SAN, emphasizing its utility as a new specific molecular marker of the human SAN and as a potential target of specific treatments intended to modify sinus rhythm.
Collapse
Affiliation(s)
- Ning Li
- From the Department of Physiology & Cell Biology and Dorothy M. Davis Heart & Lung Research Institute (N.L., T.A.C., B.J.H., P.J.M., P.M.L.J., B.J.B., V.V.F.), Department of Surgery and Dorothy M. Davis Heart & Lung Research Institute (R.S.D.H., A.K.), The Ohio State University Wexner Medical Center, Columbus; Institute of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom (H.D.); and Departments of Pharmacology and Pediatrics, Columbia University, New York, NY (M.R.R.)
| | - Thomas A Csepe
- From the Department of Physiology & Cell Biology and Dorothy M. Davis Heart & Lung Research Institute (N.L., T.A.C., B.J.H., P.J.M., P.M.L.J., B.J.B., V.V.F.), Department of Surgery and Dorothy M. Davis Heart & Lung Research Institute (R.S.D.H., A.K.), The Ohio State University Wexner Medical Center, Columbus; Institute of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom (H.D.); and Departments of Pharmacology and Pediatrics, Columbia University, New York, NY (M.R.R.)
| | - Brian J Hansen
- From the Department of Physiology & Cell Biology and Dorothy M. Davis Heart & Lung Research Institute (N.L., T.A.C., B.J.H., P.J.M., P.M.L.J., B.J.B., V.V.F.), Department of Surgery and Dorothy M. Davis Heart & Lung Research Institute (R.S.D.H., A.K.), The Ohio State University Wexner Medical Center, Columbus; Institute of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom (H.D.); and Departments of Pharmacology and Pediatrics, Columbia University, New York, NY (M.R.R.)
| | - Halina Dobrzynski
- From the Department of Physiology & Cell Biology and Dorothy M. Davis Heart & Lung Research Institute (N.L., T.A.C., B.J.H., P.J.M., P.M.L.J., B.J.B., V.V.F.), Department of Surgery and Dorothy M. Davis Heart & Lung Research Institute (R.S.D.H., A.K.), The Ohio State University Wexner Medical Center, Columbus; Institute of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom (H.D.); and Departments of Pharmacology and Pediatrics, Columbia University, New York, NY (M.R.R.)
| | - Robert S D Higgins
- From the Department of Physiology & Cell Biology and Dorothy M. Davis Heart & Lung Research Institute (N.L., T.A.C., B.J.H., P.J.M., P.M.L.J., B.J.B., V.V.F.), Department of Surgery and Dorothy M. Davis Heart & Lung Research Institute (R.S.D.H., A.K.), The Ohio State University Wexner Medical Center, Columbus; Institute of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom (H.D.); and Departments of Pharmacology and Pediatrics, Columbia University, New York, NY (M.R.R.)
| | - Ahmet Kilic
- From the Department of Physiology & Cell Biology and Dorothy M. Davis Heart & Lung Research Institute (N.L., T.A.C., B.J.H., P.J.M., P.M.L.J., B.J.B., V.V.F.), Department of Surgery and Dorothy M. Davis Heart & Lung Research Institute (R.S.D.H., A.K.), The Ohio State University Wexner Medical Center, Columbus; Institute of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom (H.D.); and Departments of Pharmacology and Pediatrics, Columbia University, New York, NY (M.R.R.)
| | - Peter J Mohler
- From the Department of Physiology & Cell Biology and Dorothy M. Davis Heart & Lung Research Institute (N.L., T.A.C., B.J.H., P.J.M., P.M.L.J., B.J.B., V.V.F.), Department of Surgery and Dorothy M. Davis Heart & Lung Research Institute (R.S.D.H., A.K.), The Ohio State University Wexner Medical Center, Columbus; Institute of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom (H.D.); and Departments of Pharmacology and Pediatrics, Columbia University, New York, NY (M.R.R.)
| | - Paul M L Janssen
- From the Department of Physiology & Cell Biology and Dorothy M. Davis Heart & Lung Research Institute (N.L., T.A.C., B.J.H., P.J.M., P.M.L.J., B.J.B., V.V.F.), Department of Surgery and Dorothy M. Davis Heart & Lung Research Institute (R.S.D.H., A.K.), The Ohio State University Wexner Medical Center, Columbus; Institute of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom (H.D.); and Departments of Pharmacology and Pediatrics, Columbia University, New York, NY (M.R.R.)
| | - Michael R Rosen
- From the Department of Physiology & Cell Biology and Dorothy M. Davis Heart & Lung Research Institute (N.L., T.A.C., B.J.H., P.J.M., P.M.L.J., B.J.B., V.V.F.), Department of Surgery and Dorothy M. Davis Heart & Lung Research Institute (R.S.D.H., A.K.), The Ohio State University Wexner Medical Center, Columbus; Institute of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom (H.D.); and Departments of Pharmacology and Pediatrics, Columbia University, New York, NY (M.R.R.)
| | - Brandon J Biesiadecki
- From the Department of Physiology & Cell Biology and Dorothy M. Davis Heart & Lung Research Institute (N.L., T.A.C., B.J.H., P.J.M., P.M.L.J., B.J.B., V.V.F.), Department of Surgery and Dorothy M. Davis Heart & Lung Research Institute (R.S.D.H., A.K.), The Ohio State University Wexner Medical Center, Columbus; Institute of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom (H.D.); and Departments of Pharmacology and Pediatrics, Columbia University, New York, NY (M.R.R.)
| | - Vadim V Fedorov
- From the Department of Physiology & Cell Biology and Dorothy M. Davis Heart & Lung Research Institute (N.L., T.A.C., B.J.H., P.J.M., P.M.L.J., B.J.B., V.V.F.), Department of Surgery and Dorothy M. Davis Heart & Lung Research Institute (R.S.D.H., A.K.), The Ohio State University Wexner Medical Center, Columbus; Institute of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom (H.D.); and Departments of Pharmacology and Pediatrics, Columbia University, New York, NY (M.R.R.).
| |
Collapse
|
45
|
Günther A, Baumann A. Distinct expression patterns of HCN channels in HL-1 cardiomyocytes. BMC Cell Biol 2015; 16:18. [PMID: 26141616 PMCID: PMC4490601 DOI: 10.1186/s12860-015-0065-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 06/05/2015] [Indexed: 01/05/2023] Open
Abstract
Background Cardiac rhythmic activity is initiated in functionally specialized areas of the heart. Hyperpolarization-activated and cyclic nucleotide-gated (HCN) channels are fundamental for these processes of cardiac physiology. Results Here we investigated transcript and protein expression patterns of HCN channels in HL-1 cardiomyocytes using a combination of quantitative PCR analysis and immunocytochemistry. Gene expression profiles of hcn1, hcn2 and hcn4 were acutely affected during HL-1 cell propagation. In addition, distinct expression patterns were uncovered for HCN1, HCN2 and HCN4 proteins. Conclusions Our results suggest that HCN channel isoforms might be involved in the concerted differentiation of HL-1 cells and may indirectly affect the occurrence of contractile HL-1 cell activity. We expect that these findings will promote studies on other molecular markers that contribute to cardiac physiology. Electronic supplementary material The online version of this article (doi:10.1186/s12860-015-0065-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anne Günther
- Institute of Complex Systems, Cellular Biophysics (ICS-4), Wilhelm-Johnen-Straße, Forschungszentrum Jülich, D-52425, Jülich, Germany.
| | - Arnd Baumann
- Institute of Complex Systems, Cellular Biophysics (ICS-4), Wilhelm-Johnen-Straße, Forschungszentrum Jülich, D-52425, Jülich, Germany.
| |
Collapse
|
46
|
Ye W, Wang J, Song Y, Yu D, Sun C, Liu C, Chen F, Zhang Y, Wang F, Harvey RP, Schrader L, Martin JF, Chen Y. A common Shox2-Nkx2-5 antagonistic mechanism primes the pacemaker cell fate in the pulmonary vein myocardium and sinoatrial node. Development 2015; 142:2521-32. [PMID: 26138475 DOI: 10.1242/dev.120220] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 06/04/2015] [Indexed: 12/26/2022]
Abstract
In humans, atrial fibrillation is often triggered by ectopic pacemaking activity in the myocardium sleeves of the pulmonary vein (PV) and systemic venous return. The genetic programs that abnormally reinforce pacemaker properties at these sites and how this relates to normal sinoatrial node (SAN) development remain uncharacterized. It was noted previously that Nkx2-5, which is expressed in the PV myocardium and reinforces a chamber-like myocardial identity in the PV, is lacking in the SAN. Here we present evidence that in mice Shox2 antagonizes the transcriptional output of Nkx2-5 in the PV myocardium and in a functional Nkx2-5(+) domain within the SAN to determine cell fate. Shox2 deletion in the Nkx2-5(+) domain of the SAN caused sick sinus syndrome, associated with the loss of the pacemaker program. Explanted Shox2(+) cells from the embryonic PV myocardium exhibited pacemaker characteristics including node-like electrophysiological properties and the capability to pace surrounding Shox2(-) cells. Shox2 deletion led to Hcn4 ablation in the developing PV myocardium. Nkx2-5 hypomorphism rescued the requirement for Shox2 for the expression of genes essential for SAN development in Shox2 mutants. Similarly, the pacemaker-like phenotype induced in the PV myocardium in Nkx2-5 hypomorphs reverted back to a working myocardial phenotype when Shox2 was simultaneously deleted. A similar mechanism is also adopted in differentiated embryoid bodies. We found that Shox2 interacts with Nkx2-5 directly, and discovered a substantial genome-wide co-occupancy of Shox2, Nkx2-5 and Tbx5, further supporting a pivotal role for Shox2 in the core myogenic program orchestrating venous pole and pacemaker development.
Collapse
Affiliation(s)
- Wenduo Ye
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Jun Wang
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine and the Texas Heart Institute, Houston, TX 77030, USA
| | - Yingnan Song
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA Southern Center for Biomedical Research and Fujian Key Laboratory of Developmental and Neural Biology, College of Life Science, Fujian Normal University, Fuzhou, Fujian 350108, P.R. China
| | - Diankun Yu
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Cheng Sun
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Chao Liu
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Fading Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Yanding Zhang
- Southern Center for Biomedical Research and Fujian Key Laboratory of Developmental and Neural Biology, College of Life Science, Fujian Normal University, Fuzhou, Fujian 350108, P.R. China
| | - Fen Wang
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA
| | - Richard P Harvey
- Developmental and Stem Cell Biology Division, The Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales 2010, Australia St. Vincent's Clinical School and School of Biological and Biomolecular Sciences, University of New South Wales, Kensington, New South Wales 2052, Australia
| | - Laura Schrader
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - James F Martin
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine and the Texas Heart Institute, Houston, TX 77030, USA
| | - YiPing Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA Southern Center for Biomedical Research and Fujian Key Laboratory of Developmental and Neural Biology, College of Life Science, Fujian Normal University, Fuzhou, Fujian 350108, P.R. China
| |
Collapse
|
47
|
Atrial Fibrillation and Fibrosis: Beyond the Cardiomyocyte Centric View. BIOMED RESEARCH INTERNATIONAL 2015; 2015:798768. [PMID: 26229964 PMCID: PMC4502285 DOI: 10.1155/2015/798768] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 03/27/2015] [Accepted: 03/30/2015] [Indexed: 12/31/2022]
Abstract
Atrial fibrillation (AF) associated with fibrosis is characterized by the appearance of interstitial myofibroblasts. These cells are responsible for the uncontrolled deposition of the extracellular matrix, which pathologically separate cardiomyocyte bundles. The enhanced fibrosis is thought to contribute to arrhythmias “indirectly” because a collagenous septum is a passive substrate for propagation, resulting in impulse conduction block and/or zigzag conduction. However, the emerging results demonstrate that myofibroblasts in vitro also promote arrhythmogenesis due to direct implications upon cardiomyocyte electrophysiology. This electrical interference may be considered beneficial as it resolves any conduction blocks; however, the passive properties of myofibroblasts might cause a delay in impulse propagation, thus promoting AF due to discontinuous slow conduction. Moreover, low-polarized myofibroblasts reduce, via cell-density dependence, the fast driving inward current for cardiac impulse conduction, therefore resulting in arrhythmogenic uniformly slow propagation. Critically, the subsequent reduction in cardiomyocytes resting membrane potential in vitro significantly increases the likelihood of ectopic activity. Myofibroblast densities and the degree of coupling at cellular border zones also impact upon this likelihood. By considering future in vivo studies, which identify myofibroblasts “per se” as a novel targets for cardiac arrhythmias, this review aims to describe the implications of noncardiomyocyte view in the context of AF.
Collapse
|
48
|
Choudhury M, Boyett MR, Morris GM. Biology of the Sinus Node and its Disease. Arrhythm Electrophysiol Rev 2015; 4:28-34. [PMID: 26835096 DOI: 10.15420/aer.2015.4.1.28] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 03/05/2015] [Indexed: 12/21/2022] Open
Abstract
The sinoatrial node (SAN) is the normal pacemaker of the heart and SAN dysfunction (SND) is common, but until recently the pathophysiology was incompletely understood. It was usually attributed to idiopathic age-related fibrosis and cell atrophy or ischaemia. It is now evident that changes in the electrophysiology of the SAN, known as electrical remodelling, is an important process that has been demonstrated in SND associated with heart failure, ageing, diabetes, atrial fibrillation and endurance exercise. Furthermore, familial SND has been identified and mutations have been characterised in key pacemaker genes of the SAN. This review summarises the current evidence regarding SAN function and the pathophysiology of SND.
Collapse
Affiliation(s)
- Moinuddin Choudhury
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Mark R Boyett
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Gwilym M Morris
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
49
|
St Clair JR, Sharpe EJ, Proenza C. Culture and adenoviral infection of sinoatrial node myocytes from adult mice. Am J Physiol Heart Circ Physiol 2015; 309:H490-8. [PMID: 26001410 DOI: 10.1152/ajpheart.00068.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 05/19/2015] [Indexed: 12/19/2022]
Abstract
Pacemaker myocytes in the sinoatrial node of the heart initiate each heartbeat by firing spontaneous action potentials. However, the molecular processes that underlie pacemaking are incompletely understood, in part because of our limited ability to manipulate protein expression within the native cellular context of sinoatrial node myocytes (SAMs). Here we describe a new method for the culture of fully differentiated SAMs from adult mice, and we demonstrate that robust expression of introduced proteins can be achieved within 24-48 h in vitro via adenoviral gene transfer. Comparison of morphological and electrophysiological characteristics of 48 h-cultured versus acutely isolated SAMs revealed only minor changes in vitro. Specifically, we found that cells tended to flatten in culture but retained an overall normal morphology, with no significant changes in cellular dimensions or membrane capacitance. Cultured cells beat spontaneously and, in patch-clamp recordings, the spontaneous action potential firing rate did not differ between cultured and acutely isolated cells, despite modest changes in a subset of action potential waveform parameters. The biophysical properties of two membrane currents that are critical for pacemaker activity in SAMs, the "funny current" (If) and voltage-gated Ca(2+) currents (ICa), were also indistinguishable between cultured and acutely isolated cells. This new method for culture and adenoviral infection of fully-differentiated SAMs from the adult mouse heart expands the range of experimental techniques that can be applied to study the molecular physiology of cardiac pacemaking because it will enable studies in which protein expression levels can be modified or genetically encoded reporter molecules expressed within SAMs.
Collapse
Affiliation(s)
- Joshua R St Clair
- Department of Physiology and Biophysics, University of Colorado - Anschutz Medical Campus, Denver, Colorado; and
| | - Emily J Sharpe
- Department of Physiology and Biophysics, University of Colorado - Anschutz Medical Campus, Denver, Colorado; and
| | - Catherine Proenza
- Department of Physiology and Biophysics, University of Colorado - Anschutz Medical Campus, Denver, Colorado; and Department of Medicine, Division of Cardiology - Anschutz Medical Campus, Denver, Colorado
| |
Collapse
|
50
|
Egom EE, Kruzliak P, Rotrekl V, Lei M. The effect of the sphingosine-1-phosphate analogue FTY720 on atrioventricular nodal tissue. J Cell Mol Med 2015; 19:1729-34. [PMID: 25864579 PMCID: PMC4511369 DOI: 10.1111/jcmm.12549] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 01/08/2015] [Indexed: 12/12/2022] Open
Abstract
The sphingosine‐1‐phosphate (S1P) receptor modulator, fingolimod (FTY720), has been used for the treatment of patients with relapsing forms of multiple sclerosis, but atrioventricular (AV) conduction block have been reported in some patients after the first dose. The underlying mechanism of this AV node conduction blockade is still not well‐understood. In this study, we hypothesize that expression of this particular arrhythmia might be related to a direct effect of FTY720 on AV node rather than a parasympathetic mimetic action. We, therefore, investigated the effect of FTY720 on AV nodal, using in vitro rat model preparation, under both basal as well as ischaemia/reperfusion conditions. We first look at the expression pattern of S1P receptors on the AV node using real‐time PCR. Although all three S1P receptor isoforms were expressed in AVN tissues, S1P1 receptor isoform expression level was higher than S1P2 and S1P3. The effect of 25 nM FTY720 on cycle length (CL) was subsequently studied via extracellular potentials recordings. FTY720 caused a mild to moderate prolongation in CL by an average 9% in AVN (n = 10, P < 0.05) preparations. We also show that FTY720 attenuated both ischaemia and reperfusion induced AVN rhythmic disturbance. To our knowledge, these remarkable findings have not been previously reported in the literature, and stress the importance for extensive monitoring period in certain cases, especially in patients taking concurrently AV node blocker agents.
Collapse
Affiliation(s)
- Emmanuel E Egom
- Egom Clinical and Translational Research Services, Halifax, NS, Canada
| | - Peter Kruzliak
- Department of Cardiovascular Diseases, International Clinical Research Center, St. Anne's University Hospital and Masaryk University, Brno, Czech Republic
| | - Vladimir Rotrekl
- Department of Cardiovascular Diseases, International Clinical Research Center, St. Anne's University Hospital and Masaryk University, Brno, Czech Republic.,Department of Medical Physics and Biophysics, Faculty of Medicine, Pavol Jozef Safarik University, Kosice, Slovak Republic
| | - Ming Lei
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Department of Pharmacology, University of Oxford, Oxford & Institute of Cardiovascular Sciences, UK
| |
Collapse
|