1
|
Bukaeva AA, Blokhina AV, Kharlap MS, Zaicenoka M, Zotova ED, Petukhova AV, Garbuzova EV, Zharikova AA, Divashuk MG, Kiseleva AV, Ershova AI, Meshkov AN, Drapkina OM. A Novel Bradycardia-Associated Variant in HCN4 as a Candidate Modifier in Type 3 Long QT Syndrome: Case Report and Deep In Silico Analysis. Biomedicines 2025; 13:1008. [PMID: 40299689 PMCID: PMC12025296 DOI: 10.3390/biomedicines13041008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 05/01/2025] Open
Abstract
Background: Genetic testing for long QT syndrome (LQTS) is straightforward in many families; however, in severe and complex cases, a single disease-causing variant may not be enough to explain all clinical features. In such cases, the search for genetic modifiers may be beneficial for precise diagnosis and management. Case presentation: We describe a three-generational family affected with clinically heterogeneous LQTS type 3 and bradycardia in which a novel missense variant p.V642M in HCN4 was identified in addition to the known pathogenic variant p.E1784K in SCN5A. We performed the detailed clinical investigation of the family and a deep in silico analysis of the discovered variants, showing the causal role of a new HCN4 variant in sinus bradycardia and its possible contribution to the phenotypic heterogeneity of LQTS type 3. Conclusions: This case is the first description of a functional variant in HCN4 as a candidate modifier in LQTS type 3 and demonstrates the importance of analyzing additional genetic variations in families with complex LQTS phenotypes.
Collapse
Affiliation(s)
- Anna A. Bukaeva
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.V.B.); (M.S.K.); (M.Z.); (A.V.P.); (E.V.G.); (A.A.Z.); (M.G.D.); (A.V.K.); (A.I.E.); (A.N.M.); (O.M.D.)
| | - Anastasia V. Blokhina
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.V.B.); (M.S.K.); (M.Z.); (A.V.P.); (E.V.G.); (A.A.Z.); (M.G.D.); (A.V.K.); (A.I.E.); (A.N.M.); (O.M.D.)
| | - Maria S. Kharlap
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.V.B.); (M.S.K.); (M.Z.); (A.V.P.); (E.V.G.); (A.A.Z.); (M.G.D.); (A.V.K.); (A.I.E.); (A.N.M.); (O.M.D.)
| | - Marija Zaicenoka
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.V.B.); (M.S.K.); (M.Z.); (A.V.P.); (E.V.G.); (A.A.Z.); (M.G.D.); (A.V.K.); (A.I.E.); (A.N.M.); (O.M.D.)
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
| | - Evgenia D. Zotova
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.V.B.); (M.S.K.); (M.Z.); (A.V.P.); (E.V.G.); (A.A.Z.); (M.G.D.); (A.V.K.); (A.I.E.); (A.N.M.); (O.M.D.)
| | - Anna V. Petukhova
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.V.B.); (M.S.K.); (M.Z.); (A.V.P.); (E.V.G.); (A.A.Z.); (M.G.D.); (A.V.K.); (A.I.E.); (A.N.M.); (O.M.D.)
| | - Elizaveta V. Garbuzova
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.V.B.); (M.S.K.); (M.Z.); (A.V.P.); (E.V.G.); (A.A.Z.); (M.G.D.); (A.V.K.); (A.I.E.); (A.N.M.); (O.M.D.)
| | - Anastasia A. Zharikova
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.V.B.); (M.S.K.); (M.Z.); (A.V.P.); (E.V.G.); (A.A.Z.); (M.G.D.); (A.V.K.); (A.I.E.); (A.N.M.); (O.M.D.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Mikhail G. Divashuk
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.V.B.); (M.S.K.); (M.Z.); (A.V.P.); (E.V.G.); (A.A.Z.); (M.G.D.); (A.V.K.); (A.I.E.); (A.N.M.); (O.M.D.)
- All-Russia Research Institute of Agricultural Biotechnology, 127550 Moscow, Russia
| | - Anna V. Kiseleva
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.V.B.); (M.S.K.); (M.Z.); (A.V.P.); (E.V.G.); (A.A.Z.); (M.G.D.); (A.V.K.); (A.I.E.); (A.N.M.); (O.M.D.)
| | - Alexandra I. Ershova
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.V.B.); (M.S.K.); (M.Z.); (A.V.P.); (E.V.G.); (A.A.Z.); (M.G.D.); (A.V.K.); (A.I.E.); (A.N.M.); (O.M.D.)
| | - Alexey N. Meshkov
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.V.B.); (M.S.K.); (M.Z.); (A.V.P.); (E.V.G.); (A.A.Z.); (M.G.D.); (A.V.K.); (A.I.E.); (A.N.M.); (O.M.D.)
| | - Oxana M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.V.B.); (M.S.K.); (M.Z.); (A.V.P.); (E.V.G.); (A.A.Z.); (M.G.D.); (A.V.K.); (A.I.E.); (A.N.M.); (O.M.D.)
| |
Collapse
|
2
|
Copier JS, Verkerk AO, Lodder EM. HCN4 in the atrioventricular node. Heart Rhythm 2025:S1547-5271(25)00200-0. [PMID: 39988103 DOI: 10.1016/j.hrthm.2025.02.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 02/25/2025]
Abstract
Hyperpolarization-activated cyclic nucleotide-gated channel 4 (HCN4) drives the funny current in cardiac pacemaker regions. Its involvement in sinoatrial node pacemaker generation is well known, but its function in the atrioventricular (AV) node (AVN) has not intensively been studied. HCN4 is expressed in the AVN, and its expression within the AVN seems similar across mammalian species with HCN4 presence in the inferior nodal extensions, compact node, and AV bundle. The main direct regulators of HCN4 are cAMP and protein kinase A. In addition, indirect regulators may affect HCN4 via trafficking and localization. However, these effects are underexplored in the AVN. AVN-specific effects in knockout and knockin mice include reduced funny current density and increased AV block. HCN4 expression in the AVN could be affected by aging, exercise, heart failure, and diabetes. This could underlie changes in PR interval, atria-His interval, Wenckebach cycle length, and AVN effective refractory period. Clinical reports link the HCN4 variant G1097W to AV block. Other clinical data come from studies assessing ivabradine, an HCN4 inhibitor. In animals, ivabradine resulted in prolonged PR and atrial-his intervals. To date, uncertainty regarding the role of HCN4 in the AVN remains. However, AVN-focused studies suggest HCN4's importance for AVN function. This review summarizes recent findings and highlights the involvement of HCN4 in normal and pathological AVN function.
Collapse
Affiliation(s)
- Jaël S Copier
- Experimental Cardiology, Amsterdam UMC, Amsterdam, The Netherlands; Heart Failure & Arrhythmias, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Arie O Verkerk
- Experimental Cardiology, Amsterdam UMC, Amsterdam, The Netherlands; Heart Failure & Arrhythmias, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands; Medical Biology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Elisabeth M Lodder
- Experimental Cardiology, Amsterdam UMC, Amsterdam, The Netherlands; Heart Failure & Arrhythmias, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
| |
Collapse
|
3
|
Zhang W, Wang F, Yin L, Tang Y, Wang X, Huang C. Cadherin-5 facilitated the differentiation of human induced pluripotent stem cells into sinoatrial node-like pacemaker cells by regulating β-catenin. J Cell Physiol 2024; 239:212-226. [PMID: 38149479 DOI: 10.1002/jcp.31161] [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] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/16/2023] [Accepted: 11/10/2023] [Indexed: 12/28/2023]
Abstract
Our study was conducted to investigate whether cadherin-5 (CDH5), a vascular endothelial cell adhesion glycoprotein, could facilitate the differentiation of human induced pluripotent stem cells (hiPSCs) into sinoatrial node-like pacemaker cells (SANLPCs), following previous findings of silk-fibroin hydrogel-induced direct conversion of quiescent cardiomyocytes into pacemaker cells in rats through the activation of CDH5. In this study, the differentiating hiPSCs were treated with CDH5 (40 ng/mL) between Day 5 and 7 during cardiomyocytes differentiation. The findings in the present study demonstrated that CDH5 stimulated the expression of pacemaker-specific markers while suppressing markers associated with working cardiomyocytes, resulting in an increased proportion of SANLPCs among hiPSCs-derived cardiomyocytes (hiPSC-CMs) population. Moreover, CDH5 induced typical electrophysiological characteristics resembling cardiac pacemaker cells in hiPSC-CMs. Further mechanistic investigations revealed that the enriched differentiation of hiPSCs into SANLPCs induced by CDH5 was partially reversed by iCRT14, an inhibitor of β-catenin. Therefore, based on the aforementioned findings, it could be inferred that the regulation of β-catenin by CDH5 played a crucial role in promoting the enriched differentiation of hiPSCs into SANLPCs, which presents a novel avenue for the construction of biological pacemakers in forthcoming research.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Fengyuan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Lin Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
4
|
Verkerk AO, Wilders R. The Action Potential Clamp Technique as a Tool for Risk Stratification of Sinus Bradycardia Due to Loss-of-Function Mutations in HCN4: An In Silico Exploration Based on In Vitro and In Vivo Data. Biomedicines 2023; 11:2447. [PMID: 37760888 PMCID: PMC10525944 DOI: 10.3390/biomedicines11092447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
These days, in vitro functional analysis of gene variants is becoming increasingly important for risk stratification of cardiac ion channelopathies. So far, such risk stratification has been applied to SCN5A, KCNQ1, and KCNH2 gene variants associated with Brugada syndrome and long QT syndrome types 1 and 2, respectively, but risk stratification of HCN4 gene variants related to sick sinus syndrome has not yet been performed. HCN4 is the gene responsible for the hyperpolarization-activated 'funny' current If, which is an important modulator of the spontaneous diastolic depolarization underlying the sinus node pacemaker activity. In the present study, we carried out a risk classification assay on those loss-of-function mutations in HCN4 for which in vivo as well as in vitro data have been published. We used the in vitro data to compute the charge carried by If (Qf) during the diastolic depolarization phase of a prerecorded human sinus node action potential waveform and assessed the extent to which this Qf predicts (1) the beating rate of the comprehensive Fabbri-Severi model of a human sinus node cell with mutation-induced changes in If and (2) the heart rate observed in patients carrying the associated mutation in HCN4. The beating rate of the model cell showed a very strong correlation with Qf from the simulated action potential clamp experiments (R2 = 0.95 under vagal tone). The clinically observed minimum or resting heart rates showed a strong correlation with Qf (R2 = 0.73 and R2 = 0.71, respectively). While a translational perspective remains to be seen, we conclude that action potential clamp on transfected cells, without the need for further voltage clamp experiments and data analysis to determine individual biophysical parameters of If, is a promising tool for risk stratification of sinus bradycardia due to loss-of-function mutations in HCN4. In combination with an If blocker, this tool may also prove useful when applied to human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) obtained from mutation carriers and non-carriers.
Collapse
Affiliation(s)
- Arie O. Verkerk
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
- Department of Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| |
Collapse
|
5
|
Crotti L, Brugada P, Calkins H, Chevalier P, Conte G, Finocchiaro G, Postema PG, Probst V, Schwartz PJ, Behr ER. From gene-discovery to gene-tailored clinical management: 25 years of research in channelopathies and cardiomyopathies. Europace 2023; 25:euad180. [PMID: 37622577 PMCID: PMC10450790 DOI: 10.1093/europace/euad180] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 06/03/2023] [Indexed: 08/26/2023] Open
Abstract
In the early nineties, few years before the birth of Europace, the clinical and scientific world of familial arrhythmogenic conditions was revolutionized by the identification of the first disease-causing genes. The explosion of genetic studies over a 15-year period led to the discovery of major disease-causing genes in practically all channelopathies and cardiomyopathies, bringing insight into the pathophysiological mechanisms of these conditions. The birth of next generation sequencing allowed a further step forward and other significant genes, as CALM1-3 in channelopathies and FLN C and TTN in cardiomyopathies were identified. Genotype-phenotype studies allowed the implementation of the genetic results in diagnosis, risk stratification, and therapeutic management with a different level of evidence in different arrhythmogenic conditions. The influence of common genetic variants, i.e. SNPs, on disease manifestation was proved in mid-twenties, and in the last 10 years with the advent of genome-wide association studies performed in familial arrhythmogenic diseases, the concept of polygenic risk score has been consolidated. Now, we are at the start of another amazing phase, i.e. the initiation of first gene therapy clinical trials.
Collapse
Affiliation(s)
- Lia Crotti
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Piazza dell'Ateneo Nuovo, 1 - 20126, Italy
- IRCCS Istituto Auxologico Italiano, Department of Cardiology, Cardiomyopathy Unit, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Piazzale Brescia, 20, 20149 Milan, Italy
| | - Pedro Brugada
- Heart Rhythm Management Centre, Postgraduate Program in Cardiac Electrophysiology and Pacing, Universitair Ziekenhuis Brussel-Vrije Universiteit Brussel, European Reference Networks Guard-Heart, Laarbeeklaan 101, Brussels 1090, Belgium
| | - Hugh Calkins
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Philippe Chevalier
- Neuromyogene Institute, Claude Bernard University, Lyon 1, Lyon, France
- Service de Rythmologie, Hospices Civils de Lyon, Lyon, France
| | - Giulio Conte
- Division of Cardiology, Istituto Cardiocentro Ticino, Ente Cantonale Ospedaliero, Lugano, Switzerland
| | - Gherardo Finocchiaro
- Cardiovascular Sciences Research Centre, St. George’s, University of London, London, UK
| | - Pieter G Postema
- Department of Cardiology, Amsterdam University Medical Centers, location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, the Netherlands
| | - Vincent Probst
- Centre Hospitalier Universitaire Nantes, Nantes Université, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Peter J Schwartz
- IRCCS Istituto Auxologico Italiano, Center for Cardiac Arrhythmias of Genetic Origin, Milan, Italy
| | - Elijah R Behr
- Cardiology Section, Institute of Molecular and Clinical Sciences, St. George's, University of London, London SW17 0RE, UK
- Department of Cardiology, Mayo Clinic Healthcare, 15 Portland Pl, London W1B 1PT, UK
- Department of Cardiology, St. George's University Hospitals NHS Foundation Trust, London SW17 0QT
| |
Collapse
|
6
|
Patel KK, Venkatesan C, Abdelhalim H, Zeeshan S, Arima Y, Linna-Kuosmanen S, Ahmed Z. Genomic approaches to identify and investigate genes associated with atrial fibrillation and heart failure susceptibility. Hum Genomics 2023; 17:47. [PMID: 37270590 DOI: 10.1186/s40246-023-00498-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/31/2023] [Indexed: 06/05/2023] Open
Abstract
Atrial fibrillation (AF) and heart failure (HF) contribute to about 45% of all cardiovascular disease (CVD) deaths in the USA and around the globe. Due to the complex nature, progression, inherent genetic makeup, and heterogeneity of CVDs, personalized treatments are believed to be critical. To improve the deciphering of CVD mechanisms, we need to deeply investigate well-known and identify novel genes that are responsible for CVD development. With the advancements in sequencing technologies, genomic data have been generated at an unprecedented pace to foster translational research. Correct application of bioinformatics using genomic data holds the potential to reveal the genetic underpinnings of various health conditions. It can help in the identification of causal variants for AF, HF, and other CVDs by moving beyond the one-gene one-disease model through the integration of common and rare variant association, the expressed genome, and characterization of comorbidities and phenotypic traits derived from the clinical information. In this study, we examined and discussed variable genomic approaches investigating genes associated with AF, HF, and other CVDs. We collected, reviewed, and compared high-quality scientific literature published between 2009 and 2022 and accessible through PubMed/NCBI. While selecting relevant literature, we mainly focused on identifying genomic approaches involving the integration of genomic data; analysis of common and rare genetic variants; metadata and phenotypic details; and multi-ethnic studies including individuals from ethnic minorities, and European, Asian, and American ancestries. We found 190 genes associated with AF and 26 genes linked to HF. Seven genes had implications in both AF and HF, which are SYNPO2L, TTN, MTSS1, SCN5A, PITX2, KLHL3, and AGAP5. We listed our conclusion, which include detailed information about genes and SNPs associated with AF and HF.
Collapse
Affiliation(s)
- Kush Ketan Patel
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson St, New Brunswick, NJ, USA
| | - Cynthia Venkatesan
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson St, New Brunswick, NJ, USA
| | - Habiba Abdelhalim
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson St, New Brunswick, NJ, USA
| | - Saman Zeeshan
- Rutgers Cancer Institute of New Jersey, Rutgers University, 195 Little Albany St, New Brunswick, NJ, USA
| | - Yuichiro Arima
- Developmental Cardiology Laboratory, International Research Center for Medical Sciences, Kumamoto University, 2-2-1 Honjo, Kumamoto City, Kumamoto, Japan
| | - Suvi Linna-Kuosmanen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Zeeshan Ahmed
- Department of Genetics and Genome Sciences, UConn Health, 400 Farmington Ave, Farmington, CT, USA.
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers Biomedical and Health Sciences, 125 Paterson St, New Brunswick, NJ, USA.
| |
Collapse
|
7
|
Münch JL, Paul F, Schmauder R, Benndorf K. Bayesian inference of kinetic schemes for ion channels by Kalman filtering. eLife 2022; 11:e62714. [PMID: 35506659 PMCID: PMC9342998 DOI: 10.7554/elife.62714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 04/22/2022] [Indexed: 11/16/2022] Open
Abstract
Inferring adequate kinetic schemes for ion channel gating from ensemble currents is a daunting task due to limited information in the data. We address this problem by using a parallelized Bayesian filter to specify hidden Markov models for current and fluorescence data. We demonstrate the flexibility of this algorithm by including different noise distributions. Our generalized Kalman filter outperforms both a classical Kalman filter and a rate equation approach when applied to patch-clamp data exhibiting realistic open-channel noise. The derived generalization also enables inclusion of orthogonal fluorescence data, making unidentifiable parameters identifiable and increasing the accuracy of the parameter estimates by an order of magnitude. By using Bayesian highest credibility volumes, we found that our approach, in contrast to the rate equation approach, yields a realistic uncertainty quantification. Furthermore, the Bayesian filter delivers negligibly biased estimates for a wider range of data quality. For some data sets, it identifies more parameters than the rate equation approach. These results also demonstrate the power of assessing the validity of algorithms by Bayesian credibility volumes in general. Finally, we show that our Bayesian filter is more robust against errors induced by either analog filtering before analog-to-digital conversion or by limited time resolution of fluorescence data than a rate equation approach.
Collapse
Affiliation(s)
- Jan L Münch
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich Schiller University JenaJenaGermany
| | - Fabian Paul
- Department of Biochemistry and Molecular Biology, University of ChicagoChicagoUnited States
| | - Ralf Schmauder
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich Schiller University JenaJenaGermany
| | - Klaus Benndorf
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich Schiller University JenaJenaGermany
| |
Collapse
|
8
|
Zhang W, Zhao H, Quan D, Tang Y, Wang X, Huang C. Tbx18 promoted the conversion of human-induced pluripotent stem cell-derived cardiomyocytes into sinoatrial node-like pacemaker cells. Cell Biol Int 2021; 46:403-414. [PMID: 34882885 DOI: 10.1002/cbin.11738] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/16/2021] [Accepted: 12/04/2021] [Indexed: 01/22/2023]
Abstract
Sinoatrial node (SAN) pacemaker cells originate from T-box transcription factor 18 (Tbx18)-expressing progenitor cells. The present study aimed to investigate whether overexpression of human transcription factor Tbx18 could reprogram human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) into SAN-like pacemaker cells (SANLPCs) in vitro. In the study, hiPSCs were first differentiated into hiPSC-CMs through regulating the Wnt/β-catenin pathway, then purified hiPSC-CMs were transfected by Tbx18 adenovirus (Tbx18-CMs group) or green fluorescent protein (GFP) adenovirus (GFP-CMs group). The beating frequency of the Tbx18-CMs group was significantly higher than that of the hiPSC-CMs group and GFP-CMs group. Compared with the other two groups, the expression levels of hyperpolarization-activated cyclic nucleotide-gated potassium channel isoform 4, connexin-45 in the Tbx18-CMs group were markedly upregulated, while the expressions of transcription factor NKX2.5, CX43 were significantly downregulated. Whole-cell patch-clamp results illustrated that action potential and "funny" current (If ) similar to SAN pacemaker cells could be recorded in the Tbx18-CMs group. In conclusion, this present study demonstrated that overexpression of Tbx18 promoted the conversion of hiPSC-CMs into SANLPCs.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P. R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P. R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P. R. China
| | - Hongyi Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P. R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P. R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P. R. China
| | - Dajun Quan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P. R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P. R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P. R. China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P. R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P. R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P. R. China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P. R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P. R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P. R. China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P. R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P. R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P. R. China
| |
Collapse
|
9
|
Iop L, Iliceto S, Civieri G, Tona F. Inherited and Acquired Rhythm Disturbances in Sick Sinus Syndrome, Brugada Syndrome, and Atrial Fibrillation: Lessons from Preclinical Modeling. Cells 2021; 10:3175. [PMID: 34831398 PMCID: PMC8623957 DOI: 10.3390/cells10113175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/03/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
Rhythm disturbances are life-threatening cardiovascular diseases, accounting for many deaths annually worldwide. Abnormal electrical activity might arise in a structurally normal heart in response to specific triggers or as a consequence of cardiac tissue alterations, in both cases with catastrophic consequences on heart global functioning. Preclinical modeling by recapitulating human pathophysiology of rhythm disturbances is fundamental to increase the comprehension of these diseases and propose effective strategies for their prevention, diagnosis, and clinical management. In silico, in vivo, and in vitro models found variable application to dissect many congenital and acquired rhythm disturbances. In the copious list of rhythm disturbances, diseases of the conduction system, as sick sinus syndrome, Brugada syndrome, and atrial fibrillation, have found extensive preclinical modeling. In addition, the electrical remodeling as a result of other cardiovascular diseases has also been investigated in models of hypertrophic cardiomyopathy, cardiac fibrosis, as well as arrhythmias induced by other non-cardiac pathologies, stress, and drug cardiotoxicity. This review aims to offer a critical overview on the effective ability of in silico bioinformatic tools, in vivo animal studies, in vitro models to provide insights on human heart rhythm pathophysiology in case of sick sinus syndrome, Brugada syndrome, and atrial fibrillation and advance their safe and successful translation into the cardiology arena.
Collapse
Affiliation(s)
- Laura Iop
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padua, Via Giustiniani, 2, I-35124 Padua, Italy; (S.I.); (G.C.)
| | | | | | - Francesco Tona
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padua, Via Giustiniani, 2, I-35124 Padua, Italy; (S.I.); (G.C.)
| |
Collapse
|
10
|
Bai X, Wang K, Boyett MR, Hancox JC, Zhang H. The Functional Role of Hyperpolarization Activated Current ( I f) on Cardiac Pacemaking in Human vs. in the Rabbit Sinoatrial Node: A Simulation and Theoretical Study. Front Physiol 2021; 12:582037. [PMID: 34489716 PMCID: PMC8417414 DOI: 10.3389/fphys.2021.582037] [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: 07/10/2020] [Accepted: 07/23/2021] [Indexed: 01/01/2023] Open
Abstract
The cardiac hyperpolarization-activated "funny" current (I f), which contributes to sinoatrial node (SAN) pacemaking, has a more negative half-maximal activation voltage and smaller fully-activated macroscopic conductance in human than in rabbit SAN cells. The consequences of these differences for the relative roles of I f in the two species, and for their responses to the specific bradycardic agent ivabradine at clinical doses have not been systematically explored. This study aims to address these issues, through incorporating rabbit and human I f formulations developed by Fabbri et al. into the Severi et al. model of rabbit SAN cells. A theory was developed to correlate the effect of I f reduction with the total inward depolarising current (I total) during diastolic depolarization. Replacing the rabbit I f formulation with the human one increased the pacemaking cycle length (CL) from 355 to 1,139 ms. With up to 20% I f reduction (a level close to the inhibition of I f by ivabradine at clinical concentrations), a modest increase (~5%) in the pacemaking CL was observed with the rabbit I f formulation; however, the effect was doubled (~12.4%) for the human I f formulation, even though the latter has smaller I f density. When the action of acetylcholine (ACh, 0.1 nM) was considered, a 20% I f reduction markedly increased the pacemaking CL by 37.5% (~27.3% reduction in the pacing rate), which is similar to the ivabradine effect at clinical concentrations. Theoretical analysis showed that the resultant increase of the pacemaking CL is inversely proportional to the magnitude of I total during diastolic depolarization phase: a smaller I f in the model resulted in a smaller I total amplitude, resulting in a slower pacemaking rate; and the same reduction in I f resulted in a more significant change of CL in the cell model with a smaller I total. This explained the mechanism by which a low dose of ivabradine slows pacemaking rate more in humans than in the rabbit. Similar results were seen in the Fabbri et al. model of human SAN cells, suggesting our observations are model-independent. Collectively, the results of study explain why low dose ivabradine at clinically relevant concentrations acts as an effective bradycardic agent in modulating human SAN pacemaking.
Collapse
Affiliation(s)
- Xiangyun Bai
- Biological Physics Group, Department of Physics and Astronomy, The University of Manchester, Manchester, United Kingdom
- School of Computer Science and Technology, Xi'an University of Posts and Telecommunications, Xi'an, China
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Kuanquan Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Mark R. Boyett
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, København, Denmark
| | - Jules C. Hancox
- Biological Physics Group, Department of Physics and Astronomy, The University of Manchester, Manchester, United Kingdom
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, Bristol, United Kingdom
| | - Henggui Zhang
- Biological Physics Group, Department of Physics and Astronomy, The University of Manchester, Manchester, United Kingdom
- Peng Cheng Laboratory, Shenzhen, China
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
11
|
Hoekstra M, van Ginneken ACG, Wilders R, Verkerk AO. HCN4 current during human sinoatrial node-like action potentials. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 166:105-118. [PMID: 34153331 DOI: 10.1016/j.pbiomolbio.2021.05.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/07/2021] [Accepted: 05/14/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Despite the many studies carried out over the past 40 years, the contribution of the HCN4 encoded hyperpolarization-activated 'funny' current (If) to pacemaker activity in the mammalian sinoatrial node (SAN), and the human SAN in particular, is still controversial and not fully established. OBJECTIVE To study the contribution of If to diastolic depolarization of human SAN cells and its dependence on heart rate, cAMP levels, and atrial load. METHODS HCN4 channels were expressed in human cardiac myocyte progenitor cells (CMPCs) and HCN4 currents assessed using perforated patch-clamp in traditional voltage clamp mode and during action potential clamp with human SAN-like action potential waveforms with 500-1500 ms cycle length, in absence or presence of forskolin to mimic β-adrenergic stimulation and a -15 mV command potential offset to mimic atrial load. RESULTS Forskolin significantly increased the fully-activated HCN4 current density at -140 mV by 14% and shifted the steady-state activation curve by +7.4 mV without affecting its slope. In addition, forskolin significantly accelerated current activation but slowed deactivation. The HCN4 current did not completely deactivate before the subsequent diastolic depolarization during action potential clamp. The amplitude of HCN4 current increased with increasing cycle length, was significantly larger in the presence of forskolin at all cycle lengths, and was significantly increased upon the negative offset to the command potential. CONCLUSIONS If is active during a human SAN action potential waveform and its amplitude is modulated by heart rate, β-adrenergic stimulation, and diastolic voltage range, such that If is under delicate control.
Collapse
Affiliation(s)
- Maaike Hoekstra
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Antoni C G van Ginneken
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
| | - Arie O Verkerk
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
12
|
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: 2.0] [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
|
13
|
Yang L, Gong Y, Tan Y, Wu L, Witman N, Zheng J, Zhang J, Fu W, Wang W. Dexmedetomidine exhibits antiarrhythmic effects on human-induced pluripotent stem cell-derived cardiomyocytes through a Na/Ca channel-mediated mechanism. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:399. [PMID: 33842620 PMCID: PMC8033317 DOI: 10.21037/atm-20-5898] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background Ventricular-like human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) exhibit the electrophysiological characteristics of spontaneous beating. Previous studies demonstrated that dexmedetomidine (DMED), a highly selective and widely used α2-adrenoceptor agonist for sedation, analgesia, and stress management, may induce antiarrhythmic effects, especially ventricular tachycardia. However, the underlying mechanisms of the DMED-mediated antiarrhythmic effects remain to be fully elucidated. Methods A conventional patch-clamp recording method was used to investigate the direct effects of DMED on spontaneous action potentials, pacemaker currents (If), potassium (K+) channel currents (IK1 and IKr), sodium (Na+) channel currents (INa), and calcium (Ca2+) channel currents (ICa) in ventricular-like hiPSC-CMs. Results DMED dose-dependently altered the frequency of ventricular-like spontaneous action potentials with a half-maximal inhibitory concentration (IC50) of 27.9 µM (n=6) and significantly prolonged the action potential duration at 90% repolarization (APD90). DMED also inhibited the amplitudes of the INa and ICa without affecting the activation and inactivation curves of these channels. DMED decreased the time constant of the Na+ and Ca2+ channel activation at potential –40 to –20 mv, and –20 mv. DMED increased the time constant of inactivation of the Na+ and Ca2+ channels. However, DMED did not affect the IK1, IKr, If, and their current-voltage relationship. The ability of DMED to decrease the spontaneous action potential frequency and the Na+ and Ca2+ channel amplitudes, were not blocked by yohimbine, idazoxan, or phentolamine. Conclusions DMED could inhibit the frequency of spontaneous action potentials and decrease the INa and ICa of hiPSC-CMs via mechanisms that were independent of the α2-adrenoceptor, the imidazoline receptor, and the α1-adrenoceptor. These inhibitory effects on hiPSC-CMs may contribute to the antiarrhythmic effects of DMED.
Collapse
Affiliation(s)
- Li Yang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yiqi Gong
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center; School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yao Tan
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center; School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Wu
- Department of Anesthesiology, Shanghai Children's Medical Center; School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Nevin Witman
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Jijian Zheng
- Department of Anesthesiology, Shanghai Children's Medical Center; School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Zhang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Fu
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center; School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Wang
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center; School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
14
|
cAMP-dependent regulation of HCN4 controls the tonic entrainment process in sinoatrial node pacemaker cells. Nat Commun 2020; 11:5555. [PMID: 33144559 PMCID: PMC7641277 DOI: 10.1038/s41467-020-19304-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 10/08/2020] [Indexed: 11/13/2022] Open
Abstract
It is highly debated how cyclic adenosine monophosphate-dependent regulation (CDR) of the major pacemaker channel HCN4 in the sinoatrial node (SAN) is involved in heart rate regulation by the autonomic nervous system. We addressed this question using a knockin mouse line expressing cyclic adenosine monophosphate-insensitive HCN4 channels. This mouse line displayed a complex cardiac phenotype characterized by sinus dysrhythmia, severe sinus bradycardia, sinus pauses and chronotropic incompetence. Furthermore, the absence of CDR leads to inappropriately enhanced heart rate responses of the SAN to vagal nerve activity in vivo. The mechanism underlying these symptoms can be explained by the presence of nonfiring pacemaker cells. We provide evidence that a tonic and mutual interaction process (tonic entrainment) between firing and nonfiring cells slows down the overall rhythm of the SAN. Most importantly, we show that the proportion of firing cells can be increased by CDR of HCN4 to efficiently oppose enhanced responses to vagal activity. In conclusion, we provide evidence for a novel role of CDR of HCN4 for the central pacemaker process in the sinoatrial node. The involvement of cAMP-dependent regulation of HCN4 in the chronotropic heart rate response is a matter of debate. Here the authors use a knockin mouse model expressing cAMP-insensitive HCN4 channels to discover an inhibitory nonfiring cell pool in the sinoatrial node and a tonic and mutual interaction between firing and nonfiring pacemaker cells that is controlled by cAMP-dependent regulation of HCN4, with implications in chronotropic heart rate responses.
Collapse
|
15
|
Baruscotti M, Bucchi A, Milanesi R, Paina M, Barbuti A, Gnecchi-Ruscone T, Bianco E, Vitali-Serdoz L, Cappato R, DiFrancesco D. A gain-of-function mutation in the cardiac pacemaker HCN4 channel increasing cAMP sensitivity is associated with familial Inappropriate Sinus Tachycardia. Eur Heart J 2019; 38:280-288. [PMID: 28182231 DOI: 10.1093/eurheartj/ehv582] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 10/01/2015] [Accepted: 10/07/2015] [Indexed: 01/09/2023] Open
Affiliation(s)
- Mirko Baruscotti
- Department of Biosciences, The PaceLab and 'Centro Interuniversitario di Medicina Molecolare e Biofisica Applicata', Università degli Studi di Milano, via Celoria 26, 20133 Milano, Italy
| | - Annalisa Bucchi
- Department of Biosciences, The PaceLab and 'Centro Interuniversitario di Medicina Molecolare e Biofisica Applicata', Università degli Studi di Milano, via Celoria 26, 20133 Milano, Italy
| | - Raffaella Milanesi
- Department of Biosciences, The PaceLab and 'Centro Interuniversitario di Medicina Molecolare e Biofisica Applicata', Università degli Studi di Milano, via Celoria 26, 20133 Milano, Italy
| | - Manuel Paina
- Department of Biosciences, The PaceLab and 'Centro Interuniversitario di Medicina Molecolare e Biofisica Applicata', Università degli Studi di Milano, via Celoria 26, 20133 Milano, Italy
| | - Andrea Barbuti
- Department of Biosciences, The PaceLab and 'Centro Interuniversitario di Medicina Molecolare e Biofisica Applicata', Università degli Studi di Milano, via Celoria 26, 20133 Milano, Italy
| | | | - Elisabetta Bianco
- Cardiovascular Department, 'Ospedali Riuniti di Trieste', University Hospital, Trieste, Italy
| | | | | | - Dario DiFrancesco
- Department of Biosciences, The PaceLab and 'Centro Interuniversitario di Medicina Molecolare e Biofisica Applicata', Università degli Studi di Milano, via Celoria 26, 20133 Milano, Italy
| |
Collapse
|
16
|
Liu R, Li J, Liu Y, Peng J, Guan X. The Effect of Astragaloside on Pacemaker Current and the Cytoskeleton in Rabbit Sinoatrial Node Cells Under the Ischemia and Reperfusion Condition. Front Pharmacol 2018; 9:551. [PMID: 29899698 PMCID: PMC5988886 DOI: 10.3389/fphar.2018.00551] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 05/08/2018] [Indexed: 11/13/2022] Open
Abstract
Objective: We investigated the role of astragaloside in the treatment of sick sinus syndrome (SSS). Methods: Neonatal New Zealand rabbits were selected for the study. Rabbit sinoatrial node (SAN) cells were isolated by the method of dual enzymatic digestion and differential adherence. The injury model was prepared through simulated ischemia and reperfusion (I/R), and changes in the pacemaker current (If) were recorded using the whole-cell patch-clamp technique. The proteins F-actin and vinculin were examined between various groups of SAN cells using a microplate reader and laser scanning confocal microscopy. The mRNA level and protein expression of hyperpolarization-activated cyclic nucleotide gated potassium channel 4 (HCN4) were assessed by q-PCR and western blot method. Results: The peak current density of If was decreased to -19.64 ± 2.14 pA/pF in SAN cells after simulated I/R, and the difference was highly significant (P < 0.01). Following simulated I/R, 100, 200, or 300 μmol L-1 astragaloside was added to the extracellular solution of SAN cells; the peak current density of the If increased to -30.43 ± 1.98, -34.83 ± 1.6, and -52.72 ± 1.7 pA/pF, respectively (P < 0.01). Adding 100 μmol L-1 astragaloside to normal SAN cells also led to an enhanced peak current density of the If (P < 0.05). In a concentration-dependent manner, especially at 300 μmol/L, astragaloside was capable of increasing the expression of HCN4 and protecting the structural stability of F-actin and vinculin in the damaged SAN cells. Conclusion: We estimated that astragaloside could shorten the action potential duration 20 (APD20) and APD50 in damaged SAN cells of neonatal rabbits, thereby increasing the expression of HCN4 and the If current density in damaged SAN cells of neonatal rabbits in a voltage-dependent manner, accelerating the steady-state activation of the If channels, and protecting damaged cytoskeleton.
Collapse
Affiliation(s)
- Ruxiu Liu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Li
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Liu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Peng
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuanke Guan
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
17
|
Yokoyama R, Kinoshita K, Hata Y, Abe M, Matsuoka K, Hirono K, Kano M, Nakazawa M, Ichida F, Nishida N, Tabata T. A mutant HCN4 channel in a family with bradycardia, left bundle branch block, and left ventricular noncompaction. Heart Vessels 2018; 33:802-819. [DOI: 10.1007/s00380-018-1116-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Accepted: 01/05/2018] [Indexed: 01/09/2023]
|
18
|
Mengesha HG, Tafesse TB, Bule MH. If Channel as an Emerging Therapeutic Target for Cardiovascular Diseases: A Review of Current Evidence and Controversies. Front Pharmacol 2017; 8:874. [PMID: 29225577 PMCID: PMC5705549 DOI: 10.3389/fphar.2017.00874] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 11/13/2017] [Indexed: 01/09/2023] Open
Abstract
In 2015, non-communicable diseases accounted for 39.5 million (70%) of the total 56.4 million deaths that occurred globally, of which 17.7 million (45%) were due to cardiovascular diseases. An elevated heart rate is considered to be one of the independent predictors and markers of future cardiovascular diseases. A variety of experimental and epidemiological studies have found that atherosclerosis, heart failure, coronary artery disease, stroke, and arrhythmia are linked to elevated heart rate. Although there are established drugs to reduce the heart rate, these drugs have undesirable side effects. Hence, the development of new drugs that selectively inhibit the heart rate is considered necessary. In the search for such drugs, almost four decades ago the If channel, also known as the “funny channel,” emerged as a novel site for the selective inhibition of heart rate. These If channels, with a mixed sodium and potassium inward current, have been identified in the sinoatrial node of the heart, which mediates the slow diastolic depolarization of the pacemaker of the spontaneous rhythmic cells. The hyperpolarization-activated cyclic nucleotide-gated (HCN) subfamily is primarily articulated in the heart and neurons that are encoded by a family of four genes (HCN1-4) and they identify the funny channel. Of these, HCN-4 is the principal protein in the sinoatrial node. Currently, funny channel inhibition is being targeted for the treatment and prevention of cardiovascular diseases such as atherosclerosis and stroke. A selective If channel inhibitor named ivabradine was discovered for clinical use in treating heart failure and coronary artery disease. However, inconsistencies regarding the clinical effects of ivabradine have been reported in the literature, suggesting the need for a rigorous analysis of the available evidence. The objective of this review is therefore to assess the current advances in targeting the If channel associated with ivabradine and related challenges.
Collapse
Affiliation(s)
- Hayelom G Mengesha
- Pharmacology and Toxicology Research Unit, School of Pharmacy, Mekelle University, Mekelle, Ethiopia.,College of Medicine and Health Science, Adigrat University, Adigrat, Ethiopia
| | - Tadesse B Tafesse
- School of Pharmacy, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Mohammed H Bule
- Department of Pharmacy, College of Medicine and Health Sciences, Ambo University, Ambo, Ethiopia
| |
Collapse
|
19
|
Li Y, Yang M, Zhang G, Li L, Ye B, Huang C, Tang Y. Transcription factor TBX18 promotes adult rat bone mesenchymal stem cell differentiation to biological pacemaker cells. Int J Mol Med 2017; 41:845-851. [PMID: 29207072 PMCID: PMC5752232 DOI: 10.3892/ijmm.2017.3259] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 11/01/2017] [Indexed: 01/17/2023] Open
Abstract
Bone mesenchymal stem cells (BMSCs) are currently considered the optimal stem cells for biological pacemaker cell transformation. The cardiac-specific transcription factor T-Box protein 18 (TBX18) is essential for sinoatrial node (SAN) formation, particularly formation of the head region that generates the electrical impulses that induce heart contraction. The present study aimed to confirm the effects of TBX18 on biological pace-maker differentiation of rat BMSCs. Flow cytometry was used to identify the surface markers of BMSCs, in order to acquire pure mesenchymal stem cells. Subsequently, BMSCs were transduced with TBX18 or green fluorescent protein adenovirus vectors. The effects of TBX18 were evaluated using SAN-specific makers including TBX18, α-actin, cardiac troponin I, hyperpolarization-activated cyclic nucleotide-gated channel 4 and connexin 43 by reverse transcription-quantitative polymerase chain reaction, western blotting and immunofluorescence. The findings demonstrated that direct conversion of BMSCs to biological pacemaker cells via TBX18 is a feasible method in the field of cardiology.
Collapse
Affiliation(s)
- Yanjun Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, Hubei 430060, P.R. China
| | - Mei Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, Hubei 430060, P.R. China
| | - Gege Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, Hubei 430060, P.R. China
| | - Le Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, Hubei 430060, P.R. China
| | - Bingjie Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, Hubei 430060, P.R. China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, Hubei 430060, P.R. China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
20
|
Sartiani L, Mannaioni G, Masi A, Novella Romanelli M, Cerbai E. The Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels: from Biophysics to Pharmacology of a Unique Family of Ion Channels. Pharmacol Rev 2017; 69:354-395. [PMID: 28878030 DOI: 10.1124/pr.117.014035] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/07/2017] [Indexed: 12/22/2022] Open
Abstract
Hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels are important members of the voltage-gated pore loop channels family. They show unique features: they open at hyperpolarizing potential, carry a mixed Na/K current, and are regulated by cyclic nucleotides. Four different isoforms have been cloned (HCN1-4) that can assemble to form homo- or heterotetramers, characterized by different biophysical properties. These proteins are widely distributed throughout the body and involved in different physiologic processes, the most important being the generation of spontaneous electrical activity in the heart and the regulation of synaptic transmission in the brain. Their role in heart rate, neuronal pacemaking, dendritic integration, learning and memory, and visual and pain perceptions has been extensively studied; these channels have been found also in some peripheral tissues, where their functions still need to be fully elucidated. Genetic defects and altered expression of HCN channels are linked to several pathologies, which makes these proteins attractive targets for translational research; at the moment only one drug (ivabradine), which specifically blocks the hyperpolarization-activated current, is clinically available. This review discusses current knowledge about HCN channels, starting from their biophysical properties, origin, and developmental features, to (patho)physiologic role in different tissues and pharmacological modulation, ending with their present and future relevance as drug targets.
Collapse
Affiliation(s)
- Laura Sartiani
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| | - Guido Mannaioni
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| | - Alessio Masi
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| | - Maria Novella Romanelli
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| | - Elisabetta Cerbai
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| |
Collapse
|
21
|
Bai X, Wang K, Yuan Y, Li Q, Dobrzynski H, Boyett MR, Hancox JC, Zhang H. Mechanism underlying impaired cardiac pacemaking rhythm during ischemia: A simulation study. CHAOS (WOODBURY, N.Y.) 2017; 27:093934. [PMID: 28964153 DOI: 10.1063/1.5002664] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Ischemia in the heart impairs function of the cardiac pacemaker, the sinoatrial node (SAN). However, the ionic mechanisms underlying the ischemia-induced dysfunction of the SAN remain elusive. In order to investigate the ionic mechanisms by which ischemia causes SAN dysfunction, action potential models of rabbit SAN and atrial cells were modified to incorporate extant experimental data of ischemia-induced changes to membrane ion channels and intracellular ion homeostasis. The cell models were incorporated into an anatomically detailed 2D model of the intact SAN-atrium. Using the multi-scale models, the functional impact of ischemia-induced electrical alterations on cardiac pacemaking action potentials (APs) and their conduction was investigated. The effects of vagal tone activity on the regulation of cardiac pacemaker activity in control and ischemic conditions were also investigated. The simulation results showed that at the cellular level ischemia slowed the SAN pacemaking rate, which was mainly attributable to the altered Na+-Ca2+ exchange current and the ATP-sensitive potassium current. In the 2D SAN-atrium tissue model, ischemia slowed down both the pacemaking rate and the conduction velocity of APs into the surrounding atrial tissue. Simulated vagal nerve activity, including the actions of acetylcholine in the model, amplified the effects of ischemia, leading to possible SAN arrest and/or conduction exit block, which are major features of the sick sinus syndrome. In conclusion, this study provides novel insights into understanding the mechanisms by which ischemia alters SAN function, identifying specific conductances as contributors to bradycardia and conduction block.
Collapse
Affiliation(s)
- Xiangyun Bai
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Kuanquan Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Yongfeng Yuan
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Qince Li
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Halina Dobrzynski
- Institute of Cardiovascular Sciences, The University of Manchester, M13 9PL Manchester, United Kingdom
| | - Mark R Boyett
- Institute of Cardiovascular Sciences, The University of Manchester, M13 9PL Manchester, United Kingdom
| | - Jules C Hancox
- Biological Physics Group, School of Physics and Astronomy, The University of Manchester, M13 9PL Manchester, United Kingdom
| | - Henggui Zhang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| |
Collapse
|
22
|
Lodder EM, Verkerk AO, Bezzina CR. Pacing Discovery: G-Protein β Subunit Mutations in Sinus Node Dysfunction. Circ Res 2017; 120:1524-1526. [PMID: 28495981 DOI: 10.1161/circresaha.117.310953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Elisabeth M Lodder
- From the Heart Center, Department of Clinical and Experimental Cardiology (E.M.L., A.O.V., C.R.B.) and Department of Medical Biology (A.O.V.), Academic Medical Center, Amsterdam, the Netherlands
| | - Arie O Verkerk
- From the Heart Center, Department of Clinical and Experimental Cardiology (E.M.L., A.O.V., C.R.B.) and Department of Medical Biology (A.O.V.), Academic Medical Center, Amsterdam, the Netherlands
| | - Connie R Bezzina
- From the Heart Center, Department of Clinical and Experimental Cardiology (E.M.L., A.O.V., C.R.B.) and Department of Medical Biology (A.O.V.), Academic Medical Center, Amsterdam, the Netherlands.
| |
Collapse
|
23
|
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: 1.1] [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
|
24
|
Rezazadeh S, Duff HJ. Genetic Determinants of Hereditary Bradyarrhythmias: A Contemporary Review of a Diverse Group of Disorders. Can J Cardiol 2017; 33:758-767. [PMID: 28545623 DOI: 10.1016/j.cjca.2017.03.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/03/2017] [Accepted: 03/10/2017] [Indexed: 11/25/2022] Open
Abstract
Bradyarrhythmia is a common clinical presentation. Although the majority of cases are acquired, genetic screening of families with bradyarrhythmia has led to the discovery of a growing number of causative hereditary mutations. These mutations can interfere with any of the steps required for the occurrence of each cardiac cycle, including generation of an action potential in the sinoatrial node, successful exit of the action potential from the node, propagation of the action potential throughout the atria until the depolarization waves reach the atrioventricular node, and finally transmission of the action potential to the ventricles through the His-Purkinje system. As expected, channelopathies are the predominant culprit for hereditary bradyarrhythmias, because they play a crucial role in action potential generation and propagation. Interestingly, there are an increasing number of genes that encode for various regulatory or structural cellular components that have been linked to hereditary bradyarrhythmias. Furthermore, population-based genetic screening has revealed that age-related conduction defects may in fact be caused by genetic predispositions rather than the simple process of aging. With recent advances in genetic testing and the creation of animal models, not only have we discovered new culprit genes but it has also has become evident that there are still significant gaps in our knowledge of cardiac pathophysiology. In this review, we discuss the clinical presentations of known hereditary bradyarrhythmias and their associated conditions in addition to detailing our current molecular understanding of the mechanisms by which they are manifested.
Collapse
Affiliation(s)
- Saman Rezazadeh
- Department of Cardiac Sciences and Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Henry J Duff
- Department of Cardiac Sciences and Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
25
|
Boulton S, Akimoto M, Akbarizadeh S, Melacini G. Free energy landscape remodeling of the cardiac pacemaker channel explains the molecular basis of familial sinus bradycardia. J Biol Chem 2017; 292:6414-6428. [PMID: 28174302 DOI: 10.1074/jbc.m116.773697] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/28/2017] [Indexed: 12/21/2022] Open
Abstract
The hyperpolarization-activated and cyclic nucleotide-modulated ion channel (HCN) drives the pacemaker activity in the heart, and its malfunction can result in heart disorders. One such disorder, familial sinus bradycardia, is caused by the S672R mutation in HCN, whose electrophysiological phenotypes include a negative shift in the channel activation voltage and an accelerated HCN deactivation. The outcomes of these changes are abnormally low resting heart rates. However, the molecular mechanism underlying these electrophysiological changes is currently not fully understood. Crystallographic investigations indicate that the S672R mutation causes limited changes in the structure of the HCN intracellular gating tetramer, but its effects on protein dynamics are unknown. Here, we utilize comparative S672R versus WT NMR analyses to show that the S672R mutation results in extensive perturbations of the dynamics in both apo- and holo-forms of the HCN4 isoform, reflecting how S672R remodels the free energy landscape for the modulation of HCN4 by cAMP, i.e. the primary cyclic nucleotide modulator of HCN channels. We show that the S672R mutation results in a constitutive shift of the dynamic auto-inhibitory equilibrium toward inactive states of HCN4 and broadens the free-energy well of the apo-form, enhancing the millisecond to microsecond dynamics of the holo-form at sites critical for gating cAMP binding. These S672R-induced variations in dynamics provide a molecular basis for the electrophysiological phenotypes of this mutation and demonstrate that the pathogenic effects of the S672R mutation can be rationalized primarily in terms of modulations of protein dynamics.
Collapse
Affiliation(s)
- Stephen Boulton
- From the Departments of Biochemistry and Biomedical Sciences and
| | - Madoka Akimoto
- Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario L8S 4M1, Canada
| | - Sam Akbarizadeh
- Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario L8S 4M1, Canada
| | - Giuseppe Melacini
- From the Departments of Biochemistry and Biomedical Sciences and .,Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario L8S 4M1, Canada
| |
Collapse
|
26
|
George SA, Faye NR, Murillo-Berlioz A, Lee KB, Trachiotis GD, Efimov IR. At the Atrioventricular Crossroads: Dual Pathway Electrophysiology in the Atrioventricular Node and its Underlying Heterogeneities. Arrhythm Electrophysiol Rev 2017; 6:179-185. [PMID: 29326832 DOI: 10.15420/aer.2017.30.1] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The atrioventricular node (AVN) is a complex structure that performs a variety of functions in the heart. The AVN is primarily an electrical gatekeeper between the atria and ventricles and introduces a delay between atrial and ventricular excitation, allowing for efficient ventricular filling. The AVN is composed of several compartments that safely transmit electrical excitation from the atria to the ventricles via the fast or slow pathways. There are many electrophysiological differences between these pathways, including conduction time and electrical refractoriness, that increase the predisposition of the atrioventricular junction to arrhythmias such as atrioventricular nodal re-entrant tachycardia. These varied electrophysiological characteristics of the fast and slow pathways stem from their unique structural and molecular composition (tissue and cellular geometry, ion channels and gap junctions). This review summarises the structural and molecular heterogeneities of the human AVN and how they result in electrophysiological variations and arrhythmias.
Collapse
Affiliation(s)
- Sharon A George
- Department of Biomedical Engineering, The George Washington University,Washington, DC, USA
| | - N Rokhaya Faye
- Department of Biomedical Engineering, The George Washington University,Washington, DC, USA
| | - Alejandro Murillo-Berlioz
- Department of Biomedical Engineering, The George Washington University,Washington, DC, USA.,Division of Cardiothoracic Surgery and Cardiothoracic Research, Veterans Affairs Medical Center,Washington, DC, USA
| | - K Benjamin Lee
- Department of Biomedical Engineering, The George Washington University,Washington, DC, USA.,Division of Cardiothoracic Surgery and Cardiothoracic Research, Veterans Affairs Medical Center,Washington, DC, USA
| | - Gregory D Trachiotis
- Division of Cardiothoracic Surgery and Cardiothoracic Research, Veterans Affairs Medical Center,Washington, DC, USA
| | - Igor R Efimov
- Department of Biomedical Engineering, The George Washington University,Washington, DC, USA
| |
Collapse
|
27
|
Ishikawa T, Tsuji Y, Makita N. Inherited bradyarrhythmia: A diverse genetic background. J Arrhythm 2015; 32:352-358. [PMID: 27761158 PMCID: PMC5063261 DOI: 10.1016/j.joa.2015.09.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 09/03/2015] [Accepted: 09/16/2015] [Indexed: 12/31/2022] Open
Abstract
Bradyarrhythmia is a common heart rhythm abnormality comprising number of diseases and is associated with decreased heart rate due to the failure of action potential generation and propagation at the sinus node. Permanent pacemaker implantation is often used therapeutically to compensate for decreased heart rate and cardiac output. The vast majority of bradyarrhythmia cases are attributable either to aging or to structural abnormalities of the cardiac conduction system, caused by underlying structural heart disease. However, there is a subset of bradyarrhythmia primarily caused by genetic defects in the absence of aging or underlying structural heart disease. These include several genes that play principal roles in cardiac electrophysiology, heart development, cardioprotection, and the structural integrity of the membrane and sarcomere. Recent advances in the functional analysis of mutations using a heterologous expression system and genetically engineered animal models have provided significant insights into the underlying molecular mechanisms responsible for inherited arrhythmia. In this review, current understandings of the genetic and molecular basis of inherited bradyarrhythmia are presented.
Collapse
Affiliation(s)
- Taisuke Ishikawa
- Department of Molecular Physiology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Yukiomi Tsuji
- Department of Molecular Physiology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Naomasa Makita
- Department of Molecular Physiology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| |
Collapse
|
28
|
DiFrancesco D. HCN4, Sinus Bradycardia and Atrial Fibrillation. Arrhythm Electrophysiol Rev 2015; 4:9-13. [PMID: 26835093 DOI: 10.15420/aer.2015.4.1.9] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 01/29/2015] [Indexed: 01/01/2023] Open
Abstract
Based on their established role in the generation of spontaneous activity in pacemaker cells and control of cardiac rate, funny/ hyperpolarisation-activated, cyclic nucleotide gated 4 (HCN4) channels are natural candidates in the search for causes of sinus arrhythmias. Investigation of funny current-related inheritable arrhythmias has led to the identification of several mutations of the HCN4 gene associated with bradycardia and/or more complex arrhythmias. More recently, the search has been extended to include auxiliary proteins such as the minK-related peptide 1 (MiRP1) β-subunit. All mutations described so far are loss-of-function and in agreement with the role of funny channels, the predominant type of arrhythmia found is bradycardia. Funny channel-linked arrhythmias, however, also include atrioventricular (AV) block and atrial fibrillation, in agreement with an emerging new concept according to which defective funny channels have a still unexplored role in impairing AV conduction and triggering atrial fibrillation. Also, importantly, recent work shows that HCN4 mutations can be associated with cardiac structural abnormalities. In this short review I briefly address the current knowledge of funny/HCN4 channel mutations and associated sinus and more complex arrhythmias.
Collapse
Affiliation(s)
- Dario DiFrancesco
- PaceLab, University of Milan and Centro Interuniversitario di Medicina Molecolare e Biofisica Applicata (CIMMBA), Milan, Italy
| |
Collapse
|
29
|
Zhou C, Ke B, Zhao Y, Liang P, Liao D, Li T, Liu J, Chen X. Hyperpolarization-activated cyclic nucleotide-gated channels may contribute to regional anesthetic effects of lidocaine. Anesthesiology 2015; 122:606-618. [PMID: 25485469 DOI: 10.1097/aln.0000000000000557] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Local anesthetics (e.g., lidocaine) have been found to inhibit hyperpolarization-activated cyclic nucleotide-gated (HCN) channels besides sodium channels. However, the exact role of HCN channels in regional anesthesia in vivo is still elusive. METHODS Sciatic nerve block and intrathecal anesthesia were performed using lidocaine in wild-type and HCN1 channel knockout (HCN1) mice. EC50 of lidocaine and durations of 1% lidocaine were determined. In electrophysiologic recordings, effects of lidocaine on HCN channel currents, voltage-gated sodium channel currents, and neural membrane properties were recorded on dorsal root ganglia neurons. RESULTS In both sciatic nerve block and intrathecal anesthesia, EC50 of lidocaine for tactile sensory blockade (2 g von Frey fiber) was significantly increased in HCN1 mice, whereas EC50 of lidocaine for pinprick blockade was unaffected. Durations of 1% lidocaine were significantly shorter in HCN1 mice for both sciatic nerve block and intrathecal anesthesia (n = 10). ZD7288 (HCN blocker) could significantly prolong durations of 1% lidocaine including pinprick blockade in sciatic nerve block (n = 10). Forskolin (raising cyclic adenosine monophosphate to enhance HCN2) could significantly shorten duration of pinprick blockade of 1% lidocaine in sciatic nerve block (n = 10). In electrophysiologic recordings, lidocaine could nonselectively inhibit HCN channel and sodium channel currents both in large and in small dorsal root ganglia neurons (n = 5 to 6). Meanwhile, lidocaine caused neural membrane hyperpolarization and increased input resistance of dorsal root ganglia neurons but not in large dorsal root ganglia neurons from HCN1 mice (n = 5-7). CONCLUSIONS These data indicate that HCN channels may contribute to regional anesthetic effects of lidocaine. By inhibiting HCN channels, lidocaine could alter membrane properties of neurons.
Collapse
Affiliation(s)
- Cheng Zhou
- From the Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu, China (C.Z., B.K., D.L., T.L., J.L., X.C.); Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China (Y.Z., P.L., J.L.); and Department of Anesthesiology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (X.C.)
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Verkerk AO, Wilders R. Pacemaker activity of the human sinoatrial node: an update on the effects of mutations in HCN4 on the hyperpolarization-activated current. Int J Mol Sci 2015; 16:3071-94. [PMID: 25642760 PMCID: PMC4346881 DOI: 10.3390/ijms16023071] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 01/22/2015] [Indexed: 11/22/2022] Open
Abstract
Since 2003, several loss-of-function mutations in the HCN4 gene, which encodes the HCN4 protein, have been associated with sinus node dysfunction. In human sinoatrial node (SAN), HCN4 is the most abundant of the four isoforms of the HCN family. Tetramers of HCN subunits constitute the ion channels that conduct the hyperpolarization-activated "funny" current (If), which plays an important modulating role in SAN pacemaker activity. Voltage-clamp experiments on HCN4 channels expressed in COS-7, CHO and HEK-293 cells, as well as in Xenopus oocytes have revealed changes in the expression and kinetics of mutant channels, but the extent to which especially the kinetic changes would affect If flowing during a human SAN action potential often remains unresolved. In our contribution to the Topical Collection on Human Single Nucleotide Polymorphisms and Disease Diagnostics, we provide an updated review of the mutation-induced changes in the expression and kinetics of HCN4 channels and provide an overview of their effects on If during the time course of a human SAN action potential, as assessed in simulated action potential clamp experiments. Future research may solve apparent inconsistencies between data from clinical studies and data from in vitro and in silico experiments.
Collapse
Affiliation(s)
- Arie O Verkerk
- Department of Anatomy, Embryology and Physiology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Ronald Wilders
- Department of Anatomy, Embryology and Physiology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| |
Collapse
|
31
|
Barbuti A, Robinson RB. Stem cell-derived nodal-like cardiomyocytes as a novel pharmacologic tool: insights from sinoatrial node development and function. Pharmacol Rev 2015; 67:368-88. [PMID: 25733770 DOI: 10.1124/pr.114.009597] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Since the first reports on the isolation and differentiation of stem cells, and in particular since the early success in driving these cells down a cardiac lineage, there has been interest in the potential of such preparations in cardiac regenerative therapy. Much of the focus of such research has been on improving mechanical function after myocardial infarction; however, electrophysiologic studies of these preparations have revealed a heterogeneous mix of action potential characteristics, including some described as "pacemaker" or "nodal-like," which in turn led to interest in the therapeutic potential of these preparations in the treatment of rhythm disorders; several proof-of-concept studies have used these cells to create a biologic alternative to electronic pacemakers. Further, there are additional potential applications of a preparation of pacemaker cells derived from stem cells, for example, in high-throughput screens of new chronotropic agents. All such applications require reasonably efficient methods for selecting or enriching the "nodal-like" cells, however, which in turn depends on first defining what constitutes a nodal-like cell since not all pacemaking cells are necessarily of nodal lineage. This review discusses the current state of the field in terms of characterizing sinoatrial-like cardiomyocytes derived from embryonic and induced pluripotent stem cells, markers that might be appropriate based on the current knowledge of the gene program leading to sinoatrial node development, what functional characteristics might be expected and desired based on studies of the sinoatrial node, and recent efforts at enrichment and selection of nodal-like cells.
Collapse
Affiliation(s)
- Andrea Barbuti
- Department of Biosciences, Università degli Studi di Milano, Milano, Italy (A.B.); and Department of Pharmacology, Columbia University Medical Center, New York, New York (R.B.R.)
| | - Richard B Robinson
- Department of Biosciences, Università degli Studi di Milano, Milano, Italy (A.B.); and Department of Pharmacology, Columbia University Medical Center, New York, New York (R.B.R.)
| |
Collapse
|
32
|
Severi S, Rodriguez B, Zaza A. Computational cardiac electrophysiology is ready for prime time. Europace 2014; 16:382-3. [DOI: 10.1093/europace/euu044] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|