1
|
Benndorf K, Enke U, Tewari D, Kusch J, Liu H, Sun H, Schmauder R, Sattler C. Subunit-specific conductance of single homomeric and heteromeric HCN pacemaker channels at femtosiemens resolution. Proc Natl Acad Sci U S A 2025; 122:e2422533122. [PMID: 39879240 PMCID: PMC11804576 DOI: 10.1073/pnas.2422533122] [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: 10/30/2024] [Accepted: 01/03/2025] [Indexed: 01/31/2025] Open
Abstract
In mammals, the four subunit isoforms HCN1-4 assemble to form functional homotetrameric and heterotetrameric hyperpolarization-activated cyclic nucleotide-modulated (HCN) ion channels. Despite the outstanding relevance of HCN channels for organisms, including generating electrical rhythmicity in cardiac pacemaker cells and diverse types of brain neurons, key channel properties are still elusive. In particular, the unitary conductance, γ, of HCN channels is highly controversial. We analyzed the unitary conductance at femtosiemens resolution of all four homotetrameric channels of the mouse, mHCN1-4. All conductance values are in the range of 1 pS which is exceptionally small compared to most other ion channels. Surprisingly, the conductance among the isoforms differs up to threefold (γmHCN2 = 1.54 pS > γmHCN1 = 0.84 pS > γmHCN3 = 0.54 pS ≈ γmHCN4 = 0.51 pS) though the residues in the two narrow parts of the pore, the selectivity filter and the inner gate, are conserved. Mutagenesis and all-atom molecular dynamics simulations demonstrate that the differences in the conductance are generated by different amounts of negative charges in the outer channel vestibule, which control ion accumulation. In line with these results, heterotetrameric channels exhibit intermediate unitary conductance values with respect to the homotetrameric channels. Our approach demonstrates how HCN channels can be functionally differentiated at the single-channel level, paving the way to target specific channels with selective drugs.
Collapse
Affiliation(s)
- Klaus Benndorf
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena07740, Germany
| | - Uta Enke
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena07740, Germany
| | - Debanjan Tewari
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena07740, Germany
| | - Jana Kusch
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena07740, Germany
| | - Haoran Liu
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin13125, Germany
- Institute of Chemistry, Technical University of Berlin, Berlin10623, Germany
| | - Han Sun
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin13125, Germany
- Institute of Chemistry, Technical University of Berlin, Berlin10623, Germany
| | - Ralf Schmauder
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena07740, Germany
| | - Christian Sattler
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena07740, Germany
| |
Collapse
|
2
|
Sharma S, Rana P, Chadha VD, Dhingra N, Kaur T. Exploring characteristic features for effective HCN1 channel inhibition using integrated analytical approaches: 3D QSAR, molecular docking, homology modelling, ADME and molecular dynamics. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2024; 53:447-464. [PMID: 39488633 DOI: 10.1007/s00249-024-01726-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/14/2024] [Accepted: 10/03/2024] [Indexed: 11/04/2024]
Abstract
Neuropathic pain (NP) is characterized by hyperalgesia, allodynia, and spontaneous pain. Hyperpolarization-activated cyclic nucleotide-gated (HCN) channel involved in neuronal hyperexcitability, has emerged as an important target for the drug development of NP. HCN channels exist in four different isoforms, where HCN1 is majorly expressed in dorsal root ganglion having an imperative role in NP pathophysiology. A specific HCN1 channel inhibitor will hold the better potential to treat NP without disturbing the physiological roles of other HCN isoforms. The main objective is to identify and analyze the chemical properties of scaffolds with higher HCN1 channel specificity. The 3D-QSAR studies highlight the hydrophobic & hydrogen bond donor groups enhance specificity towards the HCN1 channel. Further, the molecular interaction of the scaffolds with the HCN1 pore was studied by generating an open-pore model of the HCN1 channel using homology modelling and then docking the molecules with it. In addition, the important residues involved in the interaction between HCN1 pore and scaffolds were also identified. Moreover, ADME predictions revealed that compounds had good oral bioavailability and solubility characteristics. Subsequently, molecular dynamics simulation studies revealed the better stability of the lead molecules A7 and A9 during interactions and ascertained them as potential drug candidates. Cumulative studies provided the important structural features for enhancing HCN1 channel-specific inhibition, paving the way to design and develop novel specific HCN1 channel inhibitors.
Collapse
Affiliation(s)
- Shiwani Sharma
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | - Priyanka Rana
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India
| | | | - Neelima Dhingra
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India.
| | - Tanzeer Kaur
- Department of Biophysics, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
3
|
Saponaro A, Krumbach JH, Chaves-Sanjuan A, Sharifzadeh AS, Porro A, Castelli R, Hamacher K, Bolognesi M, DiFrancesco D, Clarke OB, Thiel G, Moroni A. Structural determinants of ivabradine block of the open pore of HCN4. Proc Natl Acad Sci U S A 2024; 121:e2402259121. [PMID: 38917012 PMCID: PMC11228525 DOI: 10.1073/pnas.2402259121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/26/2024] [Indexed: 06/27/2024] Open
Abstract
HCN1-4 channels are the molecular determinants of the If/Ih current that crucially regulates cardiac and neuronal cell excitability. HCN dysfunctions lead to sinoatrial block (HCN4), epilepsy (HCN1), and chronic pain (HCN2), widespread medical conditions awaiting subtype-specific treatments. Here, we address the problem by solving the cryo-EM structure of HCN4 in complex with ivabradine, to date the only HCN-specific drug on the market. Our data show ivabradine bound inside the open pore at 3 Å resolution. The structure unambiguously proves that Y507 and I511 on S6 are the molecular determinants of ivabradine binding to the inner cavity, while F510, pointing outside the pore, indirectly contributes to the block by controlling Y507. Cysteine 479, unique to the HCN selectivity filter (SF), accelerates the kinetics of block. Molecular dynamics simulations further reveal that ivabradine blocks the permeating ion inside the SF by electrostatic repulsion, a mechanism previously proposed for quaternary ammonium ions.
Collapse
Affiliation(s)
- Andrea Saponaro
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan 20133, Italy
| | - Jan H Krumbach
- Department of Physics, Technische Universität Darmstadt, Darmstadt 64289, Germany
- Department of Biology and Centre for Synthetic Biology, Technische Universität Darmstadt, Darmstadt 64287, Germany
| | | | | | - Alessandro Porro
- Department of Biosciences, University of Milan, Milan 20133, Italy
| | - Roberta Castelli
- Department of Biosciences, University of Milan, Milan 20133, Italy
| | - Kay Hamacher
- Department of Physics, Technische Universität Darmstadt, Darmstadt 64289, Germany
- Department of Biology and Centre for Synthetic Biology, Technische Universität Darmstadt, Darmstadt 64287, Germany
| | | | - Dario DiFrancesco
- Department of Biosciences, University of Milan, Milan 20133, Italy
- Institute of Biophysics-Milan, Consiglio Nazionale delle Ricerche, Milan 20133, Italy
| | - Oliver B Clarke
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY 10032
- Irving Institute for Clinical and Translational Research, Columbia University, New York, NY 10032
| | - Gerhard Thiel
- Department of Biology and Centre for Synthetic Biology, Technische Universität Darmstadt, Darmstadt 64287, Germany
- Department of Biosciences, University of Milan, Milan 20133, Italy
| | - Anna Moroni
- Department of Biosciences, University of Milan, Milan 20133, Italy
- Institute of Biophysics-Milan, Consiglio Nazionale delle Ricerche, Milan 20133, Italy
| |
Collapse
|
4
|
Page DA, Ruben PC. Cannabidiol potentiates hyperpolarization-activated cyclic nucleotide-gated (HCN4) channels. J Gen Physiol 2024; 156:e202313505. [PMID: 38652080 PMCID: PMC11040500 DOI: 10.1085/jgp.202313505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/15/2024] [Accepted: 04/09/2024] [Indexed: 04/25/2024] Open
Abstract
Cannabidiol (CBD), the main non-psychotropic phytocannabinoid produced by the Cannabis sativa plant, blocks a variety of cardiac ion channels. We aimed to identify whether CBD regulated the cardiac pacemaker channel or the hyperpolarization-activated cyclic nucleotide-gated channel (HCN4). HCN4 channels are important for the generation of the action potential in the sinoatrial node of the heart and increased heart rate in response to β-adrenergic stimulation. HCN4 channels were expressed in HEK 293T cells, and the effect of CBD application was examined using a whole-cell patch clamp. We found that CBD depolarized the V1/2 of activation in holo-HCN4 channels, with an EC50 of 1.6 µM, without changing the current density. CBD also sped activation kinetics by approximately threefold. CBD potentiation of HCN4 channels occurred via binding to the closed state of the channel. We found that CBD's mechanism of action was distinct from cAMP, as CBD also potentiated apo-HCN4 channels. The addition of an exogenous PIP2 analog did not alter the ability of CBD to potentiate HCN4 channels, suggesting that CBD also acts using a unique mechanism from the known HCN4 potentiator PIP2. Lastly, to gain insight into CBD's mechanism of action, computational modeling and targeted mutagenesis were used to predict that CBD binds to a lipid-binding pocket at the C-terminus of the voltage sensor. CBD represents the first FDA-approved drug to potentiate HCN4 channels, and our findings suggest a novel starting point for drug development targeting HCN4 channels.
Collapse
Affiliation(s)
- Dana A. Page
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
| | - Peter C. Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
| |
Collapse
|
5
|
Cho MS, Lee JH, Nam GB, Hwang KW, Cha MJ, Kim J, Choi KJ. Comparison between catheter ablation versus permanent pacemaker implantation as an initial treatment for tachycardia-bradycardia syndrome patients: a prospective, randomized trial. BMC Cardiovasc Disord 2024; 24:246. [PMID: 38730404 PMCID: PMC11088091 DOI: 10.1186/s12872-024-03920-0] [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: 08/23/2023] [Accepted: 05/03/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Clinical outcomes after catheter ablation (CA) or pacemaker (PM) implantation for the tachycardia-bradycardia syndrome (TBS) has not been evaluated adequately. We tried to compare the efficacy and safety outcomes of CA and PM implantation as an initial treatment option for TBS in paroxysmal atrial fibrillation (AF) patients. METHODS Sixty-eight patients with paroxysmal AF and TBS (mean 63.7 years, 63.2% male) were randomized, and received CA (n = 35) or PM (n = 33) as initial treatments. The primary outcomes were unexpected emergency room visits or hospitalizations attributed to cardiovascular causes. RESULTS In the intention-to-treatment analysis, the rates of primary outcomes were not significantly different between the two groups at the 2-year follow-up (19.8% vs. 25.9%; hazard ratio (HR) 0.73, 95% confidence interval (CI) 0.25-2.20, P = 0.584), irrespective of whether the results were adjusted for age (HR 1.12, 95% CI 0.34-3.64, P = 0.852). The 2-year rate of recurrent AF was significantly lower in the CA group compared to the PM group (33.9% vs. 56.8%, P = 0.038). Four patients (11.4%) in the CA group finally received PMs after CA owing to recurrent syncope episodes. The rate of major or minor procedure related complications was not significantly different between the two groups. CONCLUSION CA had a similar efficacy and safety profile with that of PM and a higher sinus rhythm maintenance rate. CA could be considered as a preferable initial treatment option over PM implantation in patients with paroxysmal AF and TBS. TRIAL REGISTRATION KCT0000155.
Collapse
Affiliation(s)
- Min Soo Cho
- Department of Cardiology, Heart Institute, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Ji Hyun Lee
- Cardiovascular Center, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Gi-Byoung Nam
- Department of Cardiology, Heart Institute, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea.
| | - Ki Won Hwang
- Division of Cardiology, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University of Medicine, Yangsan, Korea
| | - Myung-Jin Cha
- Department of Cardiology, Heart Institute, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Jun Kim
- Department of Cardiology, Heart Institute, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Kee-Joon Choi
- Department of Cardiology, Heart Institute, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| |
Collapse
|
6
|
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
|
7
|
Civil Ürkmez Y, Avcı B, Günaydın C, Çelik ZB, Ürkmez SS. Investigation of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels in vitro inflammation model at molecular level. Mol Cell Biochem 2024; 479:1223-1229. [PMID: 37432633 DOI: 10.1007/s11010-023-04788-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/12/2023] [Indexed: 07/12/2023]
Abstract
In our study, we aimed to create an inflammation model in endothelial and macrophage cell lines and to examine the changes in the expression of hyperpolarization activated cyclic nucleotide gated (HCN) channels at the molecular level. HUVEC and RAW cell lines were used in our study. 1 µg/mL LPS was applied to the cells. Cell media were taken 6 h later. TNF-α, IL-1, IL-2, IL-4, IL-10 concentrations were measured by ELISA method. Cell media were cross-applied to cells for 24 h after LPS. HCN1/HCN2 protein levels were determined by Western-Blot method. HCN-1/HCN-2 gene expressions were determined by qRT-PCR method. In the inflammation model, a significant increase in TNF-α, IL-1, and IL-2 levels was observed in RAW cell media compared to the control. While no significant difference was observed in IL-4 level, a significant decrease was observed in IL-10 level. While a significant increase in TNF-α level was observed in HUVEC cell medium, no difference was observed in other cytokines. In our inflammation model, an 8.44-fold increase in HCN1 gene expression was observed in HUVEC cells compared to the control group. No significant change was observed in HCN2 gene expression. 6.71-fold increase in HCN1 gene expression was observed in RAW cells compared to the control. The change in HCN2 expression was not statistically significant. In the Western-Blot analysis, a statistically significant increase in HCN1 level was observed in the LPS group in HUVEC cells compared to the control; no significant increase in HCN2 level was observed. While a statistically significant increase in HCN1 level was observed in the LPS group in RAW cells compared to the control; no significant increase in HCN2 level was observed. In immunofluorescence examination, it was observed that the level of HCN1 and HCN2 proteins in the cell membrane of HUVEC and RAW cells increased in the LPS group compared to the control group. While HCN1 gene/protein levels were increased in RAW and HUVEC cells in the inflammation model, no significant change was observed in HCN2 gene/protein levels. Our data suggest that the HCN1 subtype is dominant in endothelium and macrophages and may play a critical role in inflammation.
Collapse
Affiliation(s)
- Yeşim Civil Ürkmez
- Department of Biochemistry, Samsun Training and Research Hospital, University of Health Sciences, Samsun, Turkey.
| | - Bahattin Avcı
- Department of Biochemistry, School of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Caner Günaydın
- Department of Pharmacology, School of Medicine, Samsun University, Samsun, Turkey
| | - Zülfinaz Betül Çelik
- Department of Medical Biology, School of Medicine, Samsun University, Samsun, Turkey
| | - Sebati Sinan Ürkmez
- Department of Biochemistry, School of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| |
Collapse
|
8
|
Benndorf K, DiFrancesco D. Missing validation of key features in HCN single-channel recordings. Proc Natl Acad Sci U S A 2024; 121:e2400523121. [PMID: 38588429 PMCID: PMC11032480 DOI: 10.1073/pnas.2400523121] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024] Open
Affiliation(s)
- Klaus Benndorf
- Institute of Physiology II, Jena University Hospital, Jena07743, Germany
| | - Dario DiFrancesco
- Department of Biosciences, The PaceLab, University of Milano, Milano20133, Italy
| |
Collapse
|
9
|
Harde E, Hierl M, Weber M, Waiz D, Wyler R, Wach JY, Haab R, Gundlfinger A, He W, Schnider P, Paina M, Rolland JF, Greiter-Wilke A, Gasser R, Reutlinger M, Dupont A, Roberts S, O'Connor EC, Bartels B, Hall BJ. Selective and brain-penetrant HCN1 inhibitors reveal links between synaptic integration, cortical function, and working memory. Cell Chem Biol 2024; 31:577-592.e23. [PMID: 38042151 DOI: 10.1016/j.chembiol.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/28/2023] [Accepted: 11/07/2023] [Indexed: 12/04/2023]
Abstract
Hyperpolarization-activated and cyclic-nucleotide-gated 1 (HCN1) ion channels are proposed to be critical for cognitive function through regulation of synaptic integration. However, resolving the precise role of HCN1 in neurophysiology and exploiting its therapeutic potential has been hampered by minimally selective antagonists with poor potency and limited in vivo efficiency. Using automated electrophysiology in a small-molecule library screen and chemical optimization, we identified a primary carboxamide series of potent and selective HCN1 inhibitors with a distinct mode of action. In cognition-relevant brain circuits, selective inhibition of native HCN1 produced on-target effects, including enhanced excitatory postsynaptic potential summation, while administration of a selective HCN1 inhibitor to rats recovered decrement working memory. Unlike prior non-selective HCN antagonists, selective HCN1 inhibition did not alter cardiac physiology in human atrial cardiomyocytes or in rats. Collectively, selective HCN1 inhibitors described herein unmask HCN1 as a potential target for the treatment of cognitive dysfunction in brain disorders.
Collapse
Affiliation(s)
- Eva Harde
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| | - Markus Hierl
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Michael Weber
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - David Waiz
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Roger Wyler
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Jean-Yves Wach
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Rachel Haab
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Anja Gundlfinger
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Weiping He
- WuXi AppTec (Wuhan) Co., Ltd, 666 Gaoxin Road, Wuhan East Lake High-Tech Development Zone, Wuhan, Huibei, China
| | - Patrick Schnider
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | | | - Andrea Greiter-Wilke
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Rodolfo Gasser
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Michael Reutlinger
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Amanda Dupont
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Sonia Roberts
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Eoin C O'Connor
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| | - Björn Bartels
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| | - Benjamin J Hall
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| |
Collapse
|
10
|
Cámara-Checa A, Perin F, Rubio-Alarcón M, Dago M, Crespo-García T, Rapún J, Marín M, Cebrián J, Gómez R, Bermúdez-Jiménez F, Monserrat L, Tamargo J, Caballero R, Jiménez-Jáimez J, Delpón E. A gain-of-function HCN4 mutant in the HCN domain is responsible for inappropriate sinus tachycardia in a Spanish family. Proc Natl Acad Sci U S A 2023; 120:e2305135120. [PMID: 38032931 PMCID: PMC10710060 DOI: 10.1073/pnas.2305135120] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 10/12/2023] [Indexed: 12/02/2023] Open
Abstract
In a family with inappropriate sinus tachycardia (IST), we identified a mutation (p.V240M) of the hyperpolarization-activated cyclic nucleotide-gated type 4 (HCN4) channel, which contributes to the pacemaker current (If) in human sinoatrial node cells. Here, we clinically study fifteen family members and functionally analyze the p.V240M variant. Macroscopic (IHCN4) and single-channel currents were recorded using patch-clamp in cells expressing human native (WT) and/or p.V240M HCN4 channels. All p.V240M mutation carriers exhibited IST that was accompanied by cardiomyopathy in adults. IHCN4 generated by p.V240M channels either alone or in combination with WT was significantly greater than that generated by WT channels alone. The variant, which lies in the N-terminal HCN domain, increased the single-channel conductance and opening frequency and probability of HCN4 channels. Conversely, it did not modify the channel sensitivity for cAMP and ivabradine or the level of expression at the membrane. Treatment with ivabradine based on functional data reversed the IST and the cardiomyopathy of the carriers. In computer simulations, the p.V240M gain-of-function variant increases If and beating rate and thus explains the IST of the carriers. The results demonstrate the importance of the unique HCN domain in HCN4, which stabilizes the channels in the closed state.
Collapse
Affiliation(s)
- Anabel Cámara-Checa
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Francesca Perin
- Department of Pediatric Cardiology, Virgen de las Nieves University Hospital, Granada18014, Spain
- Instituto de Investigación Biosanitaria de Granada, Granada18014, Spain
| | - Marcos Rubio-Alarcón
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - María Dago
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Teresa Crespo-García
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Josu Rapún
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - María Marín
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Jorge Cebrián
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Ricardo Gómez
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Francisco Bermúdez-Jiménez
- Department of Pediatric Cardiology, Virgen de las Nieves University Hospital, Granada18014, Spain
- Instituto de Investigación Biosanitaria de Granada, Granada18014, Spain
- Centro Nacional de Investigaciones Cardiovasculares, Madrid28029, Spain
| | - Lorenzo Monserrat
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
- Health in Code Sociedad Limitada, A Coruña15008, Spain
| | - Juan Tamargo
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
| | - Ricardo Caballero
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Juan Jiménez-Jáimez
- Department of Pediatric Cardiology, Virgen de las Nieves University Hospital, Granada18014, Spain
- Instituto de Investigación Biosanitaria de Granada, Granada18014, Spain
| | - Eva Delpón
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| |
Collapse
|
11
|
Schunke KJ, Rodriguez J, Dyavanapalli J, Schloen J, Wang X, Escobar J, Kowalik G, Cheung EC, Ribeiro C, Russo R, Alber BR, Dergacheva O, Chen SW, Murillo-Berlioz AE, Lee KB, Trachiotis G, Entcheva E, Brantner CA, Mendelowitz D, Kay MW. Outcomes of hypothalamic oxytocin neuron-driven cardioprotection after acute myocardial infarction. Basic Res Cardiol 2023; 118:43. [PMID: 37801130 PMCID: PMC10558415 DOI: 10.1007/s00395-023-01013-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/07/2023]
Abstract
Altered autonomic balance is a hallmark of numerous cardiovascular diseases, including myocardial infarction (MI). Although device-based vagal stimulation is cardioprotective during chronic disease, a non-invasive approach to selectively stimulate the cardiac parasympathetic system immediately after an infarction does not exist and is desperately needed. Cardiac vagal neurons (CVNs) in the brainstem receive powerful excitation from a population of neurons in the paraventricular nucleus (PVN) of the hypothalamus that co-release oxytocin (OXT) and glutamate to excite CVNs. We tested if chemogenetic activation of PVN-OXT neurons following MI would be cardioprotective. The PVN of neonatal rats was transfected with vectors to selectively express DREADDs within OXT neurons. At 6 weeks of age, an MI was induced and DREADDs were activated with clozapine-N-oxide. Seven days following MI, patch-clamp electrophysiology confirmed the augmented excitatory neurotransmission from PVN-OXT neurons to downstream nuclei critical for parasympathetic activity with treatment (43.7 ± 10 vs 86.9 ± 9 pA; MI vs. treatment), resulting in stark improvements in survival (85% vs. 95%; MI vs. treatment), inflammation, fibrosis assessed by trichrome blue staining, mitochondrial function assessed by Seahorse assays, and reduced incidence of arrhythmias (50% vs. 10% cumulative incidence of ventricular fibrillation; MI vs. treatment). Myocardial transcriptomic analysis provided molecular insight into potential cardioprotective mechanisms, which revealed the preservation of beneficial signaling pathways, including muscarinic receptor activation, in treated animals. These comprehensive results demonstrate that the PVN-OXT network could be a promising therapeutic target to quickly activate beneficial parasympathetic-mediated cellular pathways within the heart during the early stages of infarction.
Collapse
Affiliation(s)
- Kathryn J Schunke
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA.
- Department of Anatomy, Biochemistry and Physiology, University of Hawaii, 651 Ilalo St, Honolulu, HI, BSB 211 96813, USA.
| | - Jeannette Rodriguez
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Jhansi Dyavanapalli
- Department of Pharmacology and Physiology, George Washington University, Suite 640 Ross Hall, 2300 Eye St. NW, Washington, DC, 20052, USA
| | - John Schloen
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Xin Wang
- Department of Pharmacology and Physiology, George Washington University, Suite 640 Ross Hall, 2300 Eye St. NW, Washington, DC, 20052, USA
| | - Joan Escobar
- Department of Pharmacology and Physiology, George Washington University, Suite 640 Ross Hall, 2300 Eye St. NW, Washington, DC, 20052, USA
| | - Grant Kowalik
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Emily C Cheung
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Caitlin Ribeiro
- Department of Pharmacology and Physiology, George Washington University, Suite 640 Ross Hall, 2300 Eye St. NW, Washington, DC, 20052, USA
| | - Rebekah Russo
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Bridget R Alber
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Olga Dergacheva
- Department of Pharmacology and Physiology, George Washington University, Suite 640 Ross Hall, 2300 Eye St. NW, Washington, DC, 20052, USA
| | - Sheena W Chen
- Division of Cardiothoracic Surgery and Cardiothoracic Research, Veterans Affairs Medical Center, 50 Irving St. NW, Washington, DC, 20422, USA
| | - Alejandro E Murillo-Berlioz
- Division of Cardiothoracic Surgery and Cardiothoracic Research, Veterans Affairs Medical Center, 50 Irving St. NW, Washington, DC, 20422, USA
| | - Kyongjune B Lee
- Division of Cardiothoracic Surgery and Cardiothoracic Research, Veterans Affairs Medical Center, 50 Irving St. NW, Washington, DC, 20422, USA
| | - Gregory Trachiotis
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
- Division of Cardiothoracic Surgery and Cardiothoracic Research, Veterans Affairs Medical Center, 50 Irving St. NW, Washington, DC, 20422, USA
| | - Emilia Entcheva
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Christine A Brantner
- The GWU Nanofabrication and Imaging Center, 800 22nd Street NW, Washington, DC, 20052, USA
| | - David Mendelowitz
- Department of Pharmacology and Physiology, George Washington University, Suite 640 Ross Hall, 2300 Eye St. NW, Washington, DC, 20052, USA.
| | - Matthew W Kay
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA.
| |
Collapse
|
12
|
Tibbs GR, Uprety R, Warren JD, Beyer NP, Joyce RL, Ferrer MA, Mellado W, Wong VSC, Goldberg DC, Cohen MW, Costa CJ, Li Z, Zhang G, Dephoure NE, Barman DN, Sun D, Ingólfsson HI, Sauve AA, Willis DE, Goldstein PA. An anchor-tether 'hindered' HCN1 inhibitor is antihyperalgesic in a rat spared nerve injury neuropathic pain model. Br J Anaesth 2023; 131:745-763. [PMID: 37567808 PMCID: PMC10541997 DOI: 10.1016/j.bja.2023.06.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/20/2023] [Accepted: 06/29/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Neuropathic pain impairs quality of life, is widely prevalent, and incurs significant costs. Current pharmacological therapies have poor/no efficacy and significant adverse effects; safe and effective alternatives are needed. Hyperpolarisation-activated cyclic nucleotide-regulated (HCN) channels are causally implicated in some forms of peripherally mediated neuropathic pain. Whilst 2,6-substituted phenols, such as 2,6-di-tert-butylphenol (26DTB-P), selectively inhibit HCN1 gating and are antihyperalgesic, the development of therapeutically tolerable, HCN-selective antihyperalgesics based on their inverse agonist activity requires that such drugs spare the cardiac isoforms and do not cross the blood-brain barrier. METHODS In silico molecular dynamics simulation, in vitro electrophysiology, and in vivo rat spared nerve injury methods were used to test whether 'hindered' variants of 26DTB-P (wherein a hydrophilic 'anchor' is attached in the para-position of 26DTB-P via an acyl chain 'tether') had the desired properties. RESULTS Molecular dynamics simulation showed that membrane penetration of hindered 26DTB-Ps is controlled by a tethered diol anchor without elimination of head group rotational freedom. In vitro and in vivo analysis showed that BP4L-18:1:1, a variant wherein a diol anchor is attached to 26DTB-P via an 18-carbon tether, is an HCN1 inverse agonist and an orally available antihyperalgesic. With a CNS multiparameter optimisation score of 2.25, a >100-fold lower drug load in the brain vs blood, and an absence of adverse cardiovascular or CNS effects, BP4L-18:1:1 was shown to be poorly CNS penetrant and cardiac sparing. CONCLUSIONS These findings provide a proof-of-concept demonstration that anchor-tethered drugs are a new chemotype for treatment of disorders involving membrane targets.
Collapse
Affiliation(s)
- Gareth R Tibbs
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Rajendra Uprety
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - J David Warren
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Nicole P Beyer
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Rebecca L Joyce
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Matthew A Ferrer
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | | | | | | | | | | | - Zhucui Li
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Guoan Zhang
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Noah E Dephoure
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Dipti N Barman
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Delin Sun
- Lawrence Livermore National Laboratory, Livermore, CA, USA
| | | | - Anthony A Sauve
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Dianna E Willis
- Burke Neurological Institute, White Plains, NY, USA; Feil Family Brain & Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| | - Peter A Goldstein
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA; Feil Family Brain & Mind Research Institute, Weill Cornell Medicine, New York, NY, USA; Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
13
|
Leiva O, Bohart I, Ahuja T, Park D. Off-Target Effects of Cancer Therapy on Development of Therapy-Induced Arrhythmia: A Review. Cardiology 2023; 148:324-334. [PMID: 36702116 PMCID: PMC10614257 DOI: 10.1159/000529260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/17/2023] [Indexed: 01/27/2023]
Abstract
BACKGROUND Advances in cancer therapeutics have improved overall survival and prognosis in this patient population; however, this has come at the expense of cardiotoxicity including arrhythmia. SUMMARY Cancer and its therapies are associated with cardiotoxicity via several mechanisms including inflammation, cardiomyopathy, and off-target effects. Among cancer therapies, anthracyclines and tyrosine kinase inhibitors (TKIs) are particularly known for their pro-arrhythmia effects. In addition to cardiomyopathy, anthracyclines may be pro-arrhythmogenic via reactive oxygen species (ROS) generation and altered calcium handling. TKIs may mediate their cardiotoxicity via inhibition of off-target tyrosine kinases. Ibrutinib-mediated inhibition of CSK may be responsible for the increased prevalence of atrial fibrillation. Further investigation is warranted to further elucidate the mechanisms behind arrhythmias in cancer therapies. KEY MESSAGES Arrhythmias are a common cardiotoxicity of cancer therapies. Cancer therapies may induce arrhythmias via off-target effects. Understanding the mechanisms underlying arrhythmogenesis associated with cancer therapies may help design cancer therapies that can avoid these toxicities.
Collapse
Affiliation(s)
- Orly Leiva
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| | - Isaac Bohart
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| | - Tania Ahuja
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| | - David Park
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| |
Collapse
|
14
|
Barbuti A, Baruscotti M, Bucchi A. The “Funny” Pacemaker Current. HEART RATE AND RHYTHM 2023:63-87. [DOI: 10.1007/978-3-031-33588-4_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
15
|
Ricci E, Bartolucci C, Severi S. The virtual sinoatrial node: What did computational models tell us about cardiac pacemaking? PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 177:55-79. [PMID: 36374743 DOI: 10.1016/j.pbiomolbio.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 10/17/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022]
Abstract
Since its discovery, the sinoatrial node (SAN) has represented a fascinating and complex matter of research. Despite over a century of discoveries, a full comprehension of pacemaking has still to be achieved. Experiments often produced conflicting evidence that was used either in support or against alternative theories, originating intense debates. In this context, mathematical descriptions of the phenomena underlying the heartbeat have grown in importance in the last decades since they helped in gaining insights where experimental evaluation could not reach. This review presents the most updated SAN computational models and discusses their contribution to our understanding of cardiac pacemaking. Electrophysiological, structural and pathological aspects - as well as the autonomic control over the SAN - are taken into consideration to reach a holistic view of SAN activity.
Collapse
Affiliation(s)
- Eugenio Ricci
- Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, Cesena (FC), Italy
| | - Chiara Bartolucci
- Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, Cesena (FC), Italy
| | - Stefano Severi
- Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, Cesena (FC), Italy.
| |
Collapse
|
16
|
Prodan N, Ershad F, Reyes-Alcaraz A, Li L, Mistretta B, Gonzalez L, Rao Z, Yu C, Gunaratne PH, Li N, Schwartz RJ, McConnell BK. Direct reprogramming of cardiomyocytes into cardiac Purkinje-like cells. iScience 2022; 25:105402. [PMID: 36388958 PMCID: PMC9646947 DOI: 10.1016/j.isci.2022.105402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 09/30/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022] Open
Abstract
Currently, there are no treatments that ameliorate cardiac cell death, the underlying basis of cardiovascular disease. An unexplored cell type in cardiac regeneration is cardiac Purkinje cells; specialized cells from the cardiac conduction system (CCS) responsible for propagating electrical signals. Purkinje cells have tremendous potential as a regenerative treatment because they may intrinsically integrate with the CCS of a recipient myocardium, resulting in more efficient electrical conduction in diseased hearts. This study is the first to demonstrate an effective protocol for the direct reprogramming of human cardiomyocytes into cardiac Purkinje-like cells using small molecules. The cells generated were genetically and functionally similar to native cardiac Purkinje cells, where expression of key cardiac Purkinje genes such as CNTN2, ETV1, PCP4, IRX3, SCN5a, HCN2 and the conduction of electrical signals with increased velocity was observed. This study may help to advance the quest to finding an optimized cell therapy for heart regeneration.
Collapse
Affiliation(s)
- Nicole Prodan
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 4349 Martin Luther King Blvd, Health-2 (H2) Building, Room 5024, Houston, TX 77204-5037, USA
| | - Faheem Ershad
- Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, TX 77204, USA
| | - Arfaxad Reyes-Alcaraz
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 4349 Martin Luther King Blvd, Health-2 (H2) Building, Room 5024, Houston, TX 77204-5037, USA
| | - Luge Li
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX 77030, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Brandon Mistretta
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
- Department of Biology and Biochemistry, UH-Sequencing & Gene Editing Core, University of Houston, Houston, TX 77204, USA
| | - Lei Gonzalez
- Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, TX 77204, USA
| | - Zhoulyu Rao
- Department of Mechanical Engineering, Cullen College of Engineering, University of Houston, Houston, TX 77204, USA
| | - Cunjiang Yu
- Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, TX 77204, USA
- Department of Mechanical Engineering, Cullen College of Engineering, University of Houston, Houston, TX 77204, USA
| | - Preethi H. Gunaratne
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
- Department of Biology and Biochemistry, UH-Sequencing & Gene Editing Core, University of Houston, Houston, TX 77204, USA
| | - Na Li
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX 77030, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Robert J. Schwartz
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Bradley K. McConnell
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 4349 Martin Luther King Blvd, Health-2 (H2) Building, Room 5024, Houston, TX 77204-5037, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
17
|
Farraha M, Rao R, Igoor S, Le TYL, Barry MA, Davey C, Kok C, Chong JJ, Kizana E. Recombinant Adeno-Associated Viral Vector-Mediated Gene Transfer of hTBX18 Generates Pacemaker Cells from Ventricular Cardiomyocytes. Int J Mol Sci 2022; 23:ijms23169230. [PMID: 36012498 PMCID: PMC9408910 DOI: 10.3390/ijms23169230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 12/03/2022] Open
Abstract
Sinoatrial node dysfunction can manifest as bradycardia, leading to symptoms of syncope and sudden cardiac death. Electronic pacemakers are the current standard of care but are limited due to a lack of biological chronotropic control, cost of revision surgeries, and risk of lead- and device-related complications. We therefore aimed to develop a biological alternative to electronic devices by using a clinically relevant gene therapy vector to demonstrate conversion of cardiomyocytes into sinoatrial node-like cells in an in vitro context. Neonatal rat ventricular myocytes were transduced with recombinant adeno-associated virus vector 6 encoding either hTBX18 or green fluorescent protein and maintained for 3 weeks. At the endpoint, qPCR, Western blot analysis and immunocytochemistry were used to assess for reprogramming into pacemaker cells. Cell morphology and Arclight action potentials were imaged via confocal microscopy. Compared to GFP, hTBX18-transduced cells showed that hTBX18, HCN4 and Cx45 were upregulated. Cx43 was significantly downregulated, while sarcomeric α-actinin remained unchanged. Cardiomyocytes transduced with hTBX18 acquired the tapering morphology of native pacemaker cells, as compared to the block-like, striated appearance of ventricular cardiomyocytes. Analysis of the action potentials showed phase 4 depolarization and a significant decrease in the APD50 of the hTBX18-transduced cells. We have demonstrated that rAAV-hTBX18 gene transfer to ventricular myocytes results in morphological, molecular, physiological, and functional changes, recapitulating the pacemaker phenotype in an in vitro setting. The generation of these induced pacemaker-like cells using a clinically relevant vector opens new prospects for biological pacemaker development.
Collapse
Affiliation(s)
- Melad Farraha
- Sydney Medical School, the University of Sydney, Sydney 2006, Australia
- Centre for Heart Research, the Westmead Institute for Medical Research, Sydney 2145, Australia
| | - Renuka Rao
- Centre for Heart Research, the Westmead Institute for Medical Research, Sydney 2145, Australia
| | - Sindhu Igoor
- Centre for Heart Research, the Westmead Institute for Medical Research, Sydney 2145, Australia
| | - Thi Y. L. Le
- Centre for Heart Research, the Westmead Institute for Medical Research, Sydney 2145, Australia
| | - Michael A. Barry
- Department of Cardiology, Westmead Hospital, Sydney 2145, Australia
| | - Christopher Davey
- Centre for Heart Research, the Westmead Institute for Medical Research, Sydney 2145, Australia
- School of Physics, the University of Sydney, Sydney 2006, Australia
| | - Cindy Kok
- Centre for Heart Research, the Westmead Institute for Medical Research, Sydney 2145, Australia
| | - James J.H. Chong
- Sydney Medical School, the University of Sydney, Sydney 2006, Australia
- Centre for Heart Research, the Westmead Institute for Medical Research, Sydney 2145, Australia
- Department of Cardiology, Westmead Hospital, Sydney 2145, Australia
| | - Eddy Kizana
- Sydney Medical School, the University of Sydney, Sydney 2006, Australia
- Centre for Heart Research, the Westmead Institute for Medical Research, Sydney 2145, Australia
- Department of Cardiology, Westmead Hospital, Sydney 2145, Australia
- Correspondence:
| |
Collapse
|
18
|
Dixon RE, Navedo MF, Binder MD, Santana LF. Mechanisms and Physiological Implications of Cooperative Gating of Ion Channels Clusters. Physiol Rev 2021; 102:1159-1210. [PMID: 34927454 DOI: 10.1152/physrev.00022.2021] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Ion channels play a central role in the regulation of nearly every cellular process. Dating back to the classic 1952 Hodgkin-Huxley model of the generation of the action potential, ion channels have always been thought of as independent agents. A myriad of recent experimental findings exploiting advances in electrophysiology, structural biology, and imaging techniques, however, have posed a serious challenge to this long-held axiom as several classes of ion channels appear to open and close in a coordinated, cooperative manner. Ion channel cooperativity ranges from variable-sized oligomeric cooperative gating in voltage-gated, dihydropyridine-sensitive Cav1.2 and Cav1.3 channels to obligatory dimeric assembly and gating of voltage-gated Nav1.5 channels. Potassium channels, transient receptor potential channels, hyperpolarization cyclic nucleotide-activated channels, ryanodine receptors (RyRs), and inositol trisphosphate receptors (IP3Rs) have also been shown to gate cooperatively. The implications of cooperative gating of these ion channels range from fine tuning excitation-contraction coupling in muscle cells to regulating cardiac function and vascular tone, to modulation of action potential and conduction velocity in neurons and cardiac cells, and to control of pace-making activity in the heart. In this review, we discuss the mechanisms leading to cooperative gating of ion channels, their physiological consequences and how alterations in cooperative gating of ion channels may induce a range of clinically significant pathologies.
Collapse
Affiliation(s)
- Rose Ellen Dixon
- Department of Physiology and Membrane Biology, University of California, Davis, CA, United States
| | - Manuel F Navedo
- Department of Pharmacology, University of California, Davis, CA, United States
| | - Marc D Binder
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States
| | - L Fernando Santana
- Department of Physiology and Membrane Biology, University of California, Davis, CA, United States
| |
Collapse
|
19
|
Aydin A, Gayretli Yayla K. The assessment of Tp-e interval and Tp-e/QT ratio in patients with hyperthyroidism before and after thyroid surgery. Int J Clin Pract 2021; 75:e14937. [PMID: 34606148 DOI: 10.1111/ijcp.14937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 10/01/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Ventricular repolarisation is assessed using the Tp-e interval and QT interval corrected by the heart rate (QTc) via an electrocardiogram (ECG). Prolonged Tp-e/QTc is related with an increased risk of arrhythmias and cardiac mortality. OBJECTIVE This study was conducted at a single centre in collaboration with general surgery and cardiology clinics. We aimed to appraise the assessment of Tp-e interval and Tp-e/QT ratio in patients with hyperthyroidism before and after thyroid surgery. METHODS Totally 65 patients with hyperthyroidism before and after thyroid surgery were enrolled in our study. In presurgical hospitalisation and six months after the thyroid surgery, we measured the electrocardiographic parameters again on same patients. All subjects who were investigated in this study were in sinus rhythm. Tp-e interval, Tp-e/QT and Tp-e/QTc ratios were measured from the 12-lead electrocardiogram. RESULTS Heart rate (P = .073), QT interval (P = .432) and QTc interval (P = .179) were similar before and after thyroid surgery. Tp-e interval (84.6 ± 13.1 vs 77.2 ± 10.9; P = .031), Tp-e/QT ratio (0.23 ± 0.04 vs 0.21 ± 0.04; P < .001), Tp-e/QTc ratio (0.21 ± 0.04 vs 0.19 ± 0.03; P < .001) and QTc dispersion (52.4 ± 7.2 vs 48.4 ± 7.4; P < .001) were significantly different before and after thyroid surgery. In correlation analysis, there was a significant correlation between preprocedural Tp-e/QTc ratio and preprocedural fT4 in patients with hyperthyroidism (r = 0.275, P = .026). CONCLUSION Our study demonstrated that Tp-e interval, Tp-e/QT and Tp-e/QTc ratios were shortened in patients with hyperthyroidism after thyroid surgery than before procedure. This study is considerable to display that hyperthyroidism may have a negative effect on cardiac conduction system, which potentially may induce formation of ventricular arrhythmias.
Collapse
Affiliation(s)
- Altan Aydin
- Department of General Surgery, Health Sciences University, Kanuni Education and Research Hospital, Trabzon, Turkey
| | - Kadriye Gayretli Yayla
- Department of Cardiology, Health Sciences University, Dr. Abdurrahman Yurtarslan Oncology Education and Research Hospital, Ankara, Turkey
| |
Collapse
|
20
|
Yu H, Gall B, Newman M, Hathaway Q, Brundage K, Ammer A, Mathers P, Siderovski D, Hull RW. Contribution of HCN1 variant to sinus bradycardia: A case report. J Arrhythm 2021; 37:1337-1347. [PMID: 34621433 PMCID: PMC8485797 DOI: 10.1002/joa3.12598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 06/08/2021] [Accepted: 06/26/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Missense mutations in the hyperpolarization-activated cyclic nucleotide-modulated (HCN) channel 4 (HCN4) are one of the genetic causes of cardiac sinus bradycardia. OBJECTIVE To investigate possible HCN4 channel mutation in a young patient with profound sinus bradycardia. METHODS Direct sequencing of HCN4 and whole-exome sequencing were performed on DNA samples from the indexed patient (P), the patient's son (PS), and a family unrelated healthy long-distance running volunteer (V). Resting heart rate was 31 bpm for P, 67 bpm for PS, and 50 bpm for V. Immunoblots, flow cytometry, and immunocytofluorescence confocal imaging were used to study cellular distribution of channel variants. Patch-clamp electrophysiology was used to investigate the properties of mutant HCN1 channels. RESULTS In P no missense mutations were found in the HCN4 gene; instead, we found two heterozygous variants in the HCN1 gene: deletion of an N-terminal glycine triplet (72GGG74, "N-del") and a novel missense variant, P851A, in the C-terminal region. N-del variant was found before and shared by PS. These two variations were not found in V. Compared to wild type, N-del and P851A reduced cell surface expression and negatively shifted voltage-activation with slower activation kinetics. CONCLUSION Decreased channel activity HCN1 mutant channel makes it unable to contribute to early depolarization of sinus node action potential, thus likely a main cause of the profound sinus bradycardia in this patient.
Collapse
Affiliation(s)
- Hangang Yu
- Department of Physiology and PharmacologySchool of MedicineWest Virginia UniversityMorgantownWVUSA
| | - Bryan Gall
- Department of Physiology and PharmacologySchool of MedicineWest Virginia UniversityMorgantownWVUSA
- Present address:
Variant Curator at NateraSan CarlosCAUSA
| | - Mackenzie Newman
- Department of Physiology and PharmacologySchool of MedicineWest Virginia UniversityMorgantownWVUSA
| | - Quincy Hathaway
- Department of Exercise PhysiologySchool of MedicineWest Virginia UniversityMorgantownWVUSA
| | - Kathleen Brundage
- Department of Microbiology, Immunology & Cell BiologySchool of MedicineWest Virginia UniversityMorgantownWVUSA
| | - Amanda Ammer
- Department of Microbiology, Immunology & Cell BiologySchool of MedicineWest Virginia UniversityMorgantownWVUSA
| | - Peter Mathers
- Department of NeuroscienceSchool of MedicineWest Virginia UniversityMorgantownWVUSA
| | - David Siderovski
- Department of Physiology and PharmacologySchool of MedicineWest Virginia UniversityMorgantownWVUSA
- Present address:
Pharmacology & NeuroscienceUniversity of North TexasDentonTXUSA
| | - Robert W. Hull
- Department of CardiologySchool of MedicineWest Virginia UniversityMorgantownWVUSA
- Present address:
Department of CardiologyMon General HospitalMorgantownWVUSA
| |
Collapse
|
21
|
Saponaro A, Bauer D, Giese MH, Swuec P, Porro A, Gasparri F, Sharifzadeh AS, Chaves-Sanjuan A, Alberio L, Parisi G, Cerutti G, Clarke OB, Hamacher K, Colecraft HM, Mancia F, Hendrickson WA, Siegelbaum SA, DiFrancesco D, Bolognesi M, Thiel G, Santoro B, Moroni A. Gating movements and ion permeation in HCN4 pacemaker channels. Mol Cell 2021; 81:2929-2943.e6. [PMID: 34166608 PMCID: PMC8294335 DOI: 10.1016/j.molcel.2021.05.033] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/12/2021] [Accepted: 05/27/2021] [Indexed: 10/31/2022]
Abstract
The HCN1-4 channel family is responsible for the hyperpolarization-activated cation current If/Ih that controls automaticity in cardiac and neuronal pacemaker cells. We present cryoelectron microscopy (cryo-EM) structures of HCN4 in the presence or absence of bound cAMP, displaying the pore domain in closed and open conformations. Analysis of cAMP-bound and -unbound structures sheds light on how ligand-induced transitions in the channel cytosolic portion mediate the effect of cAMP on channel gating and highlights the regulatory role of a Mg2+ coordination site formed between the C-linker and the S4-S5 linker. Comparison of open/closed pore states shows that the cytosolic gate opens through concerted movements of the S5 and S6 transmembrane helices. Furthermore, in combination with molecular dynamics analyses, the open pore structures provide insights into the mechanisms of K+/Na+ permeation. Our results contribute mechanistic understanding on HCN channel gating, cyclic nucleotide-dependent modulation, and ion permeation.
Collapse
Affiliation(s)
- Andrea Saponaro
- Department of Biosciences, University of Milan, Milan, Italy
| | - Daniel Bauer
- Department of Biology, TU-Darmstadt, Darmstadt, Germany
| | - M Hunter Giese
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Paolo Swuec
- Department of Biosciences, University of Milan, Milan, Italy; Pediatric Research Center "Romeo ed Enrica Invernizzi," University of Milan, Milan, Italy
| | | | | | | | - Antonio Chaves-Sanjuan
- Department of Biosciences, University of Milan, Milan, Italy; Pediatric Research Center "Romeo ed Enrica Invernizzi," University of Milan, Milan, Italy
| | - Laura Alberio
- Department of Biosciences, University of Milan, Milan, Italy; Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Giacomo Parisi
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Gabriele Cerutti
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - Oliver B Clarke
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA; Department of Anesthesiology, Columbia University, New York, NY, USA
| | - Kay Hamacher
- Department of Biology, TU-Darmstadt, Darmstadt, Germany
| | - Henry M Colecraft
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Filippo Mancia
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Wayne A Hendrickson
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA; Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Steven A Siegelbaum
- Department of Neuroscience, Zuckerman Institute, Columbia University, New York, NY, USA
| | - Dario DiFrancesco
- Department of Biosciences, University of Milan, Milan, Italy; Institute of Biophysics-Milano, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Martino Bolognesi
- Department of Biosciences, University of Milan, Milan, Italy; Pediatric Research Center "Romeo ed Enrica Invernizzi," University of Milan, Milan, Italy
| | - Gerhard Thiel
- Department of Biology, TU-Darmstadt, Darmstadt, Germany
| | - Bina Santoro
- Department of Neuroscience, Zuckerman Institute, Columbia University, New York, NY, USA.
| | - Anna Moroni
- Department of Biosciences, University of Milan, Milan, Italy; Institute of Biophysics-Milano, Consiglio Nazionale delle Ricerche, Rome, Italy.
| |
Collapse
|
22
|
DiFrancesco ML, Mesirca P, Bidaud I, Isbrandt D, Mangoni ME. The funny current in genetically modified mice. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 166:39-50. [PMID: 34129872 DOI: 10.1016/j.pbiomolbio.2021.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/18/2021] [Accepted: 06/07/2021] [Indexed: 12/27/2022]
Abstract
Since its first description in 1979, the hyperpolarization-activated funny current (If) has been the object of intensive research aimed at understanding its role in cardiac pacemaker activity and its modulation by the sympathetic and parasympathetic branches of the autonomic nervous system. If was described in isolated tissue strips of the rabbit sinoatrial node using the double-electrode voltage-clamp technique. Since then, the rabbit has been the principal animal model for studying pacemaker activity and If for more than 20 years. In 2001, the first study describing the electrophysiological properties of mouse sinoatrial pacemaker myocytes and those of If was published. It was soon followed by the description of murine myocytes of the atrioventricular node and the Purkinje fibres. The sinoatrial node of genetically modified mice has become a very popular model for studying the mechanisms of cardiac pacemaker activity. This field of research benefits from the impressive advancement of in-vivo exploration techniques of physiological parameters, imaging, genetics, and large-scale genomic approaches. The present review discusses the influence of mouse genetic on the most recent knowledge of the funny current's role in the physiology and pathophysiology of cardiac pacemaker activity. Genetically modified mice have provided important insights into the role of If in determining intrinsic automaticity in vivo and in myocytes of the conduction system. In addition, gene targeting of f-(HCN) channel isoforms have contributed to elucidating the current's role in the regulation of heart rate by the parasympathetic nervous system. This review is dedicated to Dario DiFrancesco on his retirement.
Collapse
Affiliation(s)
- Mattia L DiFrancesco
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy; Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France; LabEx Ion Channels Science and Therapeutics (ICST), France.
| | - Pietro Mesirca
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France; LabEx Ion Channels Science and Therapeutics (ICST), France
| | - Isabelle Bidaud
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France; LabEx Ion Channels Science and Therapeutics (ICST), France
| | - Dirk Isbrandt
- Deutsches Zentrum für Neurodegenerative Erktankungen (DZNE), Bonn, Germany; University of Cologne, Institute for Molecular and Behavioral Neuroscience, Cologne, Germany
| | - Matteo E Mangoni
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France; LabEx Ion Channels Science and Therapeutics (ICST), France.
| |
Collapse
|
23
|
Grainger N, Guarina L, Cudmore RH, Santana LF. The Organization of the Sinoatrial Node Microvasculature Varies Regionally to Match Local Myocyte Excitability. FUNCTION (OXFORD, ENGLAND) 2021; 2:zqab031. [PMID: 34250490 PMCID: PMC8259512 DOI: 10.1093/function/zqab031] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/28/2021] [Accepted: 06/10/2021] [Indexed: 01/06/2023]
Abstract
The cardiac cycle starts when an action potential is produced by pacemaking cells in the sinoatrial node. This cycle is repeated approximately 100 000 times in humans and 1 million times in mice per day, imposing a monumental metabolic demand on the heart, requiring efficient blood supply via the coronary vasculature to maintain cardiac function. Although the ventricular coronary circulation has been extensively studied, the relationship between vascularization and cellular pacemaking modalities in the sinoatrial node is poorly understood. Here, we tested the hypothesis that the organization of the sinoatrial node microvasculature varies regionally, reflecting local myocyte firing properties. We show that vessel densities are higher in the superior versus inferior sinoatrial node. Accordingly, sinoatrial node myocytes are closer to vessels in the superior versus inferior regions. Superior and inferior sinoatrial node myocytes produce stochastic subthreshold voltage fluctuations and action potentials. However, the intrinsic action potential firing rate of sinoatrial node myocytes is higher in the superior versus inferior node. Our data support a model in which the microvascular densities vary regionally within the sinoatrial node to match the electrical and Ca2+ dynamics of nearby myocytes, effectively determining the dominant pacemaking site within the node. In this model, the high vascular density in the superior sinoatrial node places myocytes with metabolically demanding, high-frequency action potentials near vessels. The lower vascularization and electrical activity of inferior sinoatrial node myocytes could limit these cells to function to support sinoatrial node periodicity with sporadic voltage fluctuations via a stochastic resonance mechanism.
Collapse
|
24
|
Dual role of miR-1 in the development and function of sinoatrial cells. J Mol Cell Cardiol 2021; 157:104-112. [PMID: 33964276 DOI: 10.1016/j.yjmcc.2021.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 04/27/2021] [Accepted: 05/03/2021] [Indexed: 11/20/2022]
Abstract
miR-1, the most abundant miRNA in the heart, modulates expression of several transcription factors and ion channels. Conditions affecting the heart rate, such as endurance training and cardiac diseases, show a concomitant miR-1 up- or down-regulation. Here, we investigated the role of miR-1 overexpression in the development and function of sinoatrial (SAN) cells using murine embryonic stem cells (mESC). We generated mESCs either overexpressing miR-1 and EGFP (miR1OE) or EGFP only (EM). SAN-like cells were selected from differentiating mESC using the CD166 marker. Gene expression and electrophysiological analysis were carried out on both early mES-derived cardiac progenitors and SAN-like cells and on beating neonatal rat ventricular cardiomyocytes (NRVC) over-expressing miR-1. miR1OE cells increased significantly the proportion of CD166+ SAN precursors compared to EM cells (23% vs 12%) and the levels of the transcription factors TBX5 and TBX18, both involved in SAN development. miR1OE SAN-like cells were bradycardic (1,3 vs 2 Hz) compared to EM cells. In agreement with data on native SAN cells, EM SAN-like cardiomyocytes show two populations of cells expressing either slow- or fast-activating If currents; miR1OE SAN-like cells instead have only fast-activating If with a significantly reduced conductance. Western Blot and immunofluorescence analysis showed a reduced HCN4 signal in miR-1OE vs EM CD166+ precursors. Together these data point out to a specific down-regulation of the slow-activating HCN4 subunit by miR-1. Importantly, the rate and If alterations were independent of the developmental effects of miR-1, being similar in NRVC transiently overexpressing miR-1. In conclusion, we demonstrated a dual role of miR-1, during development it controls the proper development of sinoatrial-precursor, while in mature SAN-like cells it modulates the HCN4 pacemaker channel translation and thus the beating rate.
Collapse
|
25
|
Co ML, Khouzam JP, Pour-Ghaz I, Minhas S, Basu-Ray I. Emerging Technologies in Cardiac Pacing From Leadless Pacers to Stem Cells. Curr Probl Cardiol 2021; 46:100797. [PMID: 33561694 DOI: 10.1016/j.cpcardiol.2021.100797] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 01/10/2021] [Indexed: 01/28/2023]
Abstract
Modern pacemakers can sense and pace multiple chambers of the heart. These pacemakers have different modes and features to optimize atrioventricular synchrony and promote intrinsic conduction. Despite recent advancements, current pacemakers have several drawbacks that limit their feasibility. In this review article, we discuss several of these limitations and detail several emerging technologies in cardiac pacing aimed to solve some of these limitations. We present several technological advancements in cardiac pacing, including the use of leadless pacemakers, physiologic pacing, battery improvements, and bioartificial pacemakers. More research still needs to be done in testing the safety and efficacy of these new developments.
Collapse
Affiliation(s)
- Michael Lawren Co
- Department of Cardiology, Loma Linda University Medical Center, Loma Linda, CA
| | | | - Issa Pour-Ghaz
- Department of Cardiology, University of Tennessee Health Science Center, Memphis, TN
| | - Sheharyar Minhas
- Department of Internal Medicine, Baptist Memorial Hospital, Memphis, TN
| | - Indranill Basu-Ray
- Arrythmia Service, Department of Cardiology, Memphis VA Medical Center, The University of Memphis, Memphis, TN.
| |
Collapse
|
26
|
Mesirca P, Fedorov VV, Hund TJ, Torrente AG, Bidaud I, Mohler PJ, Mangoni ME. Pharmacologic Approach to Sinoatrial Node Dysfunction. Annu Rev Pharmacol Toxicol 2021; 61:757-778. [PMID: 33017571 PMCID: PMC7790915 DOI: 10.1146/annurev-pharmtox-031120-115815] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The spontaneous activity of the sinoatrial node initiates the heartbeat. Sino-atrial node dysfunction (SND) and sick sinoatrial (sick sinus) syndrome are caused by the heart's inability to generate a normal sinoatrial node action potential. In clinical practice, SND is generally considered an age-related pathology, secondary to degenerative fibrosis of the heart pacemaker tissue. However, other forms of SND exist, including idiopathic primary SND, which is genetic, and forms that are secondary to cardiovascular or systemic disease. The incidence of SND in the general population is expected to increase over the next half century, boosting the need to implant electronic pacemakers. During the last two decades, our knowledge of sino-atrial node physiology and of the pathophysiological mechanisms underlying SND has advanced considerably. This review summarizes the current knowledge about SND mechanisms and discusses the possibility of introducing new pharmacologic therapies for treating SND.
Collapse
Affiliation(s)
- Pietro Mesirca
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34096 Montpellier, France;
- LabEx Ion Channels Science and Therapeutics (ICST), 06560 Nice, France
| | - Vadim V Fedorov
- Frick Center for Heart Failure and Arrhythmia at the Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Wexner Medical Center, Columbus, Ohio 43210, USA
| | - Thomas J Hund
- Frick Center for Heart Failure and Arrhythmia at the Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Angelo G Torrente
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34096 Montpellier, France;
- LabEx Ion Channels Science and Therapeutics (ICST), 06560 Nice, France
| | - Isabelle Bidaud
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34096 Montpellier, France;
- LabEx Ion Channels Science and Therapeutics (ICST), 06560 Nice, France
| | - Peter J Mohler
- Frick Center for Heart Failure and Arrhythmia at the Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Wexner Medical Center, Columbus, Ohio 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, Ohio 43210, USA
| | - Matteo E Mangoni
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34096 Montpellier, France;
- LabEx Ion Channels Science and Therapeutics (ICST), 06560 Nice, France
| |
Collapse
|
27
|
Boddum K, Skafte-Pedersen P, Rolland JF, Wilson S. Optogenetics and Optical Tools in Automated Patch Clamping. Methods Mol Biol 2021; 2188:311-330. [PMID: 33119859 DOI: 10.1007/978-1-0716-0818-0_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Automated patch clamping (APC) has been used for almost two decades to increase the throughput of electrophysiological measurements, especially in preclinical safety screening of drug compounds. Typically, cells are suctioned onto holes in planar surfaces and a stronger subsequent suction allows access to a whole cell configuration for electrical measurement of ion channel activity. The development of optogenetic tools over a wide range of wavelengths (UV to IR) provides powerful tools for improving spatiotemporal control of in vivo and in vitro experiments and is emerging as a powerful means of investigating cell networks (neuronal), single cell transduction, and subcellular pathways.Combining APC and optogenetic tools paves the way for improved investigation and control of cell kinetics and provides the opportunity for collecting robust data for new and exciting applications and therapeutic areas. Here, we present an APC optogenetics capability on the Qube Opto 384 system including experiments on light activated ion channels and photoactivated ligands.
Collapse
Affiliation(s)
- Kim Boddum
- Sophion Bioscience A/S, Ballerup, Denmark
| | | | | | | |
Collapse
|
28
|
Liang Y, Xu Z, Wu X, Pang J, Zhou P, Cao Y. Inhibition of hyperpolarization-activated cyclic nucleotide-gated channels with natural flavonoid quercetin. Biochem Biophys Res Commun 2020; 533:952-957. [PMID: 33008592 DOI: 10.1016/j.bbrc.2020.09.102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 09/22/2020] [Indexed: 11/18/2022]
Abstract
Quercetin is a natural flavonoid which has been reported to be analgesic in different animal models of pain. However, the mechanism underlying the pain-relieving effects is still unclear. Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels play critical roles in controlling pacemaker activity in cardiac and nervous systems, making the channel a new target for therapeutic exploration. In this study, we explored a series of flavonoids for their modulation on HCN channels. Among all tested flavonoids, quercetin was the most potent inhibitor for HCN channels with an IC50 value of 27.32 ± 1.19 μM for HCN2. Furthermore, quercetin prominently left shifted the voltage-dependent activation curves of HCN channels and decelerated deactivation process. The results presented herein firstly characterize quercetin as a novel and potent inhibitor for HCN channels, which represents a novel structure for future drug design of HCN channel inhibitors.
Collapse
Affiliation(s)
- Yemei Liang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Ziwei Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaoyan Wu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jianxin Pang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Pingzheng Zhou
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| | - Ying Cao
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
29
|
Lu Z, Wang HZ, Gordon CR, Ballou LM, Lin RZ, Cohen IS. Regulation of HCN2 Current by PI3K/Akt Signaling. Front Physiol 2020; 11:587040. [PMID: 33240105 PMCID: PMC7680966 DOI: 10.3389/fphys.2020.587040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/16/2020] [Indexed: 12/19/2022] Open
Abstract
It has long been known that heart rate is regulated by the autonomic nervous system. Recently, we demonstrated that the pacemaker current, I f , is regulated by phosphoinositide 3-kinase (PI3K) signaling independently of the autonomic nervous system. Inhibition of PI3K in sinus node (SN) myocytes shifts the activation of I f by almost 16 mV in the negative direction. I f in the SN is predominantly mediated by two members of the HCN gene family, HCN4 and HCN1. Purkinje fibers also possess I f and are an important secondary pacemaker in the heart. In contrast to the SN, they express HCN2 and HCN4, while ventricular myocytes, which do not normally pace, express HCN2 alone. In the current work, we investigated PI3K regulation of HCN2 expressed in HEK293 cells. Treatment with the PI3K inhibitor PI-103 caused a negative shift in the activation voltage and a dramatic reduction in the magnitude of the HCN2 current. Similar changes were also seen in cells treated with an inhibitor of the protein kinase Akt, a downstream effector of PI3K. The effects of PI-103 were reversed by perfusion of cells with phosphatidylinositol 3,4,5-trisphosphate (the second messenger produced by PI3K) or active Akt protein. We identified serine 861 in mouse HCN2 as a putative Akt phosphorylation site. Mutation of S861 to alanine mimicked the effects of Akt inhibition on voltage dependence and current magnitude. In addition, the Akt inhibitor had no effect on the mutant channel. These results suggest that Akt phosphorylation of mHCN2 S861 accounts for virtually all of the observed actions of PI3K signaling on the HCN2 current. Unexpectedly, Akt inhibition had no effect on I f in SN myocytes. This result raises the possibility that diverse PI3K signaling pathways differentially regulate HCN-induced currents in different tissues, depending on the isoforms expressed.
Collapse
Affiliation(s)
- Zhongju Lu
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, United States
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Hong Zhan Wang
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, United States
| | - Chris R. Gordon
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, United States
- Department of Nephrology, Stony Brook University, Stony Brook, NY, United States
| | - Lisa M. Ballou
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, United States
| | - Richard Z. Lin
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, United States
- Medical Service, Northport VA Medical Center, Northport, NY, United States
| | - Ira S. Cohen
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
30
|
Translating GWAS-identified loci for cardiac rhythm and rate using an in vivo image- and CRISPR/Cas9-based approach. Sci Rep 2020; 10:11831. [PMID: 32678143 PMCID: PMC7367351 DOI: 10.1038/s41598-020-68567-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 06/29/2020] [Indexed: 02/07/2023] Open
Abstract
A meta-analysis of genome-wide association studies (GWAS) identified eight loci that are associated with heart rate variability (HRV), but candidate genes in these loci remain uncharacterized. We developed an image- and CRISPR/Cas9-based pipeline to systematically characterize candidate genes for HRV in live zebrafish embryos. Nine zebrafish orthologues of six human candidate genes were targeted simultaneously in eggs from fish that transgenically express GFP on smooth muscle cells (Tg[acta2:GFP]), to visualize the beating heart. An automated analysis of repeated 30 s recordings of beating atria in 381 live, intact zebrafish embryos at 2 and 5 days post-fertilization highlighted genes that influence HRV (hcn4 and si:dkey-65j6.2 [KIAA1755]); heart rate (rgs6 and hcn4); and the risk of sinoatrial pauses and arrests (hcn4). Exposure to 10 or 25 µM ivabradine—an open channel blocker of HCNs—for 24 h resulted in a dose-dependent higher HRV and lower heart rate at 5 days post-fertilization. Hence, our screen confirmed the role of established genes for heart rate and rhythm (RGS6 and HCN4); showed that ivabradine reduces heart rate and increases HRV in zebrafish embryos, as it does in humans; and highlighted a novel gene that plays a role in HRV (KIAA1755).
Collapse
|
31
|
Şengül Ayan S, Sırcan AK, Abewa M, Kurt A, Dalaman U, Yaraş N. Mathematical model of the ventricular action potential and effects of isoproterenol-induced cardiac hypertrophy in rats. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2020; 49:323-342. [PMID: 32462262 DOI: 10.1007/s00249-020-01439-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 04/21/2020] [Accepted: 05/17/2020] [Indexed: 12/16/2022]
Abstract
Mathematical action potential (AP) modeling is a well-established but still-developing area of research to better understand physiological and pathological processes. In particular, changes in AP mechanisms in the isoproterenol (ISO) -induced hypertrophic heart model are incompletely understood. Here we present a mathematical model of the rat AP based on recordings from rat ventricular myocytes. In our model, for the first time, all channel kinetics are defined with a single type of function that is simple and easy to apply. The model AP and channels dynamics are consistent with the APs recorded from rats for both Control (absence of ISO) and ISO-treated cases. Our mathematical model helps us to understand the reason for the prolongation in AP duration after ISO application while ISO treatment helps us to validate our mathematical model. We reveal that the smaller density and the slower gating kinetics of the transient K+ current help explain the prolonged AP duration after ISO treatment and the increasing amplitude of the rapid and the slow inward rectifier currents also contribute to this prolongation alongside the flux in Ca2+ currents. ISO induced an increase in the density of the Na+ current that can explain the faster upstroke. We believe that AP dynamics from rat ventricular myocytes can be reproduced very well with this mathematical model and that it provides a powerful tool for improved insights into the underlying dynamics of clinically important AP properties such as ISO application.
Collapse
Affiliation(s)
- Sevgi Şengül Ayan
- Department of Engineering, Industrial Engineering, Antalya Bilim University, Döşemealtı, Antalya, Turkey.
| | - Ahmet K Sırcan
- Department of Engineering, Electrical and Computer Engineering, Antalya Bilim University, Döşemealtı, Antalya, Turkey
| | - Mohamedou Abewa
- Department of Engineering, Electrical and Computer Engineering, Antalya Bilim University, Döşemealtı, Antalya, Turkey
| | - Ahmet Kurt
- Department of Engineering, Electrical and Computer Engineering, Florida International University, Miami, USA
| | - Uğur Dalaman
- Department of Biophysics, Akdeniz University College of Medicine, Akdeniz University, Antalya, Turkey
| | - Nazmi Yaraş
- Department of Biophysics, Akdeniz University College of Medicine, Akdeniz University, Antalya, Turkey
| |
Collapse
|
32
|
Valiunas V, Cohen IS, Brink PR, Clausen C. A study of the outward background current conductance g K1, the pacemaker current conductance g f, and the gap junction conductance g j as determinants of biological pacing in single cells and in a two-cell syncytium using the dynamic clamp. Pflugers Arch 2020; 472:561-570. [PMID: 32415460 DOI: 10.1007/s00424-020-02378-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 12/21/2022]
Abstract
We previously demonstrated that a two-cell syncytium, composed of a ventricular myocyte and an mHCN2 expressing cell, recapitulated most properties of in vivo biological pacing induced by mHCN2-transfected hMSCs in the canine ventricle. Here, we use the two-cell syncytium, employing dynamic clamp, to study the roles of gf (pacemaker conductance), gK1 (background K+ conductance), and gj (intercellular coupling conductance) in biological pacing. We studied gf and gK1 in single HEK293 cells expressing cardiac sodium current channel Nav1.5 (SCN5A). At fixed gf, increasing gK1 hyperpolarized the cell and initiated pacing. As gK1 increased, rate increased, then decreased, finally ceasing at membrane potentials near EK. At fixed gK1, increasing gf depolarized the cell and initiated pacing. With increasing gf, rate increased reaching a plateau, then decreased, ceasing at a depolarized membrane potential. We studied gj via virtual coupling with two non-adjacent cells, a driver (HEK293 cell) in which gK1 and gf were injected without SCN5A and a follower (HEK293 cell), expressing SCN5A. At the chosen values of gK1 and gf oscillations initiated in the driver, when gj was increased synchronized pacing began, which then decreased by about 35% as gj approached 20 nS. Virtual uncoupling yielded similar insights into gj. We also studied subthreshold oscillations in physically and virtually coupled cells. When coupling was insufficient to induce pacing, passive spread of the oscillations occurred in the follower. These results show a non-monotonic relationship between gK1, gf, gj, and pacing. Further, oscillations can be generated by gK1 and gf in the absence of SCN5A.
Collapse
Affiliation(s)
- Virginijus Valiunas
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, Stony Brook, NY, 11794-8661, USA.
| | - Ira S Cohen
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, Stony Brook, NY, 11794-8661, USA
| | - Peter R Brink
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, Stony Brook, NY, 11794-8661, USA
| | - Chris Clausen
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, Stony Brook, NY, 11794-8661, USA
| |
Collapse
|
33
|
Wang ZJ, Blanco I, Hayoz S, Brelidze TI. The HCN domain is required for HCN channel cell-surface expression and couples voltage- and cAMP-dependent gating mechanisms. J Biol Chem 2020; 295:8164-8173. [PMID: 32341127 DOI: 10.1074/jbc.ra120.013281] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/23/2020] [Indexed: 11/06/2022] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are major regulators of synaptic plasticity and rhythmic activity in the heart and brain. Opening of HCN channels requires membrane hyperpolarization and is further facilitated by intracellular cyclic nucleotides (cNMPs). In HCN channels, membrane hyperpolarization is sensed by the membrane-spanning voltage sensor domain (VSD), and the cNMP-dependent gating is mediated by the intracellular cyclic nucleotide-binding domain (CNBD) connected to the pore-forming S6 transmembrane segment via the C-linker. Previous functional analysis of HCN channels has suggested a direct or allosteric coupling between the voltage- and cNMP-dependent activation mechanisms. However, the specifics of this coupling remain unclear. The first cryo-EM structure of an HCN1 channel revealed that a novel structural element, dubbed the HCN domain (HCND), forms a direct structural link between the VSD and C-linker-CNBD. In this study, we investigated the functional significance of the HCND. Deletion of the HCND prevented surface expression of HCN2 channels. Based on the HCN1 structure analysis, we identified Arg237 and Gly239 residues on the S2 of the VSD that form direct interactions with Ile135 on the HCND. Disrupting these interactions abolished HCN2 currents. We also identified three residues on the C-linker-CNBD (Glu478, Gln482, and His559) that form direct interactions with residues Arg154 and Ser158 on the HCND. Disrupting these interactions affected both voltage- and cAMP-dependent gating of HCN2 channels. These findings indicate that the HCND is necessary for the cell-surface expression of HCN channels and provides a functional link between voltage- and cAMP-dependent mechanisms of HCN channel gating.
Collapse
Affiliation(s)
- Ze-Jun Wang
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, D. C., USA
| | - Ismary Blanco
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, D. C., USA
| | - Sebastien Hayoz
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, D. C., USA
| | - Tinatin I Brelidze
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, D. C., USA .,Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, D. C., USA
| |
Collapse
|
34
|
Wei F, Wang Q, Han J, Goswamee P, Gupta A, McQuiston AR, Liu Q, Zhou L. Photodynamic Modification of Native HCN Channels Expressed in Thalamocortical Neurons. ACS Chem Neurosci 2020; 11:851-863. [PMID: 32078767 DOI: 10.1021/acschemneuro.9b00475] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The photodynamic process requires three elements: light, oxygen, and photosensitizer, and involves the formation of singlet oxygen, the molecular oxygen in excited electronic states. Previously, we reported that heterologously expressed hyperpolarization-activated cAMP-gated (HCN) channels in excised membrane patches are sensitive to photodynamic modification (PDM). Here we extend this study to native HCN channels expressed in thalamocortical (TC) neurons in the ventrobasal (VB) complex of the thalamus and dopaminergic neurons (DA) of the ventral tegmental area (VTA). To do this, we introduced the photosensitizer FITC-cAMP into TCs or DAs of rodent brain slices via a whole-cell patch-clamp recording pipette. After illumination with blue light pulses, we observed an increase in the voltage-insensitive, instantaneous Iinst component, accompanied by a long-lasting decrease in the hyperpolarization-dependent Ih component. Both Ih and the increased Iinst after PDM could be blocked by the HCN blockers Cs+ and ZD7288. When FITC and cAMP were dissociated and loaded into neurons as two separate chemicals, light application did not result in any long-lasting changes of the HCN currents. In contrast, light pulses applied to HCN2-/- neurons loaded with FITC-cAMP generated a much greater reduction in the Iinst component compared to that of WT neurons. Next, we investigated the impact of the long-lasting increases in Iinst after PDM on the cellular physiology of VB neurons. Consistent with an upregulation of HCN channel function, PDM elicited a depolarization of the resting membrane potential (RMP). Importantly, Trolox-C, an effective quencher for singlet oxygen, could block the PDM-dependent increase in Iinst and depolarization of the RMP. We propose that PDM of native HCN channels under physiological conditions may provide a photodynamic approach to alleviate HCN channelopathy in certain pathological conditions.
Collapse
Affiliation(s)
- Fusheng Wei
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330031, Jiangxi, China
| | - Qiang Wang
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Jizhong Han
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Priyodarshan Goswamee
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Ankush Gupta
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Adam Rory McQuiston
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Qinglian Liu
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Lei Zhou
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| |
Collapse
|
35
|
Shults NV, Rybka V, Suzuki YJ, Brelidze TI. Increased Smooth Muscle Kv11.1 Channel Expression in Pulmonary Hypertension and Protective Role of Kv11.1 Channel Blocker Dofetilide. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:48-56. [PMID: 31839145 PMCID: PMC6943378 DOI: 10.1016/j.ajpath.2019.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 09/05/2019] [Accepted: 09/17/2019] [Indexed: 01/08/2023]
Abstract
Kv11.1 potassium channels are essential for heart repolarization. Prescription medication that blocks Kv11.1 channels lengthens the ventricular action potential and causes cardiac arrhythmias. Surprisingly little is known about the Kv11.1 channel expression and function in the lung tissue. Here we report that Kv11.1 channels were abundantly expressed in the large pulmonary arteries (PAs) of healthy lung tissues from humans and rats. Kv11.1 channel expression was increased in the lungs of humans affected by chronic obstructive pulmonary disease-associated pulmonary hypertension and in the lungs of rats with pulmonary arterial hypertension (PAH). In healthy lung tissues from humans and rats, Kv11.1 channels were confined to the large PAs. In humans with chronic obstructive pulmonary disease-associated pulmonary hypertension and in rats with PAH, Kv11.1 channels were expressed in both the large and small PAs. The increase in Kv11.1 channel expression closely followed the time-course of the development of pulmonary vascular remodeling in PAH rats. Treatment of PAH rats with dofetilide, an Kv11.1 channel blocker approved by the US Food and Drug Administration for use in the treatment of arrythmia, inhibited PAH-associated pulmonary vascular remodeling. Taken together, the findings from this study uncovered a novel role of Kv11.1 channels in lung function and their potential as new drug targets in the treatment of pulmonary hypertension. The protective effect of dofetilide raises the possibility of repurposing this antiarrhythmic drug for the treatment of patients with pulmonary hypertension.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Animals
- Arrhythmias, Cardiac/etiology
- Arrhythmias, Cardiac/pathology
- Arrhythmias, Cardiac/prevention & control
- Case-Control Studies
- ERG1 Potassium Channel/antagonists & inhibitors
- ERG1 Potassium Channel/metabolism
- Female
- Follow-Up Studies
- Humans
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Phenethylamines/pharmacology
- Potassium Channel Blockers/pharmacology
- Prognosis
- Pulmonary Arterial Hypertension/complications
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/pathology
- Rats, Sprague-Dawley
- Sulfonamides/pharmacology
- Vascular Remodeling/drug effects
Collapse
Affiliation(s)
- Nataliia V Shults
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia
| | - Vladyslava Rybka
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia
| | - Yuichiro J Suzuki
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia
| | - Tinatin I Brelidze
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia.
| |
Collapse
|
36
|
Yang M, Zhao Q, Zhao H, Yang A, Wang F, Wang X, Tang Y, Huang C. Adipose‑derived stem cells overexpressing SK4 calcium‑activated potassium channel generate biological pacemakers. Int J Mol Med 2019; 44:2103-2112. [PMID: 31638180 PMCID: PMC6844603 DOI: 10.3892/ijmm.2019.4374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 09/11/2019] [Indexed: 01/14/2023] Open
Abstract
Recent studies have suggested that calcium-activated potassium channel (KCa) agonists increase the proportion of mouse embryonic stem cell-derived cardiomyocytes and promote the differentiation of pacemaker cells. In the present study, it was hypothesized that adipose-derived stem cells (ADSCs) can differentiate into pacemaker-like cells via over-expression of the SK4 gene. ADSCs were transduced with a recombinant adenovirus vector carrying the mouse SK4 gene, whereas the control group was transduced with GFP vector. ADSCs transduced with SK4 vector were implanted into the rat left ventricular free wall. Complete atrioventricular block (AVB) was established in isolated perfused rat hearts after 2 weeks. SK4 was successfully and stably expressed in ADSCs following transduction. The mRNA levels of the pluripotent markers Oct-4 and Sox-2 declined and that of the transcription factor Shox2 was upregulated following SK4 transduction. The expression of α-actinin and hyperpolarization-activated cyclic nucleotide-gated potassium channel 4 (HCN4) increased in the SK4 group. The hyperpolarizing activated pacemaker current If (8/20 cells) was detected in ADSCs transduced with SK4, but not in the GFP group. Furthermore, SK4 transduction induced the expression of p-ERK1/2 and p-p38 MAPK. In the ex vivo experiments, the heart rate of the SK4 group following AVB establishment was significantly higher compared with that in the GFP group. Immunofluorescence revealed that the transduced ADSCs were successfully implanted and expressed HCN4 in the SK4 group. In conclusion, SK4 induced ADSCs to differentiate into cardiomyocyte-like and pacemaker-like cells via activation of the extracellular signal-regulated kinase 1/2 and p38 mitogen-activated protein kinase pathways. Therefore, ADSCs transduced with SK4 may be used to generate biological pacemakers in ex vivo rat hearts.
Collapse
Affiliation(s)
- Mei Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qingyan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hongyi Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Ankang Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Fengyuan Wang
- 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
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
37
|
Identification of peripheral neural circuits that regulate heart rate using optogenetic and viral vector strategies. Nat Commun 2019; 10:1944. [PMID: 31028266 PMCID: PMC6486614 DOI: 10.1038/s41467-019-09770-1] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 03/27/2019] [Indexed: 11/26/2022] Open
Abstract
Heart rate is under the precise control of the autonomic nervous system. However, the wiring of peripheral neural circuits that regulate heart rate is poorly understood. Here, we develop a clearing-imaging-analysis pipeline to visualize innervation of intact hearts in 3D and employed a multi-technique approach to map parasympathetic and sympathetic neural circuits that control heart rate in mice. We identify cholinergic neurons and noradrenergic neurons in an intrinsic cardiac ganglion and the stellate ganglia, respectively, that project to the sinoatrial node. We also report that the heart rate response to optogenetic versus electrical stimulation of the vagus nerve displays different temporal characteristics and that vagal afferents enhance parasympathetic and reduce sympathetic tone to the heart via central mechanisms. Our findings provide new insights into neural regulation of heart rate, and our methodology to study cardiac circuits can be readily used to interrogate neural control of other visceral organs. The wiring of peripheral neural circuits that regulate heart rate is poorly understood. In this study, authors used tissue clearing for high-resolution characterization of nerves in the heart in 3D and transgenic and novel viral vector approaches to identify peripheral parasympathetic and sympathetic neuronal populations involved in heart rate control in mice.
Collapse
|
38
|
Santos-Vera B, Vaquer-Alicea ADC, Maria-Rios CE, Montiel-Ramos A, Ramos-Cardona A, Vázquez-Torres R, Sanabria P, Jiménez-Rivera CA. Protein and surface expression of HCN2 and HCN4 subunits in mesocorticolimbic areas after cocaine sensitization. Neurochem Int 2019; 125:91-98. [PMID: 30794847 DOI: 10.1016/j.neuint.2019.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/05/2019] [Accepted: 02/06/2019] [Indexed: 01/20/2023]
Abstract
The Ih is a mixed depolarizing current present in neurons which, upon activation by hyperpolarization, modulates neuronal excitability in the mesocorticolimbic (MCL) system, an area which regulates emotions such as pleasure, reward, and motivation. Its biophysical properties are determined by HCN protein expression profiles, specifically HCN subunits 1-4. Previously, we reported that cocaine-induced behavioral sensitization increases HCN2 protein expression in all MCL areas with the Ventral Tegmental Area (VTA) showing the most significant increase. Recent evidence suggests that HCN4 also has an important expression in the MCL system. Although there is a significant expression of HCN channels in the MCL system their role in addictive processes is largely unknown. Thus, in this study we aim to compare HCN2 and HCN4 expression profiles and their cellular compartmental distribution in the MCL system, before and after cocaine sensitization. Surface/intracellular (S/I) ratio analysis indicates that VTA HCN2 subunits are mostly expressed in the cell surface in contrast to other areas tested. Our findings demonstrate that after cocaine sensitization, the HCN2 S/I ratio in the VTA was decreased whereas in the Prefrontal Cortex it was increased. In addition, HCN4 total expression in the VTA was decreased after cocaine sensitization, although the S/I ratio was not altered. Together, these results demonstrate differential cocaine effects on HCN2 and HCN4 protein expression profiles and therefore suggest a diverse Ih modulation of cellular activity during cocaine addictive processes.
Collapse
Affiliation(s)
- Bermary Santos-Vera
- Physiology Department, University of Puerto Rico, School of Medicine, Medical Sciences Campus, San Juan, 00936-5067, Puerto Rico
| | - Ana Del C Vaquer-Alicea
- Physiology Department, University of Puerto Rico, School of Medicine, Medical Sciences Campus, San Juan, 00936-5067, Puerto Rico
| | - Cristina E Maria-Rios
- Biology Department, University of Puerto Rico, Rio Piedras Campus, San Juan, 00936-5067, Puerto Rico
| | - Alan Montiel-Ramos
- Biology Department, University of Puerto Rico, Rio Piedras Campus, San Juan, 00936-5067, Puerto Rico
| | - Aynette Ramos-Cardona
- Psychology Department, University of Puerto Rico, Rio Piedras Campus, San Juan, 00936-5067, Puerto Rico
| | - Rafael Vázquez-Torres
- Physiology Department, University of Puerto Rico, School of Medicine, Medical Sciences Campus, San Juan, 00936-5067, Puerto Rico
| | - Priscila Sanabria
- Physiology Department, Universidad Central del Caribe, Bayamon, 00960, Puerto Rico
| | - Carlos A Jiménez-Rivera
- Physiology Department, University of Puerto Rico, School of Medicine, Medical Sciences Campus, San Juan, 00936-5067, Puerto Rico.
| |
Collapse
|
39
|
Analysis of Anasplatyrhynchos genome resequencing data reveals genetic signatures of artificial selection. PLoS One 2019; 14:e0211908. [PMID: 30735526 PMCID: PMC6368380 DOI: 10.1371/journal.pone.0211908] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 01/22/2019] [Indexed: 02/05/2023] Open
Abstract
Three artificially selected duck populations (AS), higher lean meat ratios (LTPD), higher fat ratios (FTPD) and higher quality meat (CMD), have been developed in China, providing excellent populations for investigation of artificial selection effects. However, the genetic signatures of artificial selection are unclear. In this study, we sequenced the genome sequences of these three artificially selected populations and their ancestral population (mallard, M). We then compared the genome sequences between AS and M and between LTPD and FTPD using integrated strategies such as anchoring scaffolds to pseudo-chromosomes, mutation detection, selective screening, GO analysis, qRT-PCR, and protein multiple sequences alignment to uncover genetic signatures of selection. We anchored duck scaffolds to pseudo-chromosomes and obtained 28 pseudo-chromosomes, accounting for 84% of duck genome in length. Totally 78 and 99 genes were found to be under selection between AS and M and between LTPD and FTPD. Genes under selection between AS and M mainly involved in pigmentation and heart rates, while genes under selection between LTPD and FTPD involved in muscle development and fat deposition. A heart rate regulator (HCN1), the strongest selected gene between AS and M, harbored a GC deletion in AS and displayed higher mRNA expression level in M than in AS. IGF2R, a regulator of skeletal muscle mass, was found to be under selection between FTPD and LTPD. We also found two nonsynonymous substitutions in IGF2R, which might lead to higher IGF2R mRNA expression level in FTPD than LTPD, indicating the two nonsynonymous substitutions might play a key role for the regulation of duck skeletal muscle mass. Taken together, these results of this study provide valuable insight for the genetic basis of duck artificial selection.
Collapse
|
40
|
Tanguay J, Callahan KM, D'Avanzo N. Characterization of drug binding within the HCN1 channel pore. Sci Rep 2019; 9:465. [PMID: 30679654 PMCID: PMC6345760 DOI: 10.1038/s41598-018-37116-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 11/29/2018] [Indexed: 11/09/2022] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels mediate rhythmic electrical activity of cardiac pacemaker cells, and in neurons play important roles in setting resting membrane potentials, dendritic integration, neuronal pacemaking, and establishing action potential threshold. Block of HCN channels slows the heart rate and is currently used to treat angina. However, HCN block also provides a promising approach to the treatment of neuronal disorders including epilepsy and neuropathic pain. While several molecules that block HCN channels have been identified, including clonidine and its derivative alinidine, lidocaine, mepivacaine, bupivacaine, ZD7288, ivabradine, zatebradine, and cilobradine, their low affinity and lack of specificity prevents wide-spread use. Different studies suggest that the binding sites of these inhibitors are located in the inner vestibule of HCN channels, but the molecular details of their binding remain unknown. We used computational docking experiments to assess the binding sites and mode of binding of these inhibitors against the recently solved atomic structure of human HCN1 channels, and a homology model of the open pore derived from a closely related CNG channel. We identify a possible hydrophobic groove in the pore cavity that plays an important role in conformationally restricting the location and orientation of drugs bound to the inner vestibule. Our results also help explain the molecular basis of the low-affinity binding of these inhibitors, paving the way for the development of higher affinity molecules.
Collapse
Affiliation(s)
- Jérémie Tanguay
- Department of Physics, Université de Montréal, Montréal, Canada
| | - Karen M Callahan
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Canada
| | - Nazzareno D'Avanzo
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Canada.
| |
Collapse
|
41
|
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: 0.1] [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
|
42
|
Cao Y, Chen S, Liang Y, Wu T, Pang J, Liu S, Zhou P. Inhibition of hyperpolarization-activated cyclic nucleotide-gated channels by β-blocker carvedilol. Br J Pharmacol 2018; 175:3963-3975. [PMID: 30098004 DOI: 10.1111/bph.14469] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 07/02/2018] [Accepted: 07/30/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Carvedilol is a clinically effective β-blocker broadly used for treating congestive heart failure (CHF), and several clinical trials have demonstrated that it shows a favourable effect compared with other β-blockers in patients with CHF. The mechanism underlying this beneficial effect of carvedilol compared to other β-blockers is not clearly understood. In addition to β-blockers, inhibitors of hyperpolarization-activated cyclic nucleotide (HCN)-gated channels, which play a critical role in spontaneous rhythmic activity in the heart, have also been proposed to be suitable drugs for reducing heart rate and, therefore, beneficial for treating CHF. In the present study, we investigated the effect of carvedilol on HCN channels. EXPERIMENTAL APPROACH Whole-cell patch-clamp recordings were used to assess the effect of carvedilol on currents from wild-type and mutant HCN1, HCN2 and HCN4 channels expressed in CHO cells. KEY RESULTS Carvedilol was the only β-blocker tested that showed inhibitory effects on the major sinoatrial HCN channel isoform HCN4. Carvedilol inhibited HCN4 in a concentration-dependent manner with an EC50 of 4.4 μM. In addition, carvedilol also inhibited HCN1 and HCN2 channels. Carvedilol blocked HCN channels by decelerating the rate of channel activation and increasing that of deactivation, and shifted the voltage-dependence of activation leftwards. Our data also showed that carvedilol, unlike other inhibitors of this channel (ivabradine and ZD7288), is not an 'open-channel' inhibitor of HCN4. CONCLUSIONS AND IMPLICATIONS Carvedilol is a negative gating modulator of HCN channels. It represents a novel structure for future drug design of HCN channel inhibitors.
Collapse
Affiliation(s)
- Ying Cao
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Shujun Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Yemei Liang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Ting Wu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jianxin Pang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Shuwen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Southern Medical University, Guangzhou, China
| | - Pingzheng Zhou
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
43
|
HCN Channels: New Therapeutic Targets for Pain Treatment. Molecules 2018; 23:molecules23092094. [PMID: 30134541 PMCID: PMC6225464 DOI: 10.3390/molecules23092094] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/17/2018] [Accepted: 08/18/2018] [Indexed: 12/28/2022] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are highly regulated proteins which respond to different cellular stimuli. The HCN currents (Ih) mediated by HCN1 and HCN2 drive the repetitive firing in nociceptive neurons. The role of HCN channels in pain has been widely investigated as targets for the development of new therapeutic drugs, but the comprehensive design of HCN channel modulators has been restricted due to the lack of crystallographic data. The three-dimensional structure of the human HCN1 channel was recently reported, opening new possibilities for the rational design of highly-selective HCN modulators. In this review, we discuss the structural and functional properties of HCN channels, their pharmacological inhibitors, and the potential strategies for designing new drugs to block the HCN channel function associated with pain perception.
Collapse
|
44
|
Vetulli HM, Elizari MV, Naccarelli GV, Gonzalez MD. Cardiac automaticity: basic concepts and clinical observations. J Interv Card Electrophysiol 2018; 52:263-270. [PMID: 30112616 DOI: 10.1007/s10840-018-0423-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 07/19/2018] [Indexed: 10/28/2022]
Abstract
PURPOSE The purpose of this report was to review the basic mechanisms underlying cardiac automaticity. Second, we describe our clinical observations related to the anatomical and functional characteristics of sinus automaticity. METHODS We first reviewed the main discoveries regarding the mechanisms responsible for cardiac automaticity. We then analyzed our clinical experience regarding the location of sinus automaticity in two unique populations: those with inappropriate sinus tachycardia and those with a dominant pacemaker located outside the crista terminalis region. RESULTS We studied 26 patients with inappropriate sinus tachycardia (age 34 ± 8 years; 21 females). Non-contact endocardial mapping (Ensite 3000, Endocardial Solutions) was performed in 19 patients and high-density contact mapping (Carto-3, Biosense Webster with PentaRay catheter) in 7 patients. The site of earliest atrial activation shifted after each RF application within and outside the crista terminalis region, indicating a wide distribution of atrial pacemaker sites. We also analyzed 11 patients with dominant pacemakers located outside the crista terminalis (age 27 ± 7 years; five females). In all patients, the rhythm was the dominant pacemaker both at rest and during exercise and located in the right atrial appendage in 6 patients, in the left atrial appendage in 4 patients, and in the mitral annulus in 1 patient. Following ablation, earliest atrial activation shifted to the region of the crista terminalis at a slower rate. CONCLUSIONS Membrane and sub-membrane mechanisms interact to generate cardiac automaticity. The present observations in patients with inappropriate sinus tachycardia and dominant pacemakers are consistent with a wide distribution of pacemaker sites within and outside the boundaries of the crista terminalis.
Collapse
Affiliation(s)
- Hector M Vetulli
- Electrophysiology Department, Sanatorio Otamendi and Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | - Gerald V Naccarelli
- Clinical Electrophysiology, The Milton S. Hershey Medical Center, Penn State University Heart and Vascular Institute, Penn State University College of Medicine, 500 University Dr. Room H 1344K, Hershey, PA, 17033, USA
| | - Mario D Gonzalez
- Clinical Electrophysiology, The Milton S. Hershey Medical Center, Penn State University Heart and Vascular Institute, Penn State University College of Medicine, 500 University Dr. Room H 1344K, Hershey, PA, 17033, USA.
| |
Collapse
|
45
|
Campostrini G, DiFrancesco JC, Castellotti B, Milanesi R, Gnecchi-Ruscone T, Bonzanni M, Bucchi A, Baruscotti M, Ferrarese C, Franceschetti S, Canafoglia L, Ragona F, Freri E, Labate A, Gambardella A, Costa C, Gellera C, Granata T, Barbuti A, DiFrancesco D. A Loss-of-Function HCN4 Mutation Associated With Familial Benign Myoclonic Epilepsy in Infancy Causes Increased Neuronal Excitability. Front Mol Neurosci 2018; 11:269. [PMID: 30127718 PMCID: PMC6089338 DOI: 10.3389/fnmol.2018.00269] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 07/16/2018] [Indexed: 01/03/2023] Open
Abstract
HCN channels are highly expressed and functionally relevant in neurons and increasing evidence demonstrates their involvement in the etiology of human epilepsies. Among HCN isoforms, HCN4 is important in cardiac tissue, where it underlies pacemaker activity. Despite being expressed also in deep structures of the brain, mutations of this channel functionally shown to be associated with epilepsy have not been reported yet. Using Next Generation Sequencing for the screening of patients with idiopathic epilepsy, we identified the p.Arg550Cys (c.1648C>T) heterozygous mutation on HCN4 in two brothers affected by benign myoclonic epilepsy of infancy. Functional characterization in heterologous expression system and in neurons showed that the mutation determines a loss of function of HCN4 contribution to activity and an increase of neuronal discharge, potentially predisposing to epilepsy. Expressed in cardiomyocytes, mutant channels activate at slightly more negative voltages than wild-type (WT), in accordance with borderline bradycardia. While HCN4 variants have been frequently associated with cardiac arrhythmias, these data represent the first experimental evidence that functional alteration of HCN4 can also be involved in human epilepsy through a loss-of-function effect and associated increased neuronal excitability. Since HCN4 appears to be highly expressed in deep brain structures only early during development, our data provide a potential explanation for a link between dysfunctional HCN4 and infantile epilepsy. These findings suggest that it may be useful to include HCN4 screening to extend the knowledge of the genetic causes of infantile epilepsies, potentially paving the way for the identification of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Giulia Campostrini
- Molecular Physiology and Neurobiology, The PaceLab, Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Jacopo C DiFrancesco
- Clinical Neurophysiology and Epilepsy Center, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,Laboratory of Neurobiology, Department of Neurology, Milan Center for Neuroscience, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Barbara Castellotti
- Unit of Genetics of Neurodegenerative and Metabolic Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Raffaella Milanesi
- Molecular Physiology and Neurobiology, The PaceLab, Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | | | - Mattia Bonzanni
- Molecular Physiology and Neurobiology, The PaceLab, Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Annalisa Bucchi
- Molecular Physiology and Neurobiology, The PaceLab, Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Mirko Baruscotti
- Molecular Physiology and Neurobiology, The PaceLab, Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Carlo Ferrarese
- Laboratory of Neurobiology, Department of Neurology, Milan Center for Neuroscience, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Silvana Franceschetti
- Clinical Neurophysiology and Epilepsy Center, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Laura Canafoglia
- Clinical Neurophysiology and Epilepsy Center, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Francesca Ragona
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Elena Freri
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Angelo Labate
- Institute of Neurology, Università degli Studi Magna Græcia di Catanzaro, Catanzaro, Italy
| | - Antonio Gambardella
- Institute of Neurology, Università degli Studi Magna Græcia di Catanzaro, Catanzaro, Italy
| | - Cinzia Costa
- Neurology Unit, Ospedale S. Maria della Misericordia, Department of Medicine, University of Perugia, Perugia, Italy
| | - Cinzia Gellera
- Unit of Genetics of Neurodegenerative and Metabolic Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Tiziana Granata
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Andrea Barbuti
- Molecular Physiology and Neurobiology, The PaceLab, Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Dario DiFrancesco
- Molecular Physiology and Neurobiology, The PaceLab, Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
46
|
Mader F, Müller S, Krause L, Springer A, Kernig K, Protzel C, Porath K, Rackow S, Wittstock T, Frank M, Hakenberg OW, Köhling R, Kirschstein T. Hyperpolarization-Activated Cyclic Nucleotide-Gated Non-selective (HCN) Ion Channels Regulate Human and Murine Urinary Bladder Contractility. Front Physiol 2018; 9:753. [PMID: 29971015 PMCID: PMC6018223 DOI: 10.3389/fphys.2018.00753] [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: 03/12/2018] [Accepted: 05/29/2018] [Indexed: 11/16/2022] Open
Abstract
Purpose: Hyperpolarization-activated cyclic nucleotide gated non-selective (HCN) channels have been demonstrated in the urinary bladder in various species. Since they play a major role in governing rhythmic activity in pacemaker cells like in the sinoatrial node, we explored the role of these channels in human and murine detrusor smooth muscle. Methods: In an organ bath, human and murine detrusor smooth muscle specimens were challenged with the HCN channel blocker ZD7288. In human tissue derived from macroscopically tumor-free cancer resections, the urothelium was removed. In addition, HCN1-deficient mice were used to identify the contribution of this particular isoform. Expression of HCN channels in the urinary bladder was analyzed using histological and ultrastructural analyses as well as quantitative reverse transcriptase polymerase chain reaction (RT-PCR). Results: We found that the HCN channel blocker ZD7288 (50 μM) both induced tonic contractions and increased phasic contraction amplitudes in human and murine detrusor specimens. While these responses were not sensitive to tetrodotoxin, they were significantly reduced by the gap junction inhibitor 18β-glycyrrhetic acid suggesting that HCN channels are located within the gap junction-interconnected smooth muscle cell network rather than on efferent nerve fibers. Immunohistochemistry suggested HCN channel expression on smooth muscle tissue, and immunoelectron microscopy confirmed the scattered presence of HCN2 on smooth muscle cell membranes. HCN channels seem to be down-regulated with aging, which is paralleled by an increasing effect of ZD7288 in aging detrusor tissue. Importantly, the anticonvulsant and HCN channel activator lamotrigine relaxed the detrusor which could be reversed by ZD7288. Conclusion: These findings demonstrate that HCN channels are functionally present and localized on smooth muscle cells of the urinary bladder. Given the age-dependent decline of these channels in humans, activation of HCN channels by compounds such as lamotrigine opens up the opportunity to combat detrusor hyperactivity in the elderly by drugs already approved for epilepsy.
Collapse
Affiliation(s)
- Felix Mader
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Steffen Müller
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Ludwig Krause
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Armin Springer
- Department of Medical Biology, Electron Microscopy Center, University of Rostock, Rostock, Germany
| | - Karoline Kernig
- Department of Urology, University of Rostock, Rostock, Germany
| | - Chris Protzel
- Department of Urology, University of Rostock, Rostock, Germany
| | - Katrin Porath
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Simone Rackow
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Tristan Wittstock
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Marcus Frank
- Department of Medical Biology, Electron Microscopy Center, University of Rostock, Rostock, Germany
| | | | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Timo Kirschstein
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| |
Collapse
|
47
|
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]
|
48
|
Abstract
Electronic pacemakers have been used in patients with heart rhythm disorders for device-supported pacing. While effective, there are such shortcomings as limited battery life, permanent implantation of catheters, the lack of autonomic neurohumoral responses, and risks of lead dislodging. Here we describe protocols for establishing porcine models of sick sinus syndrome and complete heart block, and the generation of bioartificial pacemaker by delivering a strategically engineered form of hyperpolarization-activated cyclic nucleotide-gated pacemaker channel protein via somatic gene transfer to convert atrial or ventricular muscle cardiomyocytes into nodal-like cells that rhythmically fire action potentials.
Collapse
|
49
|
AAV-mediated conversion of human pluripotent stem cell-derived pacemaker. Biochem Biophys Res Commun 2017; 494:346-351. [DOI: 10.1016/j.bbrc.2017.10.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 10/04/2017] [Indexed: 12/21/2022]
|
50
|
Zhao X, Gu T. Dysfunctional Hyperpolarization-Activated Cyclic Nucleotide-gated Ion Channels in Cardiac Diseases. Braz J Cardiovasc Surg 2017; 31:203-6. [PMID: 27556324 PMCID: PMC5062718 DOI: 10.5935/1678-9741.20160030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are reverse
voltage-dependent, and their activation depends on the hyperpolarization of the
membrane and may be directly or indirectly regulated by the cyclic adenosine
monophosphate (cAMP) or other signal-transduction cascades. The distribution,
quantity and activation states of HCN channels differ in tissues throughout the
body. Evidence exhibits that HCN channels play critical roles in the generation
and conduction of the electrical impulse and the physiopathological process of
some cardiac diseases. They may constitute promising drug targets in the
treatment of these cardiac diseases. Pharmacological treatment targeting HCN
channels is of benefit to these cardiac conditions.
Collapse
Affiliation(s)
- Xiaoqi Zhao
- Department of Cardiac Surgery ICU, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Tianxiang Gu
- Department of Cardiac Surgery ICU, First Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|