1
|
Clancy CE, Santana LF. Advances in induced pluripotent stem cell-derived cardiac myocytes: technological breakthroughs, key discoveries and new applications. J Physiol 2024; 602:3871-3892. [PMID: 39032073 PMCID: PMC11326976 DOI: 10.1113/jp282562] [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: 02/21/2024] [Accepted: 07/02/2024] [Indexed: 07/22/2024] Open
Abstract
A transformation is underway in precision and patient-specific medicine. Rapid progress has been enabled by multiple new technologies including induced pluripotent stem cell-derived cardiac myocytes (iPSC-CMs). Here, we delve into these advancements and their future promise, focusing on the efficiency of reprogramming techniques, the fidelity of differentiation into the cardiac lineage, the functional characterization of the resulting cardiac myocytes, and the many applications of in silico models to understand general and patient-specific mechanisms controlling excitation-contraction coupling in health and disease. Furthermore, we explore the current and potential applications of iPSC-CMs in both research and clinical settings, underscoring the far-reaching implications of this rapidly evolving field.
Collapse
Affiliation(s)
- Colleen E Clancy
- Department of Physiology & Membrane Biology, School of Medicine, University of California Davis, Davis, CA, USA
- Center for Precision Medicine and Data Sciences, University of California Davis, School of Medicine, Sacramento, CA, USA
| | - L Fernando Santana
- Department of Physiology & Membrane Biology, School of Medicine, University of California Davis, Davis, CA, USA
- Center for Precision Medicine and Data Sciences, University of California Davis, School of Medicine, Sacramento, CA, USA
| |
Collapse
|
2
|
Verkerk AO, Wilders R. Injection of I K1 through dynamic clamp can make all the difference in patch-clamp studies on hiPSC-derived cardiomyocytes. Front Physiol 2023; 14:1326160. [PMID: 38152247 PMCID: PMC10751953 DOI: 10.3389/fphys.2023.1326160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 11/24/2023] [Indexed: 12/29/2023] Open
Abstract
Human-induced stem cell-derived cardiomyocytes (hiPSC-CMs) are a valuable tool for studying development, pharmacology, and (inherited) arrhythmias. Unfortunately, hiPSC-CMs are depolarized and spontaneously active, even the working cardiomyocyte subtypes such as atrial- and ventricular-like hiPSC-CMs, in contrast to the situation in the atria and ventricles of adult human hearts. Great efforts have been made, using many different strategies, to generate more mature, quiescent hiPSC-CMs with more close-to-physiological resting membrane potentials, but despite promising results, it is still difficult to obtain hiPSC-CMs with such properties. The dynamic clamp technique allows to inject a current with characteristics of the inward rectifier potassium current (IK1), computed in real time according to the actual membrane potential, into patch-clamped hiPSC-CMs during action potential measurements. This results in quiescent hiPSC-CMs with a close-to-physiological resting membrane potential. As a result, action potential measurements can be performed with normal ion channel availability, which is particularly important for the physiological functioning of the cardiac SCN5A-encoded fast sodium current (INa). We performed in vitro and in silico experiments to assess the beneficial effects of the dynamic clamp technique in dissecting the functional consequences of the SCN5A-1795insD+/- mutation. In two separate sets of patch-clamp experiments on control hiPSC-CMs and on hiPSC-CMs with mutations in ACADVL and GNB5, we assessed the value of dynamic clamp in detecting delayed afterdepolarizations and in investigating factors that modulate the resting membrane potential. We conclude that the dynamic clamp technique has highly beneficial effects in all of the aforementioned settings and should be widely used in patch-clamp studies on hiPSC-CMs while waiting for the ultimate fully mature hiPSC-CMs.
Collapse
Affiliation(s)
- Arie O. Verkerk
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
3
|
Han JL, Heinson YW, Chua CJ, Liu W, Entcheva E. CRISPRi gene modulation and all-optical electrophysiology in post-differentiated human iPSC-cardiomyocytes. Commun Biol 2023; 6:1236. [PMID: 38062109 PMCID: PMC10703822 DOI: 10.1038/s42003-023-05627-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Uncovering gene-phenotype relationships can be enabled by precise gene modulation in human induced pluripotent stem-cell-derived cardiomyocytes (iPSC-CMs) and follow up phenotyping using scalable all-optical electrophysiology platforms. Such efforts towards human functional genomics can be aided by recent CRISPR-derived technologies for reversible gene inhibition or activation (CRISPRi/a). We set out to characterize the performance of CRISPRi in post-differentiated iPSC-CMs, targeting key cardiac ion channel genes, KCNH2, KCNJ2, and GJA1, and providing a multiparametric quantification of the effects on cardiac repolarization, stability of the resting membrane potential and conduction properties using all-optical tools. More potent CRISPRi effectors, e.g., Zim3, and optimized viral delivery led to improved performance on par with the use of CRISPRi iPSC lines. Confirmed mild yet specific phenotype changes when CRISPRi is deployed in non-dividing differentiated heart cells is an important step towards more holistic pre-clinical cardiotoxicity testing and for future therapeutic use in vivo.
Collapse
Affiliation(s)
- Julie L Han
- Department of Biomedical Engineering, The George Washington University, Washington, DC, 20052, USA
| | - Yuli W Heinson
- Department of Biomedical Engineering, The George Washington University, Washington, DC, 20052, USA
| | - Christianne J Chua
- Department of Biomedical Engineering, The George Washington University, Washington, DC, 20052, USA
| | - Wei Liu
- Department of Biomedical Engineering, The George Washington University, Washington, DC, 20052, USA
| | - Emilia Entcheva
- Department of Biomedical Engineering, The George Washington University, Washington, DC, 20052, USA.
| |
Collapse
|
4
|
Chapotte-Baldacci CA, Pierre M, Djemai M, Pouliot V, Chahine M. Biophysical properties of Na V1.5 channels from atrial-like and ventricular-like cardiomyocytes derived from human induced pluripotent stem cells. Sci Rep 2023; 13:20685. [PMID: 38001331 PMCID: PMC10673932 DOI: 10.1038/s41598-023-47310-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 11/12/2023] [Indexed: 11/26/2023] Open
Abstract
Generating atrial-like cardiomyocytes derived from human induced pluripotent stem cells (hiPSCs) is crucial for modeling and treating atrial-related diseases, such as atrial arrythmias including atrial fibrillations. However, it is essential to obtain a comprehensive understanding of the electrophysiological properties of these cells. The objective of the present study was to investigate the molecular, electrical, and biophysical properties of several ion channels, especially NaV1.5 channels, in atrial hiPSC cardiomyocytes. Atrial cardiomyocytes were obtained by the differentiation of hiPSCs treated with retinoic acid (RA). The quality of the atrial specification was assessed by qPCR, immunocytofluorescence, and western blotting. The electrophysiological properties of action potentials (APs), Ca2+ dynamics, K+ and Na+ currents were investigated using patch-clamp and optical mapping approaches. We evaluated mRNA transcript and protein expressions to show that atrial cardiomyocytes expressed higher atrial- and sinoatrial-specific markers (MYL7, CACNA1D) and lower ventricular-specific markers (MYL2, CACNA1C, GJA1) than ventricular cardiomyocytes. The amplitude, duration, and steady-state phase of APs in atrial cardiomyocytes decreased, and had a shape similar to that of mature atrial cardiomyocytes. Interestingly, NaV1.5 channels in atrial cardiomyocytes exhibited lower mRNA transcripts and protein expression, which could explain the lower current densities recorded by patch-clamp. Moreover, Na+ currents exhibited differences in activation and inactivation parameters. These differences could be explained by an increase in SCN2B regulatory subunit expression and a decrease in SCN1B and SCN4B regulatory subunit expressions. Our results show that a RA treatment made it possible to obtain atrial cardiomyocytes and investigate differences in NaV1.5 channel properties between ventricular- and atrial-like cells.
Collapse
Affiliation(s)
- Charles-Albert Chapotte-Baldacci
- Department of Medicine, Laval University, Quebec City, QC, Canada
- CERVO Brain Research Centre, 2601, chemin de la Canardière, Quebec City, QC, G1J 2G3, Canada
| | - Marion Pierre
- Department of Medicine, Laval University, Quebec City, QC, Canada
- CERVO Brain Research Centre, 2601, chemin de la Canardière, Quebec City, QC, G1J 2G3, Canada
| | - Mohammed Djemai
- Department of Medicine, Laval University, Quebec City, QC, Canada
- CERVO Brain Research Centre, 2601, chemin de la Canardière, Quebec City, QC, G1J 2G3, Canada
| | - Valérie Pouliot
- CERVO Brain Research Centre, 2601, chemin de la Canardière, Quebec City, QC, G1J 2G3, Canada
| | - Mohamed Chahine
- CERVO Brain Research Centre, 2601, chemin de la Canardière, Quebec City, QC, G1J 2G3, Canada.
| |
Collapse
|
5
|
Hermanstyne TO, Yang ND, Granados-Fuentes D, Li X, Mellor RL, Jegla T, Herzog ED, Nerbonne JM. Kv12-encoded K+ channels drive the day-night switch in the repetitive firing rates of SCN neurons. J Gen Physiol 2023; 155:e202213310. [PMID: 37516908 PMCID: PMC10373311 DOI: 10.1085/jgp.202213310] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/11/2023] [Accepted: 07/06/2023] [Indexed: 07/31/2023] Open
Abstract
Considerable evidence suggests that day-night rhythms in the functional expression of subthreshold potassium (K+) channels regulate daily oscillations in the spontaneous firing rates of neurons in the suprachiasmatic nucleus (SCN), the master circadian pacemaker in mammals. The K+ conductance(s) driving these daily rhythms in the repetitive firing rates of SCN neurons, however, have not been identified. To test the hypothesis that subthreshold Kv12.1/Kv12.2-encoded K+ channels play a role, we obtained current-clamp recordings from SCN neurons in slices prepared from adult mice harboring targeted disruptions in the Kcnh8 (Kv12.1-/-) or Kcnh3 (Kv12.2-/-) locus. We found that mean nighttime repetitive firing rates were higher in Kv12.1-/- and Kv12.2-/- than in wild type (WT), SCN neurons. In marked contrast, mean daytime repetitive firing rates were similar in Kv12.1-/-, Kv12.2-/-, and WT SCN neurons, and the day-night difference in mean repetitive firing rates, a hallmark feature of WT SCN neurons, was eliminated in Kv12.1-/- and Kv12.2-/- SCN neurons. Similar results were obtained with in vivo shRNA-mediated acute knockdown of Kv12.1 or Kv12.2 in adult SCN neurons. Voltage-clamp experiments revealed that Kv12-encoded current densities in WT SCN neurons are higher at night than during the day. In addition, the pharmacological block of Kv12-encoded currents increased the mean repetitive firing rate of nighttime, but not daytime, in WT SCN neurons. Dynamic clamp-mediated subtraction of modeled Kv12-encoded currents also selectively increased the mean repetitive firing rates of nighttime WT SCN neurons. Despite the elimination of the nighttime decrease in the mean repetitive firing rates of SCN neurons, however, locomotor (wheel-running) activity remained rhythmic in Kv12.1-/-, Kv12.2-/-, and Kv12.1-targeted shRNA-expressing, and Kv12.2-targeted shRNA-expressing animals.
Collapse
Affiliation(s)
- Tracey O. Hermanstyne
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Nien-Du Yang
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | | | - Xiaofan Li
- Department of Biology, The Pennsylvania State University, University Park, State College, PA, USA
| | - Rebecca L. Mellor
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Timothy Jegla
- Department of Biology, The Pennsylvania State University, University Park, State College, PA, USA
| | - Erik D. Herzog
- Department of Biology, Washington University, St. Louis, MO, USA
| | - Jeanne M. Nerbonne
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| |
Collapse
|
6
|
Yang ND, Mellor RL, Hermanstyne TO, Nerbonne JM. Effects of NALCN-Encoded Na + Leak Currents on the Repetitive Firing Properties of SCN Neurons Depend on K +-Driven Rhythmic Changes in Input Resistance. J Neurosci 2023; 43:5132-5141. [PMID: 37339878 PMCID: PMC10342223 DOI: 10.1523/jneurosci.0182-23.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 06/02/2023] [Accepted: 06/12/2023] [Indexed: 06/22/2023] Open
Abstract
Neurons in the suprachiasmatic nucleus (SCN) generate circadian changes in the rates of spontaneous action potential firing that regulate and synchronize daily rhythms in physiology and behavior. Considerable evidence suggests that daily rhythms in the repetitive firing rates (higher during the day than at night) of SCN neurons are mediated by changes in subthreshold potassium (K+) conductance(s). An alternative "bicycle" model for circadian regulation of membrane excitability in clock neurons, however, suggests that an increase in NALCN-encoded sodium (Na+) leak conductance underlies daytime increases in firing rates. The experiments reported here explored the role of Na+ leak currents in regulating daytime and nighttime repetitive firing rates in identified adult male and female mouse SCN neurons: vasoactive intestinal peptide-expressing (VIP+), neuromedin S-expressing (NMS+) and gastrin-releasing peptide-expressing (GRP+) cells. Whole-cell recordings from VIP+, NMS+, and GRP+ neurons in acute SCN slices revealed that Na+ leak current amplitudes/densities are similar during the day and at night, but have a larger impact on membrane potentials in daytime neurons. Additional experiments, using an in vivo conditional knockout approach, demonstrated that NALCN-encoded Na+ currents selectively regulate daytime repetitive firing rates of adult SCN neurons. Dynamic clamp-mediated manipulation revealed that the effects of NALCN-encoded Na+ currents on the repetitive firing rates of SCN neurons depend on K+ current-driven changes in input resistances. Together, these findings demonstrate that NALCN-encoded Na+ leak channels contribute to regulating daily rhythms in the excitability of SCN neurons by a mechanism that depends on K+ current-mediated rhythmic changes in intrinsic membrane properties.SIGNIFICANCE STATEMENT Elucidating the ionic mechanisms responsible for generating daily rhythms in the rates of spontaneous action potential firing of neurons in the suprachiasmatic nucleus (SCN), the master circadian pacemaker in mammals, is an important step toward understanding how the molecular clock controls circadian rhythms in physiology and behavior. While numerous studies have focused on identifying subthreshold K+ channel(s) that mediate day-night changes in the firing rates of SCN neurons, a role for Na+ leak currents has also been suggested. The results of the experiments presented here demonstrate that NALCN-encoded Na+ leak currents differentially modulate daily rhythms in the daytime/nighttime repetitive firing rates of SCN neurons as a consequence of rhythmic changes in subthreshold K+ currents.
Collapse
Affiliation(s)
- Nien-Du Yang
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri 63110
| | | | - Tracey O Hermanstyne
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Jeanne M Nerbonne
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri 63110
- Department of Medicine, Cardiovascular Division
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri 63110
| |
Collapse
|
7
|
Terrar DA. Timing mechanisms to control heart rhythm and initiate arrhythmias: roles for intracellular organelles, signalling pathways and subsarcolemmal Ca 2. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220170. [PMID: 37122228 PMCID: PMC10150226 DOI: 10.1098/rstb.2022.0170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Rhythms of electrical activity in all regions of the heart can be influenced by a variety of intracellular membrane bound organelles. This is true both for normal pacemaker activity and for abnormal rhythms including those caused by early and delayed afterdepolarizations under pathological conditions. The influence of the sarcoplasmic reticulum (SR) on cardiac electrical activity is widely recognized, but other intracellular organelles including lysosomes and mitochondria also contribute. Intracellular organelles can provide a timing mechanism (such as an SR clock driven by cyclic uptake and release of Ca2+, with an important influence of intraluminal Ca2+), and/or can act as a Ca2+ store involved in signalling mechanisms. Ca2+ plays many diverse roles including carrying electric current, driving electrogenic sodium-calcium exchange (NCX) particularly when Ca2+ is extruded across the surface membrane causing depolarization, and activation of enzymes which target organelles and surface membrane proteins. Heart function is also influenced by Ca2+ mobilizing agents (cADP-ribose, nicotinic acid adenine dinucleotide phosphate and inositol trisphosphate) acting on intracellular organelles. Lysosomal Ca2+ release exerts its effects via calcium/calmodulin-dependent protein kinase II to promote SR Ca2+ uptake, and contributes to arrhythmias resulting from excessive beta-adrenoceptor stimulation. A separate arrhythmogenic mechanism involves lysosomes, mitochondria and SR. Interacting intracellular organelles, therefore, have profound effects on heart rhythms and NCX plays a central role. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.
Collapse
Affiliation(s)
- Derek A Terrar
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| |
Collapse
|
8
|
Seibertz F, Sutanto H, Dülk R, Pronto JRD, Springer R, Rapedius M, Liutkute A, Ritter M, Jung P, Stelzer L, Hüsgen LM, Klopp M, Rubio T, Fakuade FE, Mason FE, Hartmann N, Pabel S, Streckfuss-Bömeke K, Cyganek L, Sossalla S, Heijman J, Voigt N. Electrophysiological and calcium-handling development during long-term culture of human-induced pluripotent stem cell-derived cardiomyocytes. Basic Res Cardiol 2023; 118:14. [PMID: 37020075 PMCID: PMC10076390 DOI: 10.1007/s00395-022-00973-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 04/07/2023]
Abstract
Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are increasingly used for personalised medicine and preclinical cardiotoxicity testing. Reports on hiPSC-CM commonly describe heterogenous functional readouts and underdeveloped or immature phenotypical properties. Cost-effective, fully defined monolayer culture is approaching mainstream adoption; however, the optimal age at which to utilise hiPSC-CM is unknown. In this study, we identify, track and model the dynamic developmental behaviour of key ionic currents and Ca2+-handling properties in hiPSC-CM over long-term culture (30-80 days). hiPSC-CMs > 50 days post differentiation show significantly larger ICa,L density along with an increased ICa,L-triggered Ca2+-transient. INa and IK1 densities significantly increase in late-stage cells, contributing to increased upstroke velocity and reduced action potential duration, respectively. Importantly, our in silico model of hiPSC-CM electrophysiological age dependence confirmed IK1 as the key ionic determinant of action potential shortening in older cells. We have made this model available through an open source software interface that easily allows users to simulate hiPSC-CM electrophysiology and Ca2+-handling and select the appropriate age range for their parameter of interest. This tool, together with the insights from our comprehensive experimental characterisation, could be useful in future optimisation of the culture-to-characterisation pipeline in the field of hiPSC-CM research.
Collapse
Affiliation(s)
- Fitzwilliam Seibertz
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Henry Sutanto
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Rebekka Dülk
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Julius Ryan D Pronto
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Robin Springer
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | | | - Aiste Liutkute
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Melanie Ritter
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Philipp Jung
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Lea Stelzer
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Luisa M Hüsgen
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Marie Klopp
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Tony Rubio
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Funsho E Fakuade
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Fleur E Mason
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Nico Hartmann
- Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University Göttingen, Göttingen, Germany
| | - Steffen Pabel
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany
| | - Katrin Streckfuss-Bömeke
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University Göttingen, Göttingen, Germany
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - Lukas Cyganek
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
- Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University Göttingen, Göttingen, Germany
| | - Samuel Sossalla
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University Göttingen, Göttingen, Germany
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany
| | - Jordi Heijman
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands.
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany.
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
9
|
Kodirov SA. Whole-cell patch-clamp recording and parameters. Biophys Rev 2023; 15:257-288. [PMID: 37124922 PMCID: PMC10133435 DOI: 10.1007/s12551-023-01055-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/22/2023] [Indexed: 05/02/2023] Open
Abstract
The patch-clamp technique represents an electrophysiology type of method. This is one of several insightful approaches with five major configurations, namely a loose patch, cell-attached (also known as on-cell), whole-cell, inside-out, and outside-out modes. The patch-clamp method is more advanced compared to classical electrophysiology since it elucidates single-channel activation in a tiny portion of the membrane in addition to action potential (AP), junction potential (JP), endplate potential (EP), electrical coupling between two adjacent cells via Gap junction hemi-channels, excitatory/inhibitory postsynaptic potentials, and resting membrane potential (RMP). In fact, a malfunction of only one channel or even one component will alter AP amplitude or duration in vitro. If parameters are inferred appropriately and recordings are performed properly, the patch-clamp trace readouts and results are robust. The main hallmarks of currents via voltage-dependent calcium (Cav), hyperpolarization-activated cyclic nucleotide gated non-selective cation (HCN), inwardly rectifying potassium (Kir), voltage-dependent potassium (Kv), and voltage-dependent sodium (Nav) channels are similar and tractable among cells even when they are derived from evolutionary distinct organs and species. However, the size of the membrane area, where the functional subunits reside, and current magnitudes vary among cells of the same type. Therefore, dividing current magnitudes by cell capacitance- current density enables the estimate of functional and active channels relative to recorded cytoplasmic membrane area. Since the patch-clamp recordings can be performed in both current- and voltage-clamp modes, the action potential or spike durations can be adequately elucidated. Sometimes, optical methods are preferred to patch-clamp electrophysiology, but the obtained signals and traces are not robust. Finally, not only an alternans of AP durations, but also that of 'action potential shape' is observed with electrophysiology.
Collapse
Affiliation(s)
- Sodikdjon A. Kodirov
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
- Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
- Almazov Federal Medical Research Centre, Saint Petersburg, 197341 Russia
- Institute for Physiology and Pathophysiology, Johannes Kepler University, Linz, Austria
| |
Collapse
|
10
|
Altomare C, Bartolucci C, Sala L, Balbi C, Burrello J, Pietrogiovanna N, Burrello A, Bolis S, Panella S, Arici M, Krause R, Rocchetti M, Severi S, Barile L. A dynamic clamping approach using in silico IK1 current for discrimination of chamber-specific hiPSC-derived cardiomyocytes. Commun Biol 2023; 6:291. [PMID: 36934210 PMCID: PMC10024709 DOI: 10.1038/s42003-023-04674-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 03/07/2023] [Indexed: 03/20/2023] Open
Abstract
Human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CM) constitute a mixed population of ventricular-, atrial-, nodal-like cells, limiting the reliability for studying chamber-specific disease mechanisms. Previous studies characterised CM phenotype based on action potential (AP) morphology, but the classification criteria were still undefined. Our aim was to use in silico models to develop an automated approach for discriminating the electrophysiological differences between hiPSC-CM. We propose the dynamic clamp (DC) technique with the injection of a specific IK1 current as a tool for deriving nine electrical biomarkers and blindly classifying differentiated CM. An unsupervised learning algorithm was applied to discriminate CM phenotypes and principal component analysis was used to visualise cell clustering. Pharmacological validation was performed by specific ion channel blocker and receptor agonist. The proposed approach improves the translational relevance of the hiPSC-CM model for studying mechanisms underlying inherited or acquired atrial arrhythmias in human CM, and for screening anti-arrhythmic agents.
Collapse
Affiliation(s)
- Claudia Altomare
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Euler institute, Università Svizzera italiana, Lugano, Switzerland
| | - Chiara Bartolucci
- Department of Electrical, Electronic and Information Engineering 'Guglielmo Marconi', University of Bologna, Cesena, Italy
| | - Luca Sala
- Istituto Auxologico Italiano IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milan, Italy
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Carolina Balbi
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Jacopo Burrello
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Division of Internal Medicine 4 and Hypertension Unit, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Nicole Pietrogiovanna
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Alessio Burrello
- Department of Electrical, Electronic and Information Engineering (DEI), University of Bologna, Bologna, Italy
| | - Sara Bolis
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Stefano Panella
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Martina Arici
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Rolf Krause
- Euler institute, Università Svizzera italiana, Lugano, Switzerland
| | - Marcella Rocchetti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Stefano Severi
- Department of Electrical, Electronic and Information Engineering 'Guglielmo Marconi', University of Bologna, Cesena, Italy.
| | - Lucio Barile
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, Switzerland.
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.
- Euler institute, Università Svizzera italiana, Lugano, Switzerland.
- Faculty of Biomedical Sciences, Università Svizzera italiana, Lugano, Switzerland.
- Institute of Life Science, Scuola Superiore Sant'Anna, Pisa, Italy.
| |
Collapse
|
11
|
Hermanstyne TO, Yang ND, Granados-Fuentes D, Li X, Mellor RL, Jegla T, Herzog ED, Nerbonne JM. Kv12-Encoded K + Channels Drive the Day-Night Switch in the Repetitive Firing Rates of SCN Neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.30.526323. [PMID: 36778242 PMCID: PMC9915524 DOI: 10.1101/2023.01.30.526323] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Considerable evidence suggests that day-night rhythms in the functional expression of subthreshold potassium (K + ) channels regulate daily oscillations in the rates of spontaneous action potential firing of neurons in the suprachiasmatic nucleus (SCN), the master circadian pacemaker in mammals. The K + conductance(s) driving these daily rhythms in repetitive firing rates, however, have not been identified. To test the hypothesis that subthreshold Kv12.1/Kv12.2-encoded K + channels play a role, we obtained current-clamp recordings from SCN neurons in slices prepared from adult mice harboring targeted disruptions in the Kcnh8 (Kv12.1 -/- ) or Kcnh3 (Kv12.2 -/- ) locus. We found that mean nighttime repetitive firing rates were higher in Kv12.1 -/- and Kv12.2 -/- , than in wild type (WT), SCN neurons. In marked contrast, mean daytime repetitive firing rates were similar in Kv12.1 -/- , Kv12.2 -/- and WT SCN neurons, and the day-night difference in mean repetitive firing rates, a hallmark feature of WT SCN neurons, was eliminated in Kv12.1 -/- and Kv12.2 -/- SCN neurons. Similar results were obtained with in vivo shRNA-mediated acute knockdown of Kv12.1 or Kv12.2 in adult SCN neurons. Voltage-clamp experiments revealed that Kv12-encoded current densities in WT SCN neurons are higher at night than during the day. In addition, pharmacological block of Kv12-encoded currents increased the mean repetitive firing rate of nighttime, but not daytime, in WT SCN neurons. Dynamic clamp-mediated subtraction of modeled Kv12-encoded currents also selectively increased the mean repetitive firing rates of nighttime WT SCN neurons. Despite the elimination of nighttime decrease in the mean repetitive firing rates of SCN neurons, however, locomotor (wheel-running) activity remained rhythmic in Kv12.1 -/- , Kv12.2 -/- , Kv12.1-targeted shRNA-expressing, and Kv12.2-targeted shRNA-expressing animals.
Collapse
Affiliation(s)
- Tracey O. Hermanstyne
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO
| | - Nien-Du Yang
- Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO
- Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, MO
| | | | - Xiaofan Li
- Department of Biology, The Pennsylvania State University, University Park, PA
| | - Rebecca L. Mellor
- Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO
| | - Timothy Jegla
- Department of Biology, The Pennsylvania State University, University Park, PA
| | - Erik D. Herzog
- Department of Biology, Washington University, St. Louis, MO
| | - Jeanne M. Nerbonne
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO
- Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO
- Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
12
|
Verkerk AO, Doszpod IJ, Mengarelli I, Magyar T, Polyák A, Pászti B, Efimov IR, Wilders R, Koncz I. Acetylcholine Reduces L-Type Calcium Current without Major Changes in Repolarization of Canine and Human Purkinje and Ventricular Tissue. Biomedicines 2022; 10:biomedicines10112987. [PMID: 36428555 PMCID: PMC9687254 DOI: 10.3390/biomedicines10112987] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/11/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
Vagal nerve stimulation (VNS) holds a strong basis as a potentially effective treatment modality for chronic heart failure, which explains why a multicenter VNS study in heart failure with reduced ejection fraction is ongoing. However, more detailed information is required on the effect of acetylcholine (ACh) on repolarization in Purkinje and ventricular cardiac preparations to identify the advantages, risks, and underlying cellular mechanisms of VNS. Here, we studied the effect of ACh on the action potential (AP) of canine Purkinje fibers (PFs) and several human ventricular preparations. In addition, we characterized the effects of ACh on the L-type Ca2+ current (ICaL) and AP of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and performed computer simulations to explain the observed effects. Using microelectrode recordings, we found a small but significant AP prolongation in canine PFs. In the human myocardium, ACh slightly prolonged the AP in the midmyocardium but resulted in minor AP shortening in subepicardial tissue. Perforated patch-clamp experiments on hiPSC-CMs demonstrated that 5 µM ACh caused an ≈15% decrease in ICaL density without changes in gating properties. Using dynamic clamp, we found that under blocked K+ currents, 5 µM ACh resulted in an ≈23% decrease in AP duration at 90% of repolarization in hiPSC-CMs. Computer simulations using the O'Hara-Rudy human ventricular cell model revealed that the overall effect of ACh on AP duration is a tight interplay between the ACh-induced reduction in ICaL and ACh-induced changes in K+ currents. In conclusion, ACh results in minor changes in AP repolarization and duration of canine PFs and human ventricular myocardium due to the concomitant inhibition of inward ICaL and outward K+ currents, which limits changes in net repolarizing current and thus prevents major changes in AP repolarization.
Collapse
Affiliation(s)
- Arie O. Verkerk
- Department of Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Illés J. Doszpod
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary
| | - Isabella Mengarelli
- Department of Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Tibor Magyar
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary
| | - Alexandra Polyák
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary
| | - Bence Pászti
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary
| | - Igor R. Efimov
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA
- Department of Biomedical Engineering, Northwestern University, Chicago, IL 60611, USA
- Department of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Correspondence: (R.W.); (I.K.)
| | - István Koncz
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA
- Correspondence: (R.W.); (I.K.)
| |
Collapse
|
13
|
Fassina D, Costa CM, Longobardi S, Karabelas E, Plank G, Harding SE, Niederer SA. Modelling the interaction between stem cells derived cardiomyocytes patches and host myocardium to aid non-arrhythmic engineered heart tissue design. PLoS Comput Biol 2022; 18:e1010030. [PMID: 35363778 PMCID: PMC9007348 DOI: 10.1371/journal.pcbi.1010030] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 04/13/2022] [Accepted: 03/17/2022] [Indexed: 11/18/2022] Open
Abstract
Application of epicardial patches constructed from human-induced pluripotent stem cell- derived cardiomyocytes (hiPSC-CMs) has been proposed as a long-term therapy to treat scarred hearts post myocardial infarction (MI). Understanding electrical interaction between engineered heart tissue patches (EHT) and host myocardium represents a key step toward a successful patch engraftment. EHT retain different electrical properties with respect to the host heart tissue due to the hiPSC-CMs immature phenotype, which may lead to increased arrhythmia risk. We developed a modelling framework to examine the influence of patch design on electrical activation at the engraftment site. We performed an in silico investigation of different patch design approaches to restore pre-MI activation properties and evaluated the associated arrhythmic risk. We developed an in silico cardiac electrophysiology model of a transmural cross section of host myocardium. The model featured an infarct region, an epicardial patch spanning the infarct region and a bath region. The patch is modelled as a layer of hiPSC-CM, combined with a layer of conductive polymer (CP). Tissue and patch geometrical dimensions and conductivities were incorporated through 10 modifiable model parameters. We validated our model against 4 independent experimental studies and showed that it can qualitatively reproduce their findings. We performed a global sensitivity analysis (GSA) to isolate the most important parameters, showing that the stimulus propagation is mainly governed by the scar depth, radius and conductivity when the scar is not transmural, and by the EHT patch conductivity when the scar is transmural. We assessed the relevance of small animal studies to humans by comparing simulations of rat, rabbit and human myocardium. We found that stimulus propagation paths and GSA sensitivity indices are consistent across species. We explored which EHT design variables have the potential to restore physiological propagation. Simulations predict that increasing EHT conductivity from 0.28 to 1-1.1 S/m recovered physiological activation in rat, rabbit and human. Finally, we assessed arrhythmia risk related to increasing EHT conductivity and tested increasing the EHT Na+ channel density as an alternative strategy to match healthy activation. Our results revealed a greater arrhythmia risk linked to increased EHT conductivity compared to increased Na+ channel density. We demonstrated that our modeling framework could capture the interaction between host and EHT patches observed in in vitro experiments. We showed that large (patch and tissue dimensions) and small (cardiac myocyte electrophysiology) scale differences between small animals and humans do not alter EHT patch effect on infarcted tissue. Our model revealed that only when the scar is transmural do EHT properties impact activation times and isolated the EHT conductivity as the main parameter influencing propagation. We predicted that restoring physiological activation by tuning EHT conductivity is possible but may promote arrhythmic behavior. Finally, our model suggests that acting on hiPSC-CMs low action potential upstroke velocity and lack of IK1 may restore pre-MI activation while not promoting arrhythmia.
Collapse
Affiliation(s)
- Damiano Fassina
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Caroline M. Costa
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, United Kingdom
| | - Stefano Longobardi
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, United Kingdom
| | - Elias Karabelas
- Institute of Mathematics & Scientific Computing, University of Graz, Graz, Austria
| | - Gernot Plank
- Gottfried Schatz Research Center (for Cell Signaling, Metabolism and Aging), Division Biophysics, Medical University of Graz, Graz, Austria
| | - Sian E. Harding
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Steven A. Niederer
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, United Kingdom
| |
Collapse
|
14
|
Nijak A, Simons E, Vandendriessche B, Van de Sande D, Fransen E, Sieliwończyk E, Van Gucht I, Van Craenenbroeck E, Saenen J, Heidbuchel H, Ponsaerts P, Labro AJ, Snyders D, De Vos W, Schepers D, Alaerts M, Loeys BL. Morpho-functional comparison of differentiation protocols to create iPSC-derived cardiomyocytes. Biol Open 2022; 11:274508. [PMID: 35195246 PMCID: PMC8890088 DOI: 10.1242/bio.059016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/15/2021] [Indexed: 11/30/2022] Open
Abstract
Cardiomyocytes derived from induced pluripotent stem cells (iPSC-CMs) offer an attractive platform for cardiovascular research. Patient-specific iPSC-CMs are very useful for studying disease development, and bear potential for disease diagnostics, prognosis evaluation and development of personalized treatment. Several monolayer-based serum-free protocols have been described for the differentiation of iPSCs into cardiomyocytes, but data on their performance are scarce. In this study, we evaluated two protocols that are based on temporal modulation of the Wnt/β-catenin pathway for iPSC-CM differentiation from four iPSC lines, including two control individuals and two patients carrying an SCN5A mutation. The SCN5A gene encodes the cardiac voltage-gated sodium channel (Nav1.5) and loss-of-function mutations can cause the cardiac arrhythmia Brugada syndrome. We performed molecular characterization of the obtained iPSC-CMs by immunostaining for cardiac specific markers and by expression analysis of selected cardiac structural and ionic channel protein-encoding genes with qPCR. We also investigated cell growth morphology, contractility and survival of the iPSC-CMs after dissociation. Finally, we performed electrophysiological characterization of the cells, focusing on the action potential (AP) and calcium transient (CT) characteristics using patch-clamping and optical imaging, respectively. Based on our comprehensive morpho-functional analysis, we concluded that both tested protocols result in a high percentage of contracting CMs. Moreover, they showed acceptable survival and cell quality after dissociation (>50% of cells with a smooth cell membrane, possible to seal during patch-clamping). Both protocols generated cells presenting with typical iPSC-CM AP and CT characteristics, although one protocol (that involves sequential addition of CHIR99021 and Wnt-C59) rendered iPSC-CMs, which were more accessible for patch-clamp and calcium transient experiments and showed an expression pattern of cardiac-specific markers more similar to this observed in human heart left ventricle samples. Summary: In this study, we evaluated two protocols that are based on temporal modulation of the Wnt/β -catenin pathway for iPSC-CM differentiation from four iPSC lines. We show that both protocols were successful in the generation of contracting iPSC-CMs. However, one of the tested protocols rendered cells that were more accessible for patch-clamp experiments and showed an expression pattern of cardiac-specific markers more similar to this of human heart left ventricle samples.
Collapse
Affiliation(s)
- Aleksandra Nijak
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp & Antwerp University Hospital, Antwerp 2650, Belgium
| | - Eline Simons
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp & Antwerp University Hospital, Antwerp 2650, Belgium
| | - Bert Vandendriessche
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp & Antwerp University Hospital, Antwerp 2650, Belgium
| | - Dieter Van de Sande
- Laboratory of Molecular Biophysics, Cellular and Network Excitability, Department of Biomedical Sciences, University of Antwerp, Antwerp 2610, Belgium
| | - Erik Fransen
- StatUa Center of Statistics, University of Antwerp 2650, Antwerp, Belgium
| | - Ewa Sieliwończyk
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp & Antwerp University Hospital, Antwerp 2650, Belgium
| | - Ilse Van Gucht
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp & Antwerp University Hospital, Antwerp 2650, Belgium
| | - Emeline Van Craenenbroeck
- Department of Cardiology, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp 2650, Belgium
| | - Johan Saenen
- Department of Cardiology, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp 2650, Belgium
| | - Hein Heidbuchel
- Department of Cardiology, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp 2650, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Department of Biomedical Sciences, University of Antwerp, Antwerp 2610, Belgium
| | - Alain J Labro
- Laboratory of Molecular Biophysics, Cellular and Network Excitability, Department of Biomedical Sciences, University of Antwerp, Antwerp 2610, Belgium.,Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent 9000, Belgium
| | - Dirk Snyders
- Laboratory of Molecular Biophysics, Cellular and Network Excitability, Department of Biomedical Sciences, University of Antwerp, Antwerp 2610, Belgium
| | - Winnok De Vos
- Laboratory of Cell Biology and Histology, Faculty of Veterinary Sciences, University of Antwerp, Antwerp 2610, Belgium
| | - Dorien Schepers
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp & Antwerp University Hospital, Antwerp 2650, Belgium.,Laboratory of Molecular Biophysics, Cellular and Network Excitability, Department of Biomedical Sciences, University of Antwerp, Antwerp 2610, Belgium
| | - Maaike Alaerts
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp & Antwerp University Hospital, Antwerp 2650, Belgium
| | - Bart L Loeys
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp & Antwerp University Hospital, Antwerp 2650, Belgium.,Department of Human Genetics, Radboud University Medical Centre, Nijmegen 6525, The Netherlands
| |
Collapse
|
15
|
Du C, Rasmusson RL, Bett GC, Franks B, Zhang H, Hancox JC. Investigation of the Effects of the Short QT Syndrome D172N Kir2.1 Mutation on Ventricular Action Potential Profile Using Dynamic Clamp. Front Pharmacol 2022; 12:794620. [PMID: 35115940 PMCID: PMC8806151 DOI: 10.3389/fphar.2021.794620] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/14/2021] [Indexed: 12/22/2022] Open
Abstract
The congenital short QT syndrome (SQTS) is a cardiac condition that leads to abbreviated ventricular repolarization and an increased susceptibility to arrhythmia and sudden death. The SQT3 form of the syndrome is due to mutations to the KCNJ2 gene that encodes Kir2.1, a critical component of channels underlying cardiac inwardly rectifying K+ current, IK1. The first reported SQT3 KCNJ2 mutation gives rise to the D172N Kir2.1 mutation, the consequences of which have been studied on recombinant channels in vitro and in ventricular cell and tissue simulations. The aim of this study was to establish the effects of the D172N mutation on ventricular repolarization through real-time replacement of IK1 using the dynamic clamp technique. Whole-cell patch-clamp recordings were made from adult guinea-pig left ventricular myocytes at physiological temperature. Action potentials (APs) were elicited at 1 Hz. Intrinsic IK1 was inhibited with a low concentration (50 µM) of Ba2+ ions, which led to AP prolongation and triangulation, accompanied by a ∼6 mV depolarization of resting membrane potential. Application of synthetic IK1 through dynamic clamp restored AP duration, shape and resting potential. Replacement of wild-type (WT) IK1 with heterozygotic (WT-D172N) or homozygotic (D172N) mutant formulations under dynamic clamp significantly abbreviated AP duration (APD90) and accelerated maximal AP repolarization velocity, with no significant hyperpolarization of resting potential. Across stimulation frequencies from 0.5 to 3 Hz, the relationship between APD90 and cycle length was downward shifted, reflecting AP abbreviation at all stimulation frequencies tested. In further AP measurements at 1 Hz from hiPSC cardiomyocytes, the D172N mutation produced similar effects on APD and repolarization velocity; however, resting potential was moderately hyperpolarized by application of mutant IK1 to these cells. Overall, the results of this study support the major changes in ventricular cell AP repolarization with the D172N predicted from prior AP modelling and highlight the potential utility of using adult ventricular cardiomyocytes for dynamic clamp exploration of functional consequences of Kir2.1 mutations.
Collapse
Affiliation(s)
- Chunyun Du
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Randall L. Rasmusson
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University of New York, University at Buffalo, Buffalo, NY, United States
- Cytocybernetics Inc, North Tonawanda, NY, United States
| | - Glenna C. Bett
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University of New York, University at Buffalo, Buffalo, NY, United States
- Cytocybernetics Inc, North Tonawanda, NY, United States
- Department of Obstetrics and Gynecology, Center for Cellular and Systems Electrophysiology, State University of New York, University at Buffalo, Buffalo, NY, United States
| | | | - Henggui Zhang
- Biological Physics Group, Department of Physics and Astronomy, The University of Manchester, Manchester, United Kingdom
| | - Jules C. Hancox
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
- Biological Physics Group, Department of Physics and Astronomy, The University of Manchester, Manchester, United Kingdom
- *Correspondence: Jules C. Hancox,
| |
Collapse
|
16
|
Contributions of Na V1.8 and Na V1.9 to excitability in human induced pluripotent stem-cell derived somatosensory neurons. Sci Rep 2021; 11:24283. [PMID: 34930944 PMCID: PMC8688473 DOI: 10.1038/s41598-021-03608-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 12/02/2021] [Indexed: 12/13/2022] Open
Abstract
The inhibition of voltage-gated sodium (NaV) channels in somatosensory neurons presents a promising novel modality for the treatment of pain. However, the precise contribution of these channels to neuronal excitability, the cellular correlate of pain, is unknown; previous studies using genetic knockout models or pharmacologic block of NaV channels have identified general roles for distinct sodium channel isoforms, but have never quantified their exact contributions to these processes. To address this deficit, we have utilized dynamic clamp electrophysiology to precisely tune in varying levels of NaV1.8 and NaV1.9 currents into induced pluripotent stem cell-derived sensory neurons (iPSC-SNs), allowing us to quantify how graded changes in these currents affect different parameters of neuronal excitability and electrogenesis. We quantify and report direct relationships between NaV1.8 current density and action potential half-width, overshoot, and repetitive firing. We additionally quantify the effect varying NaV1.9 current densities have on neuronal membrane potential and rheobase. Furthermore, we examined the simultaneous interplay between NaV1.8 and NaV1.9 on neuronal excitability. Finally, we show that minor biophysical changes in the gating of NaV1.8 can render human iPSC-SNs hyperexcitable, in a first-of-its-kind investigation of a gain-of-function NaV1.8 mutation in a human neuronal background.
Collapse
|
17
|
Van de Sande DV, Kopljar I, Maaike A, Teisman A, Gallacher DJ, Bart L, Snyders DJ, Leybaert L, Lu HR, Labro AJ. The resting membrane potential of hSC-CM in a syncytium is more hyperpolarised than that of isolated cells. Channels (Austin) 2021; 15:239-252. [PMID: 33465001 PMCID: PMC7817136 DOI: 10.1080/19336950.2021.1871815] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/20/2020] [Accepted: 12/31/2020] [Indexed: 01/11/2023] Open
Abstract
Human-induced pluripotent stem cell (hiPSC) and stem cell (hSC) derived cardiomyocytes (CM) are gaining popularity as in vitro model for cardiology and pharmacology studies. A remaining flaw of these cells, as shown by single-cell electrophysiological characterization, is a more depolarized resting membrane potential (RMP) compared to native CM. Most reports attribute this to a lower expression of the Kir2.1 potassium channel that generates the IK1 current. However, most RMP recordings are obtained from isolated hSC/hiPSC-CMs whereas in a more native setting these cells are interconnected with neighboring cells by connexin-based gap junctions, forming a syncytium. Hereby, these cells are electrically connected and the total pool of IK1 increases. Therefore, the input resistance (Ri) of interconnected cells is lower than that of isolated cells. During patch clamp experiments pipettes need to be well attached or sealed to the cell, which is reflected in the seal resistance (Rs), because a nonspecific ionic current can leak through this pipette-cell contact or seal and balance out small currents within the cell such as IK1. By recording the action potential of isolated hSC-CMs and that of hSC-CMs cultured in small monolayers, we show that the RMP of hSC-CMs in monolayer is approximately -20 mV more hyperpolarized compared to isolated cells. Accordingly, adding carbenoxolone, a connexin channel blocker, isolates the cell that is patch clamped from its neighboring cells of the monolayer and depolarizes the RMP. The presented data show that the recorded RMP of hSC-CMs in a syncytium is more negative than that determined from isolated hSC/hiPSC-CMs, most likely because the active pool of Kir2.1 channels increased.
Collapse
Affiliation(s)
| | - Ivan Kopljar
- Global Safety Pharmacology, Non-Clinical Safety, Janssen R&D, Beerse, Belgium
| | - Alaerts Maaike
- Centre of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Ard Teisman
- Global Safety Pharmacology, Non-Clinical Safety, Janssen R&D, Beerse, Belgium
| | - David J. Gallacher
- Global Safety Pharmacology, Non-Clinical Safety, Janssen R&D, Beerse, Belgium
| | - Loeys Bart
- Centre of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Dirk J. Snyders
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Luc Leybaert
- Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Hua Rong Lu
- Global Safety Pharmacology, Non-Clinical Safety, Janssen R&D, Beerse, Belgium
| | - Alain J. Labro
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
18
|
Yamamoto Y, Hirose S, Wuriyanghai Y, Yoshinaga D, Makiyama T. Electrophysiological Analysis of hiPSC-Derived Cardiomyocytes Using a Patch-Clamp Technique. Methods Mol Biol 2021; 2320:121-133. [PMID: 34302654 DOI: 10.1007/978-1-0716-1484-6_13] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
Abstract
Electrophysiological analysis of human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) using a patch-clamp technique enables the most precise evaluation of electrophysiological properties in single cells. Compared to multielectrode array (MEA) and membrane voltage imaging, patch-clamp recordings offer quantitative measurements of action potentials, and the relevant ionic currents which are essential for the research of disease modeling of inherited arrhythmias, safety pharmacology, and drug discovery using hiPSC-CMs. In this chapter, we describe the detail flow of patch-clamp recordings in hiPSC-CMs.
Collapse
Affiliation(s)
- Yuta Yamamoto
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center, Suita, Japan.,Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Sayako Hirose
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yimin Wuriyanghai
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Daisuke Yoshinaga
- Department of Pediatrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takeru Makiyama
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| |
Collapse
|
19
|
Modeling Cardiomyopathies in a Dish: State-of-the-Art and Novel Perspectives on hiPSC-Derived Cardiomyocytes Maturation. BIOLOGY 2021; 10:biology10080730. [PMID: 34439963 PMCID: PMC8389603 DOI: 10.3390/biology10080730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 12/23/2022]
Abstract
The stem cell technology and the induced pluripotent stem cells (iPSCs) production represent an excellent alternative tool to study cardiomyopathies, which overcome the limitations associated with primary cardiomyocytes (CMs) access and manipulation. CMs from human iPSCs (hiPSC-CMs) are genetically identical to patient primary cells of origin, with the main electrophysiological and mechanical features of CMs. The key issue to be solved is to achieve a degree of structural and functional maturity typical of adult CMs. In this perspective, we will focus on the main differences between fetal-like hiPSC-CMs and adult CMs. A viewpoint is given on the different approaches used to improve hiPSC-CMs maturity, spanning from long-term culture to complex engineered heart tissue. Further, we outline limitations and future developments needed in cardiomyopathy disease modeling.
Collapse
|
20
|
Al Sayed ZR, Canac R, Cimarosti B, Bonnard C, Gourraud JB, Hamamy H, Kayserili H, Girardeau A, Jouni M, Jacob N, Gaignerie A, Chariau C, David L, Forest V, Marionneau C, Charpentier F, Loussouarn G, Lamirault G, Reversade B, Zibara K, Lemarchand P, Gaborit N. Human model of IRX5 mutations reveals key role for this transcription factor in ventricular conduction. Cardiovasc Res 2021; 117:2092-2107. [PMID: 32898233 DOI: 10.1093/cvr/cvaa259] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/15/2020] [Accepted: 08/28/2020] [Indexed: 01/02/2023] Open
Abstract
AIMS Several inherited arrhythmic diseases have been linked to single gene mutations in cardiac ion channels and interacting proteins. However, the mechanisms underlying most arrhythmias, are thought to involve altered regulation of the expression of multiple effectors. In this study, we aimed to examine the role of a transcription factor (TF) belonging to the Iroquois homeobox family, IRX5, in cardiac electrical function. METHODS AND RESULTS Using human cardiac tissues, transcriptomic correlative analyses between IRX5 and genes involved in cardiac electrical activity showed that in human ventricular compartment, IRX5 expression strongly correlated to the expression of major actors of cardiac conduction, including the sodium channel, Nav1.5, and Connexin 40 (Cx40). We then generated human-induced pluripotent stem cells (hiPSCs) derived from two Hamamy syndrome-affected patients carrying distinct homozygous loss-of-function mutations in IRX5 gene. Cardiomyocytes derived from these hiPSCs showed impaired cardiac gene expression programme, including misregulation in the control of Nav1.5 and Cx40 expression. In accordance with the prolonged QRS interval observed in Hamamy syndrome patients, a slower ventricular action potential depolarization due to sodium current reduction was observed on electrophysiological analyses performed on patient-derived cardiomyocytes, confirming the functional role of IRX5 in electrical conduction. Finally, a cardiac TF complex was newly identified, composed by IRX5 and GATA4, in which IRX5 potentiated GATA4-induction of SCN5A expression. CONCLUSION Altogether, this work unveils a key role for IRX5 in the regulation of human ventricular depolarization and cardiac electrical conduction, providing therefore new insights into our understanding of cardiac diseases.
Collapse
Affiliation(s)
- Zeina R Al Sayed
- Université de Nantes, CNRS, INSERM, l'institut du thorax, 8 quai Moncousu, F-44000 Nantes, France
| | - Robin Canac
- Université de Nantes, CNRS, INSERM, l'institut du thorax, 8 quai Moncousu, F-44000 Nantes, France
| | - Bastien Cimarosti
- Université de Nantes, CNRS, INSERM, l'institut du thorax, 8 quai Moncousu, F-44000 Nantes, France
| | - Carine Bonnard
- Institute of Medical Biology, A*STAR, 8A Biomedical Grove, Singapore 138648, Singapore
| | - Jean-Baptiste Gourraud
- Université de Nantes, CNRS, INSERM, l'institut du thorax, 8 quai Moncousu, F-44000 Nantes, France
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, 8 quai Moncousu, F-44000 Nantes, France
| | - Hanan Hamamy
- Department of Genetic Medicine and Development, Geneva University, 1 rue Michel-Servet, Geneva 1211, Switzerland
| | - Hulya Kayserili
- Medical Genetics Department, Koç University School of Medicine(KUSOM), Rumelifeneri Yolu 34450, Istanbul, Turkey
| | - Aurore Girardeau
- Université de Nantes, CNRS, INSERM, l'institut du thorax, 8 quai Moncousu, F-44000 Nantes, France
| | - Mariam Jouni
- Université de Nantes, CNRS, INSERM, l'institut du thorax, 8 quai Moncousu, F-44000 Nantes, France
| | - Nicolas Jacob
- Université de Nantes, CNRS, INSERM, l'institut du thorax, 8 quai Moncousu, F-44000 Nantes, France
| | - Anne Gaignerie
- Université de Nantes, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, 8 Quai Moncousu, F-44000 Nantes, France
| | - Caroline Chariau
- Université de Nantes, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, 8 Quai Moncousu, F-44000 Nantes, France
| | - Laurent David
- Université de Nantes, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, 8 Quai Moncousu, F-44000 Nantes, France
- Université de Nantes, INSERM, CRTI, 30 Bd Jean Monnet, F-44093 Nantes, France
- ITUN, CHU Nantes, 30 Bd Jean Monnet, F-44093 Nantes, France
| | - Virginie Forest
- Université de Nantes, CNRS, INSERM, l'institut du thorax, 8 quai Moncousu, F-44000 Nantes, France
| | - Céline Marionneau
- Université de Nantes, CNRS, INSERM, l'institut du thorax, 8 quai Moncousu, F-44000 Nantes, France
| | - Flavien Charpentier
- Université de Nantes, CNRS, INSERM, l'institut du thorax, 8 quai Moncousu, F-44000 Nantes, France
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, 8 quai Moncousu, F-44000 Nantes, France
| | - Gildas Loussouarn
- Université de Nantes, CNRS, INSERM, l'institut du thorax, 8 quai Moncousu, F-44000 Nantes, France
| | - Guillaume Lamirault
- Université de Nantes, CNRS, INSERM, l'institut du thorax, 8 quai Moncousu, F-44000 Nantes, France
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, 8 quai Moncousu, F-44000 Nantes, France
| | - Bruno Reversade
- Institute of Medical Biology, A*STAR, 8A Biomedical Grove, Singapore 138648, Singapore
- Medical Genetics Department, Koç University School of Medicine(KUSOM), Rumelifeneri Yolu 34450, Istanbul, Turkey
- Department of Paediatrics, National University of Singapore, 1E Kent Ridge Road, Singapore 119228, Singapore
- Institute of Molecular and Cellular Biology, A*STAR, 61 Biopolis Drive, Singapore 138673, Singapore
- Reproductive Biology Laboratory, Amsterdam UMC, Meibergdreef 9 1105, Amsterdam-Zuidoost, Netherlands
| | - Kazem Zibara
- ER045, Laboratory of stem cells, DSST, Biology department, Faculty of Sciences, Lebanese University, Rafic Hariri Campus - Hadath, Beirut 1700, Lebanon
| | - Patricia Lemarchand
- Université de Nantes, CNRS, INSERM, l'institut du thorax, 8 quai Moncousu, F-44000 Nantes, France
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, 8 quai Moncousu, F-44000 Nantes, France
| | - Nathalie Gaborit
- Université de Nantes, CNRS, INSERM, l'institut du thorax, 8 quai Moncousu, F-44000 Nantes, France
| |
Collapse
|
21
|
Electrophysiology of hiPSC-Cardiomyocytes Co-Cultured with HEK Cells Expressing the Inward Rectifier Channel. Int J Mol Sci 2021; 22:ijms22126621. [PMID: 34205607 PMCID: PMC8235371 DOI: 10.3390/ijms22126621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/07/2021] [Accepted: 06/11/2021] [Indexed: 11/23/2022] Open
Abstract
The immature electrophysiology of human-induced pluripotent stem cell-derived cardiomyocytes (hiCMs) complicates their use for therapeutic and pharmacological purposes. An insufficient inward rectifying current (IK1) and the presence of a funny current (if) cause spontaneous electrical activity. This study tests the hypothesis that the co-culturing of hiCMs with a human embryonic kidney (HEK) cell-line expressing the Kir2.1 channel (HEK-IK1) can generate an electrical syncytium with an adult-like cardiac electrophysiology. The mechanical activity of co-cultures using different HEK-IK1:hiCM ratios was compared with co-cultures using wildtype (HEK–WT:hiCM) or hiCM alone on days 3–8 after plating. Only ratios of 1:3 and 1:1 showed a significant reduction in spontaneous rate at days 4 and 6, suggesting that IK1 was influencing the electrophysiology. Detailed analysis at day 4 revealed an increased incidence of quiescent wells or sub-areas. Electrical activity showed a decreased action potential duration (APD) at 20% and 50%, but not at 90%, alongside a reduced amplitude of the aggregate AP signal. A computational model of the 1:1 co-culture replicates the electrophysiological effects of HEK–WT. The addition of the IK1 conductance reduced the spontaneous rate and APD20, 50 and 90, and minor variation in the intercellular conductance caused quiescence. In conclusion, a 1:1 co-culture HEK-IK1:hiCM caused changes in electrophysiology and spontaneous activity consistent with the integration of IK1 into the electrical syncytium. However, the additional electrical effects of the HEK cell at 1:1 increased the possibility of electrical quiescence before sufficient IK1 was integrated into the syncytium.
Collapse
|
22
|
Maturation strategies and limitations of induced pluripotent stem cell-derived cardiomyocytes. Biosci Rep 2021; 41:226678. [PMID: 33057659 PMCID: PMC8209171 DOI: 10.1042/bsr20200833] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 10/06/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) have the ability to differentiate into cardiomyocytes (CMs). They are not only widely used in cardiac pharmacology screening, human heart disease modeling, and cell transplantation-based treatments, but also the most promising source of CMs for experimental and clinical applications. However, their use is largely restricted by the immature phenotype of structure and function, which is similar to embryonic or fetal CMs and has certain differences from adult CMs. In order to overcome this critical issue, many studies have explored and revealed new strategies to induce the maturity of iPSC-CMs. Therefore, this article aims to review recent induction methods of mature iPSC-CMs, related mechanisms, and limitations.
Collapse
|
23
|
Akwaboah AD, Tsevi B, Yamlome P, Treat JA, Brucal-Hallare M, Cordeiro JM, Deo M. An in silico hiPSC-Derived Cardiomyocyte Model Built With Genetic Algorithm. Front Physiol 2021; 12:675867. [PMID: 34220540 PMCID: PMC8242263 DOI: 10.3389/fphys.2021.675867] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/05/2021] [Indexed: 12/25/2022] Open
Abstract
The formulation of in silico biophysical models generally requires optimization strategies for reproducing experimentally observed phenomena. In electrophysiological modeling, robust nonlinear regressive methods are often crucial for guaranteeing high fidelity models. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), though nascent, have proven to be useful in cardiac safety pharmacology, regenerative medicine, and in the implementation of patient-specific test benches for investigating inherited cardiac disorders. This study demonstrates the potency of heuristic techniques at formulating biophysical models, with emphasis on a hiPSC-CM model using a novel genetic algorithm (GA) recipe we proposed. The proposed GA protocol was used to develop a hiPSC-CM biophysical computer model by fitting mathematical formulations to experimental data for five ionic currents recorded in hiPSC-CMs. The maximum conductances of the remaining ionic channels were scaled based on recommendations from literature to accurately reproduce the experimentally observed hiPSC-CM action potential (AP) metrics. Near-optimal parameter fitting was achieved for the GA-fitted ionic currents. The resulting model recapitulated experimental AP parameters such as AP durations (APD50, APD75, and APD90), maximum diastolic potential, and frequency of automaticity. The outcome of this work has implications for validating the biophysics of hiPSC-CMs in their use as viable substitutes for human cardiomyocytes, particularly in cardiac safety pharmacology and in the study of inherited cardiac disorders. This study presents a novel GA protocol useful for formulating robust numerical biophysical models. The proposed protocol is used to develop a hiPSC-CM model with implications for cardiac safety pharmacology.
Collapse
Affiliation(s)
- Akwasi D Akwaboah
- Department of Engineering, Norfolk State University, Norfolk, VA, United States
| | - Bright Tsevi
- Department of Engineering, Norfolk State University, Norfolk, VA, United States
| | - Pascal Yamlome
- Department of Engineering, Norfolk State University, Norfolk, VA, United States
| | | | | | | | - Makarand Deo
- Department of Engineering, Norfolk State University, Norfolk, VA, United States
| |
Collapse
|
24
|
Homan T, Delanoë-Ayari H, Meli AC, Cazorla O, Gergely C, Mejat A, Chevalier P, Moreau A. MorphoScript: a dedicated analysis to assess the morphology and contractile structures of cardiomyocytes derived from stem cells. Bioinformatics 2021; 37:4209-4215. [PMID: 34048539 DOI: 10.1093/bioinformatics/btab400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/19/2021] [Accepted: 05/27/2021] [Indexed: 01/07/2023] Open
Abstract
MOTIVATION Cardiomyocytes derived from stem cells are closely followed, notably since the discovery in 2007 of human induced pluripotent stem cells (hiPSC). Cardiomyocytes (hiPSC-CM) derived from hiPSC are indeed more and more used to study specific cardiac diseases as well as for developing novel applications such as drug safety experiments. Robust dedicated tools to characterize hiPSC-CM are now required. The hiPSC-CM morphology constitutes an important parameter since these cells do not demonstrate the expected rod shape, characteristic of native human cardiomyocytes. Similarly, the presence, the density and the organization of contractile structures would be a valuable parameter to study. Precise measurements of such characteristics would be useful in many situations: for describing pathological conditions, for pharmacological screens or even for studies focused on the hiPSC-CM maturation process. RESULTS For this purpose, we developed a MATLAB based image analysis toolbox, which gives accurate values for cellular morphology parameters as well as for the contractile cell organization. IMPLEMENTATION To demonstrate the power of this automated image analysis, we used a commercial maturation medium intended to promote the maturation status of hiPSC-CM, and compare the parameters with the ones obtained with standard culture medium, and with freshly dissociated mouse cardiomyocytes. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Tess Homan
- Claude Bernard University, Lyon 1, Université de Lyon, Lyon; Institut lumière matière, (Lyon, France)
| | - Hélène Delanoë-Ayari
- Claude Bernard University, Lyon 1, Université de Lyon, Lyon; Institut lumière matière, (Lyon, France)
| | - Albano C Meli
- PhyMedExp, INSERM, CNRS, University of Montpellier, Montpellier
| | - Olivier Cazorla
- PhyMedExp, INSERM, CNRS, University of Montpellier, Montpellier
| | - Csilla Gergely
- L2C, University of Montpellier, CNRS, Montpellier, France
| | - Alexandre Mejat
- Claude Bernard University, Lyon 1, Université de Lyon, Lyon; Neuromyogene Institut, (Lyon, France)
| | - Philippe Chevalier
- Claude Bernard University, Lyon 1, Université de Lyon, Lyon; Neuromyogene Institut, (Lyon, France).,Hospices civils de Lyon, Service de Rythmologie, (Bron, France)
| | - Adrien Moreau
- PhyMedExp, INSERM, CNRS, University of Montpellier, Montpellier
| |
Collapse
|
25
|
Verkerk AO, Marchal GA, Zegers JG, Kawasaki M, Driessen AHG, Remme CA, de Groot JR, Wilders R. Patch-Clamp Recordings of Action Potentials From Human Atrial Myocytes: Optimization Through Dynamic Clamp. Front Pharmacol 2021; 12:649414. [PMID: 33912059 PMCID: PMC8072333 DOI: 10.3389/fphar.2021.649414] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/18/2021] [Indexed: 11/29/2022] Open
Abstract
Introduction: Atrial fibrillation (AF) is the most common cardiac arrhythmia. Consequently, novel therapies are being developed. Ultimately, the impact of compounds on the action potential (AP) needs to be tested in freshly isolated human atrial myocytes. However, the frequent depolarized state of these cells upon isolation seriously hampers reliable AP recordings. Purpose: We assessed whether AP recordings from single human atrial myocytes could be improved by providing these cells with a proper inward rectifier K+ current (IK1), and consequently with a regular, non-depolarized resting membrane potential (RMP), through “dynamic clamp”. Methods: Single myocytes were enzymatically isolated from left atrial appendage tissue obtained from patients with paroxysmal AF undergoing minimally invasive surgical ablation. APs were elicited at 1 Hz and measured using perforated patch-clamp methodology, injecting a synthetic IK1 to generate a regular RMP. The injected IK1 had strong or moderate rectification. For comparison, a regular RMP was forced through injection of a constant outward current. A wide variety of ion channel blockers was tested to assess their modulatory effects on AP characteristics. Results: Without any current injection, RMPs ranged from −9.6 to −86.2 mV in 58 cells. In depolarized cells (RMP positive to −60 mV), RMP could be set at −80 mV using IK1 or constant current injection and APs could be evoked upon stimulation. AP duration differed significantly between current injection methods (p < 0.05) and was shortest with constant current injection and longest with injection of IK1 with strong rectification. With moderate rectification, AP duration at 90% repolarization (APD90) was similar to myocytes with regular non-depolarized RMP, suggesting that a synthetic IK1 with moderate rectification is the most appropriate for human atrial myocytes. Importantly, APs evoked using each injection method were still sensitive to all drugs tested (lidocaine, nifedipine, E-4031, low dose 4-aminopyridine, barium, and apamin), suggesting that the major ionic currents of the atrial cells remained functional. However, certain drug effects were quantitatively dependent on the current injection approach used. Conclusion: Injection of a synthetic IK1 with moderate rectification facilitates detailed AP measurements in human atrial myocytes. Therefore, dynamic clamp represents a promising tool for testing novel antiarrhythmic drugs.
Collapse
Affiliation(s)
- Arie O Verkerk
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Gerard A Marchal
- Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Jan G Zegers
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Makiri Kawasaki
- Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Antoine H G Driessen
- Department of Cardiothoracic Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Carol Ann Remme
- Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Joris R de Groot
- Department of Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
26
|
Verkerk AO, Wilders R. Dynamic Clamp in Electrophysiological Studies on Stem Cell-Derived Cardiomyocytes-Why and How? J Cardiovasc Pharmacol 2021; 77:267-279. [PMID: 33229908 DOI: 10.1097/fjc.0000000000000955] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/31/2020] [Indexed: 12/15/2022]
Abstract
ABSTRACT Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) are supposed to be a good human-based model, with virtually unlimited cell source, for studies on mechanisms underlying cardiac development and cardiac diseases, and for identification of drug targets. However, a major drawback of hPSC-CMs as a model system, especially for electrophysiological studies, is their depolarized state and associated spontaneous electrical activity. Various approaches are used to overcome this drawback, including the injection of "synthetic" inward rectifier potassium current (IK1), which is computed in real time, based on the recorded membrane potential ("dynamic clamp"). Such injection of an IK1-like current results in quiescent hPSC-CMs with a nondepolarized resting potential that show "adult-like" action potentials on stimulation, with functional availability of the most important ion channels involved in cardiac electrophysiology. These days, dynamic clamp has become a widely appreciated electrophysiological tool. However, setting up a dynamic clamp system can still be laborious and difficult, both because of the required hardware and the implementation of the dedicated software. In the present review, we first summarize the potential mechanisms underlying the depolarized state of hPSC-CMs and the functional consequences of this depolarized state. Next, we explain how an existing manual patch clamp setup can be extended with dynamic clamp. Finally, we shortly validate the extended setup with atrial-like and ventricular-like hPSC-CMs. We feel that dynamic clamp is a highly valuable tool in the field of cellular electrophysiological studies on hPSC-CMs and hope that our directions for setting up such dynamic clamp system may prove helpful.
Collapse
Affiliation(s)
- Arie O Verkerk
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands ; and
- Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands ; and
| |
Collapse
|
27
|
Deo M, Akwaboah A, Tsevi B, Treat JA, Cordeiro JM. Role of the rapid delayed rectifier K + current in human induced pluripotent stem cells derived cardiomyocytes. ARCHIVES OF STEM CELL AND THERAPY 2021; 1:14-18. [PMID: 33604593 PMCID: PMC7889062 DOI: 10.46439/stemcell.1.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Makarand Deo
- Department of Engineering, Norfolk State University, Norfolk, Virginia, USA
| | - Akwasi Akwaboah
- Department of Engineering, Norfolk State University, Norfolk, Virginia, USA
| | - Bright Tsevi
- Department of Engineering, Norfolk State University, Norfolk, Virginia, USA
| | - Jacqueline A Treat
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, New York, USA
| | - Jonathan M Cordeiro
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, New York, USA
| |
Collapse
|
28
|
A Scalable Approach Reveals Functional Responses of iPSC Cardiomyocyte 3D Spheroids. SLAS DISCOVERY 2020; 26:352-363. [DOI: 10.1177/2472555220975332] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cardiomyocytes (CMs) derived from induced pluripotent stem cells (iPSCs) provide an in vitro model of the human myocardium. Complex 3D scaffolded culture methods improve the phenotypical maturity of iPSC-CMs, although typically at the expense of throughput. We have developed a novel, scalable approach that enables the use of iPSC-CM 3D spheroid models in a label-free readout system in a standard 96-well plate-based format. Spheroids were accurately positioned onto recording electrodes using a magnetic gold–iron oxide nanoparticle approach. Remarkably, both contractility (impedance) and extracellular field potentials (EFPs) could be detected from the actively beating spheroids over long durations and after automated dosing with pharmacological agents. The effects on these parameters of factors, such as co-culture (including human primary cardiac fibroblasts), extracellular buffer composition, and electrical pacing, were investigated. Beat amplitudes were increased greater than 15-fold by co-culture with fibroblasts. Optimization of extracellular Ca2+ fluxes and electrical pacing promoted the proper physiological response to positive inotropic agonists of increased beat amplitude (force) rather than the increased beat rate often observed in iPSC-CM studies. Mechanistically divergent repolarizations in different spheroid models were indicated by their responses to BaCl2 compared with E-4031. These studies demonstrate a new method that enables the pharmacological responses of 3D iPSC-CM spheroids to be determined in a label-free, standardized, 96-well plate-based system. This approach could have discovery applications across cardiovascular efficacy and safety, where parameters typically sought as readouts of iPSC-CM maturity or physiological relevance have the potential to improve assay predictivity.
Collapse
|
29
|
Functional evaluation of gene mutations in Long QT Syndrome: strength of evidence from in vitro assays for deciphering variants of uncertain significance. JOURNAL OF CONGENITAL CARDIOLOGY 2020. [DOI: 10.1186/s40949-020-00037-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Genetic screening is now commonplace for patients suspected of having inherited cardiac conditions. Variants of uncertain significance (VUS) in disease-associated genes pose problems for the diagnostician and reliable methods for evaluating VUS function are required. Although function is difficult to interrogate for some genes, heritable channelopathies have established mechanisms that should be amenable to well-validated evaluation techniques.
The cellular electrophysiology techniques of ‘voltage-’ and ‘patch-’ clamp have a long history of successful use and have been central to identifying both the roles of genes involved in different forms of congenital Long QT Syndrome (LQTS) and the mechanisms by which mutations lead to aberrant ion channel function underlying clinical phenotypes. This is particularly evident for KCNQ1, KCNH2 and SCN5A, mutations in which underlie > 90% of genotyped LQTS cases (the LQT1-LQT3 subtypes). Recent studies utilizing high throughput (HT) planar patch-clamp recording have shown it to discriminate effectively between rare benign and pathological variants, studied through heterologous expression of recombinant channels. In combination with biochemical methods for evaluating channel trafficking and supported by biophysical modelling, patch clamp also provides detailed mechanistic insight into the functional consequences of identified mutations. Whilst potentially powerful, patient-specific stem-cell derived cardiomyocytes and genetically modified animal models are currently not well-suited to high throughput VUS study.
Conclusion
The widely adopted 2015 American College of Medical Genetics (ACMG) and Association for Molecular Pathology (AMP) guidelines for the interpretation of sequence variants include the PS3 criterion for consideration of evidence from well-established in vitro or in vivo assays. The wealth of information on underlying mechanisms of LQT1-LQT3 and recent HT patch clamp data support consideration of patch clamp data together (for LQT1 and LQT2) with information from biochemical trafficking assays as meeting the PS3 criterion of well established assays, able to provide ‘strong’ evidence for functional pathogenicity of identified VUS.
Collapse
|
30
|
Ashok P, Parikh A, Du C, Tzanakakis ES. Xenogeneic-Free System for Biomanufacturing of Cardiomyocyte Progeny From Human Pluripotent Stem Cells. Front Bioeng Biotechnol 2020; 8:571425. [PMID: 33195131 PMCID: PMC7644809 DOI: 10.3389/fbioe.2020.571425] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/28/2020] [Indexed: 01/14/2023] Open
Abstract
Functional heart cells and tissues sourced from human pluripotent stem cells (hPSCs) have great potential for substantially advancing treatments of cardiovascular maladies. Realization of this potential will require the development of cost-effective and tunable bioprocesses for manufacturing hPSC-based cell therapeutics. Here, we report the development of a xeno-free platform for guiding the cardiogenic commitment of hPSCs. The system is based on a fully defined, open-source formulation without complex supplements, which have varied and often undetermined effects on stem cell physiology. The formulation was used to systematically investigate factors inducing the efficient commitment to cardiac mesoderm of three hPSC lines. Contractile clusters of cells appeared within a week of differentiation in planar cultures and by day 13 over 80% of the cells expressed cardiac progeny markers such as TNNT2. In conjunction with expansion, this differentiation strategy was employed in stirred-suspension cultures of hPSCs. Scalable differentiation resulted in 0.4-2 million CMs/ml or ∼5-20 TNNT2-positive cells per seeded hPSC without further enrichment. Our findings will contribute to the engineering of bioprocesses advancing the manufacturing of stem cell-based therapeutics for heart diseases.
Collapse
Affiliation(s)
- Preeti Ashok
- Chemical and Biological Engineering, Tufts University, Medford, MA, United States
| | | | - Chuang Du
- Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Emmanuel S. Tzanakakis
- Chemical and Biological Engineering, Tufts University, Medford, MA, United States
- Clinical and Translational Science Institute, Tufts Medical Center, Boston, MA, United States
- Developmental Molecular and Chemical Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
31
|
Li W, Han JL, Entcheva E. Syncytium cell growth increases Kir2.1 contribution in human iPSC-cardiomyocytes. Am J Physiol Heart Circ Physiol 2020; 319:H1112-H1122. [PMID: 32986966 PMCID: PMC7789971 DOI: 10.1152/ajpheart.00148.2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) enable cardiotoxicity testing and personalized medicine. However, their maturity is of concern, including relatively depolarized resting membrane potential and more spontaneous activity compared with adult cardiomyocytes, implicating low or lacking inward rectifier potassium current (Ik1). Here, protein quantification confirms Kir2.1 expression in hiPSC-CM syncytia, albeit several times lower than in adult heart tissue. We find that hiPSC-CM culture density influences Kir2.1 expression at the mRNA level (potassium inwardly rectifying channel subfamily J member 2) and at the protein level and its associated electrophysiology phenotype. Namely, all-optical cardiac electrophysiology and pharmacological treatments reveal reduction of spontaneous and irregular activity and increase in action potential upstroke in denser cultures. Blocking Ik1-like currents with BaCl2 increased spontaneous frequency and blunted action potential upstrokes during pacing in a dose-dependent manner only in the highest-density cultures, in line with Ik1’s role in regulating the resting membrane potential. Our results emphasize the importance of syncytial growth of hiPSC-CMs for more physiologically relevant phenotype and the power of all-optical electrophysiology to study cardiomyocytes in their multicellular setting. NEW & NOTEWORTHY We identify cell culture density and cell-cell contact as an important factor in determining the expression of a key ion channel at the transcriptional and the protein levels, KCNJ2/Kir2.1, and its contribution to the electrophysiology of human induced pluripotent stem cell-derived cardiomyocytes. Our results indicate that studies on isolated cells, out of tissue context, may underestimate the cellular ion channel properties being characterized.
Collapse
Affiliation(s)
- Weizhen Li
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia
| | - Julie L Han
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia
| | - Emilia Entcheva
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia
| |
Collapse
|
32
|
Barajas-Martinez H, Smith M, Hu D, Goodrow RJ, Puleo C, Hasdemir C, Antzelevitch C, Pfeiffer R, Treat JA, Cordeiro JM. Susceptibility to Ventricular Arrhythmias Resulting from Mutations in FKBP1B, PXDNL, and SCN9A Evaluated in hiPSC Cardiomyocytes. Stem Cells Int 2020; 2020:8842398. [PMID: 32952569 PMCID: PMC7481990 DOI: 10.1155/2020/8842398] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/29/2020] [Accepted: 08/11/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND We report an inherited cardiac arrhythmia syndrome consisting of Brugada and Early Repolarization Syndrome associated with variants in SCN9A, PXDNL, and FKBP1B. The proband inherited the 3 mutations and exhibited palpitations and arrhythmia-mediated syncope, whereas the parents and sister, who carried one or two of the mutations, were asymptomatic. METHODS AND RESULTS We assessed the functional impact of these mutations in induced pluripotent stem cell cardiomyocytes (hiPSC-CMs) derived from the proband and an unaffected family member. Current and voltage clamp recordings, as well as confocal microscopy analysis of Ca2+ transients, were evaluated in hiPSC-CMs from the proband and compared these results with hiPSC-CMs from undiseased controls. Genetic analysis using next-generation DNA sequencing revealed heterozygous mutations in SCN9A, PXDNL, and FKBP1B in the proband. The proband displayed right bundle branch block and exhibited episodes of syncope. The father carried a mutation in FKBP1B, whereas the mother and sister carried the SCN9A mutation. None of the 3 family members screened developed cardiac events. Action potential recordings from control hiPSC-CM showed spontaneous activity and a low upstroke velocity. In contrast, the hiPSC-CM from the proband showed irregular spontaneous activity. Confocal microscopy of the hiPSC-CM of the proband revealed low fluorescence intensity Ca2+ transients that were episodic in nature. Patch-clamp measurements in hiPSC-CM showed no difference in I Na but reduced I Ca in the proband compared with control. Coexpression of PXDNL-R391Q with SCN5A-WT displayed lower I Na density compared to PXDNL-WT. In addition, coexpression of PXDNL-R391Q with KCND3-WT displayed significantly higher I to density compared to PXDNL-WT. CONCLUSION SCN9A, PXDNL, and FKBP1B variants appeared to alter spontaneous activity in hiPSC-CM. Only the proband carrying all 3 mutations displayed the ERS/BrS phenotype, whereas one nor two mutations alone did not produce the clinical phenotype. Our results suggest a polygenic cause of the BrS/ERS arrhythmic phenotype due to mutations in these three gene variants caused a very significant loss of function of I Na and I Ca and gain of function of I to.
Collapse
Affiliation(s)
- Hector Barajas-Martinez
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
- Department of Cardiovascular Research, Lakenau Institute for Medical Research, Wynnewood, PA, USA
| | - Maya Smith
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
| | - Dan Hu
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
- Department of Cardiology & Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, China
| | - Robert J. Goodrow
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
| | - Colleen Puleo
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
| | - Can Hasdemir
- Department of Cardiology, Ege University School of Medicine, Izmir, Turkey
| | - Charles Antzelevitch
- Department of Cardiovascular Research, Lakenau Institute for Medical Research, Wynnewood, PA, USA
- Kimmel College of Medicine of Thomas Jefferson University, Philadelphia, PA, USA
| | - Ryan Pfeiffer
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
| | - Jacqueline A. Treat
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
| | - Jonathan M. Cordeiro
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
| |
Collapse
|
33
|
A computational model of induced pluripotent stem-cell derived cardiomyocytes for high throughput risk stratification of KCNQ1 genetic variants. PLoS Comput Biol 2020; 16:e1008109. [PMID: 32797034 PMCID: PMC7449496 DOI: 10.1371/journal.pcbi.1008109] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/26/2020] [Accepted: 06/30/2020] [Indexed: 01/01/2023] Open
Abstract
In the last decade, there has been tremendous progress in identifying genetic anomalies linked to clinical disease. New experimental platforms have connected genetic variants to mechanisms underlying disruption of cellular and organ behavior and the emergence of proarrhythmic cardiac phenotypes. The development of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) signifies an important advance in the study of genetic disease in a patient-specific context. However, considerable limitations of iPSC-CM technologies have not been addressed: 1) phenotypic variability in apparently identical genotype perturbations, 2) low-throughput electrophysiological measurements, and 3) an immature phenotype which may impact translation to adult cardiac response. We have developed a computational approach intended to address these problems. We applied our recent iPSC-CM computational model to predict the proarrhythmic risk of 40 KCNQ1 genetic variants. An IKs computational model was fit to experimental data for each mutation, and the impact of each mutation was simulated in a population of iPSC-CM models. Using a test set of 15 KCNQ1 mutations with known clinical long QT phenotypes, we developed a method to stratify the effects of KCNQ1 mutations based on proarrhythmic markers. We utilized this method to predict the severity of the remaining 25 KCNQ1 mutations with unknown clinical significance. Tremendous phenotypic variability was observed in the iPSC-CM model population following mutant perturbations. A key novelty is our reporting of the impact of individual KCNQ1 mutant models on adult ventricular cardiomyocyte electrophysiology, allowing for prediction of mutant impact across the continuum of aging. This serves as a first step toward translating predicted response in the iPSC-CM model to predicted response of the adult ventricular myocyte given the same genetic mutation. As a whole, this study presents a new computational framework that serves as a high throughput method to evaluate risk of genetic mutations based-on proarrhythmic behavior in phenotypically variable populations. In the last decade, there has been tremendous progress in identifying genetic mutations linked to clinical diseases, such as cardiac arrhythmia. Many experimental platforms have been developed to study this link, including induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). IPSC-CMs are patient-derived cardiac cells which allow for the study of genetic variants within a patient-specific context. However, experimentally iPSC-CMs have certain limitations, including: (1) they exhibit variability in behavior within cells that are apparently genetically identical, and (2) they are immature compared to adult cardiac cells. In our study, we have developed a computational approach to model 40 genetic variants in the KCNQ1 gene and predict the proarrhythmic risk of each variant. To do this, we modeled the ionic current determined by KCNQ1, IKs, to fit experimental data for each mutation. We then simulated the impact of each mutation in a population of iPSC-CMs, incorporating variability across the population. We also simulated each variant in an adult cardiac cell model, providing a link between iPSC-CM response to mutants and adult cardiac cell response to the same mutants. Overall, this study provides a new computational framework to evaluate risk of genetic mutations based-on proarrhythmic behavior diverse populations of iPSC-CM models.
Collapse
|
34
|
Marcantoni A, Calorio C, Hidisoglu E, Chiantia G, Carbone E. Cav1.2 channelopathies causing autism: new hallmarks on Timothy syndrome. Pflugers Arch 2020; 472:775-789. [PMID: 32621084 DOI: 10.1007/s00424-020-02430-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/23/2020] [Accepted: 06/26/2020] [Indexed: 02/07/2023]
Abstract
Cav1.2 L-type calcium channels play key roles in long-term synaptic plasticity, sensory transduction, muscle contraction, and hormone release. De novo mutations in the gene encoding Cav1.2 (CACNA1C) causes two forms of Timothy syndrome (TS1, TS2), characterized by a multisystem disorder inclusive of cardiac arrhythmias, long QT, autism, and adrenal gland dysfunction. In both TS1 and TS2, the missense mutation G406R is on the alternatively spliced exon 8 and 8A coding for the IS6-helix of Cav1.2 and is responsible for the penetrant form of autism in most TS individuals. The mutation causes specific gain-of-function changes to Cav1.2 channel gating: a "leftward shift" of voltage-dependent activation, reduced voltage-dependent inactivation, and a "leftward shift" of steady-state inactivation. How this occurs and how Cav1.2 gating changes alter neuronal firing and synaptic plasticity is still largely unexplained. Trying to better understanding the molecular basis of Cav1.2 gating dysfunctions leading to autism, here, we will present and discuss the properties of recently reported typical and atypical TS phenotypes and the effective gating changes exhibited by missense mutations associated with long QTs without extracardiac symptoms, unrelated to TS. We will also discuss new emerging views achieved from using iPSCs-derived neurons and the newly available autistic TS2-neo mouse model, both appearing promising for understanding neuronal mistuning in autistic TS patients. We will also analyze and describe recent proposals of molecular pathways that might explain mistuned Ca2+-mediated and Ca2+-independent excitation-transcription signals to the nucleus. Briefly, we will also discuss possible pharmacological approaches to treat autism associated with L-type channelopathies.
Collapse
Affiliation(s)
- Andrea Marcantoni
- Department of Drug Science, Laboratory of Cellular and Molecular Neuroscience, N.I.S. Centre, Corso Raffaello 30, 10125, Torino, Italy
| | - Chiara Calorio
- Department of Drug Science, Laboratory of Cellular and Molecular Neuroscience, N.I.S. Centre, Corso Raffaello 30, 10125, Torino, Italy
| | - Enis Hidisoglu
- Department of Biophysics, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Giuseppe Chiantia
- Department of Drug Science, Laboratory of Cellular and Molecular Neuroscience, N.I.S. Centre, Corso Raffaello 30, 10125, Torino, Italy
| | - Emilio Carbone
- Department of Drug Science, Laboratory of Cellular and Molecular Neuroscience, N.I.S. Centre, Corso Raffaello 30, 10125, Torino, Italy.
| |
Collapse
|
35
|
Becker N, Horváth A, De Boer T, Fabbri A, Grad C, Fertig N, George M, Obergrussberger A. Automated Dynamic Clamp for Simulation of I
K1
in Human Induced Pluripotent Stem Cell–Derived Cardiomyocytes in Real Time Using Patchliner Dynamite
8. ACTA ACUST UNITED AC 2019; 88:e70. [DOI: 10.1002/cpph.70] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
| | | | - Teun De Boer
- Department of Medical PhysiologyUniversity Medical Center Utrecht Utrecht The Netherlands
| | - Alan Fabbri
- Department of Medical PhysiologyUniversity Medical Center Utrecht Utrecht The Netherlands
| | | | | | | | | |
Collapse
|
36
|
Garg P, Garg V, Shrestha R, Sanguinetti MC, Kamp TJ, Wu JC. Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes as Models for Cardiac Channelopathies: A Primer for Non-Electrophysiologists. Circ Res 2019; 123:224-243. [PMID: 29976690 DOI: 10.1161/circresaha.118.311209] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Life threatening ventricular arrhythmias leading to sudden cardiac death are a major cause of morbidity and mortality. In the absence of structural heart disease, these arrhythmias, especially in the younger population, are often an outcome of genetic defects in specialized membrane proteins called ion channels. In the heart, exceptionally well-orchestrated activity of a diversity of ion channels mediates the cardiac action potential. Alterations in either the function or expression of these channels can disrupt the configuration of the action potential, leading to abnormal electrical activity of the heart that can sometimes initiate an arrhythmia. Understanding the pathophysiology of inherited arrhythmias can be challenging because of the complexity of the disorder and lack of appropriate cellular and in vivo models. Recent advances in human induced pluripotent stem cell technology have provided remarkable progress in comprehending the underlying mechanisms of ion channel disorders or channelopathies by modeling these complex arrhythmia syndromes in vitro in a dish. To fully realize the potential of induced pluripotent stem cells in elucidating the mechanistic basis and complex pathophysiology of channelopathies, it is crucial to have a basic knowledge of cardiac myocyte electrophysiology. In this review, we will discuss the role of the various ion channels in cardiac electrophysiology and the molecular and cellular mechanisms of arrhythmias, highlighting the promise of human induced pluripotent stem cell-cardiomyocytes as a model for investigating inherited arrhythmia syndromes and testing antiarrhythmic strategies. Overall, this review aims to provide a basic understanding of the electrical activity of the heart and related channelopathies, especially to clinicians or research scientists in the cardiovascular field with limited electrophysiology background.
Collapse
Affiliation(s)
- Priyanka Garg
- From the Stanford Cardiovascular Institute (P.G., R.S., J.C.W.).,Department of Medicine, Division of Cardiology (P.G., R.S., J.C.W.).,Institute for Stem Cell Biology and Regenerative Medicine (P.G., R.S., J.C.W.)
| | - Vivek Garg
- Stanford University School of Medicine, CA; Department of Physiology, University of California San Francisco (V.G.)
| | - Rajani Shrestha
- From the Stanford Cardiovascular Institute (P.G., R.S., J.C.W.).,Department of Medicine, Division of Cardiology (P.G., R.S., J.C.W.).,Institute for Stem Cell Biology and Regenerative Medicine (P.G., R.S., J.C.W.)
| | | | - Timothy J Kamp
- Department of Medicine, University of Wisconsin-Madison (T.J.K.)
| | - Joseph C Wu
- From the Stanford Cardiovascular Institute (P.G., R.S., J.C.W.) .,Department of Medicine, Division of Cardiology (P.G., R.S., J.C.W.).,Institute for Stem Cell Biology and Regenerative Medicine (P.G., R.S., J.C.W.)
| |
Collapse
|
37
|
Fabbri A, Goversen B, Vos MA, van Veen TAB, de Boer TP. Required G K1 to Suppress Automaticity of iPSC-CMs Depends Strongly on I K1 Model Structure. Biophys J 2019; 117:2303-2315. [PMID: 31623886 PMCID: PMC6990378 DOI: 10.1016/j.bpj.2019.08.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 07/24/2019] [Accepted: 08/26/2019] [Indexed: 01/06/2023] Open
Abstract
Human-induced pluripotent stem cells derived cardiomyocytes (hiPSC-CMs) are a virtually endless source of human cardiomyocytes that may become a great tool for safety pharmacology; however, their electrical phenotype is immature: they show spontaneous action potentials (APs) and an unstable and depolarized resting membrane potential (RMP) because of lack of IK1. Such immaturity hampers their application in assessing drug safety. The electronic overexpression of IK1 (e.g., through the dynamic clamp (DC) technique) is an option to overcome this deficit. In this computational study, we aim to estimate how much IK1 is needed to bring hiPSC-CMs to a stable and hyperpolarized RMP and which mathematical description of IK1 is most suitable for DC experiments. We compared five mature IK1 formulations (Bett, Dhamoon, Ishihara, O’Hara-Rudy, and ten Tusscher) with the native one (Paci), evaluating the main properties (outward peak, degree of rectification), and we quantified their effects on AP features (RMP, V˙max, APD50, APD90 (AP duration at 50 and 90% of repolarization), and APD50/APD90) after including them in the hiPSC-CM mathematical model by Paci. Then, we automatically identified the critical conductance for IK1 ( GK1, critical), the minimally required amount of IK1 suppressing spontaneous activity. Preconditioning the cell model with depolarizing/hyperpolarizing prepulses allowed us to highlight time dependency of the IK1 formulations. Simulations showed that inclusion of mature IK1 formulations resulted in hyperpolarized RMP and higher V˙max, and observed GK1, critical and the effect on AP duration strongly depended on IK1 formulation. Finally, the Ishihara IK1 led to shorter (−16.3%) and prolonged (+6.5%) APD90 in response to hyperpolarizing and depolarizing prepulses, respectively, whereas other models showed negligible effects. Fine-tuning of GK1 is an important step in DC experiments. Our computational work proposes a procedure to automatically identify how much IK1 current is required to inject to stop the spontaneous activity and suggests the use of the Ishihara IK1 model to perform DC experiments in hiPSC-CMs.
Collapse
Affiliation(s)
- Alan Fabbri
- University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Marc A Vos
- University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Teun P de Boer
- University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
38
|
Treat JA, Goodrow RJ, Bot CT, Haedo RJ, Cordeiro JM. Pharmacological enhancement of repolarization reserve in human induced pluripotent stem cells derived cardiomyocytes. Biochem Pharmacol 2019; 169:113608. [PMID: 31465775 DOI: 10.1016/j.bcp.2019.08.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 08/14/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are used for many applications including safety pharmacology. However, a deficiency or complete absence of several K+ currents suggests repolarization reserve is low in hiPSC-CMs. We determined whether a dual Ito and IKr activator can improve repolarization reserve in hiPSC-CMs resulting in a more electrophysiologically mature phenotype. METHODS AND RESULTS Human iPSC were maintained on growth factor and differentiated into the cardiac phenotype by addition of selective Wnt molecules. Current and voltage clamp recordings in single cells were made using patch electrodes. Extracellular field potentials were made using a microelectrode array on hiPSC monolayers. Action potential recordings from hiPSC-CMs following application of an IKr inhibitor resulted in depolarization of the membrane potential and prolongation of the APD. A flattening of the T-wave was noted on the pseudo-ECG. In contrast, application of the IKr and Ito agonist, NS3623, resulted in hyperpolarization of the membrane, slowing of the spontaneous rate and shortening of the APD. Voltage clamp recording showed a significant increase in IKr; no enhancement of Ito in hiPSC-CMs was noted. AP clamp experiments revealed that IKr plays a role in both phase 3 repolarization and phase 4 depolarization. mRNA analysis revealed that KCNH2 is abundantly expressed in hiPSC-CM, consistent with electrophysiological recordings. CONCLUSIONS Although NS3623 is a dual Ito and IKr activator in ventricular myocytes, application of this compound to hiPSC-CMs enhanced only IKr and no effect on Ito was noted. Our results suggest IKr enhancement can improve repolarization reserve in this cell type. The disconnect between a dramatic increase in Ito in adult myocytes versus the lack of effect in hiPSC-CMs suggest that the translation of pharmacological effects in hiPSC-CM to adult myocytes should be viewed with caution.
Collapse
Affiliation(s)
- Jacqueline A Treat
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY 13501 USA
| | - Robert J Goodrow
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY 13501 USA
| | - Corina T Bot
- Nanion Technologies, 1 Naylon Ave. Suite C, Livingston, NJ 07039, USA
| | - Rodolfo J Haedo
- Nanion Technologies, 1 Naylon Ave. Suite C, Livingston, NJ 07039, USA
| | - Jonathan M Cordeiro
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY 13501 USA.
| |
Collapse
|
39
|
Phenotype-Based High-Throughput Classification of Long QT Syndrome Subtypes Using Human Induced Pluripotent Stem Cells. Stem Cell Reports 2019; 13:394-404. [PMID: 31378668 PMCID: PMC6700479 DOI: 10.1016/j.stemcr.2019.06.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 06/29/2019] [Accepted: 06/30/2019] [Indexed: 11/23/2022] Open
Abstract
For long QT syndrome (LQTS), recent progress in genome-sequencing technologies enabled the identification of rare genomic variants with diagnostic, prognostic, and therapeutic implications. However, pathogenic stratification of the identified variants remains challenging, especially in variants of uncertain significance. This study aimed to propose a phenotypic cell-based diagnostic assay for identifying LQTS to recognize pathogenic variants in a high-throughput manner suitable for screening. We investigated the response of LQT2-induced pluripotent stem cell (iPSC)-derived cardiomyocytes (iPSC-CMs) following IKr blockade using a multi-electrode array, finding that the response to IKr blockade was significantly smaller than in Control-iPSC-CMs. Furthermore, we found that LQT1-iPSC-CMs and LQT3-iPSC-CMs could be distinguished from Control-iPSC-CMs by IKs blockade and INa blockade, respectively. This strategy might be helpful in compensating for the shortcomings of genetic testing of LQTS patients.
Collapse
|
40
|
Kernik DC, Morotti S, Wu H, Garg P, Duff HJ, Kurokawa J, Jalife J, Wu JC, Grandi E, Clancy CE. A computational model of induced pluripotent stem-cell derived cardiomyocytes incorporating experimental variability from multiple data sources. J Physiol 2019; 597:4533-4564. [PMID: 31278749 PMCID: PMC6767694 DOI: 10.1113/jp277724] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 07/05/2019] [Indexed: 12/22/2022] Open
Abstract
Key points Induced pluripotent stem cell‐derived cardiomyocytes (iPSC‐CMs) capture patient‐specific genotype–phenotype relationships, as well as cell‐to‐cell variability of cardiac electrical activity Computational modelling and simulation provide a high throughput approach to reconcile multiple datasets describing physiological variability, and also identify vulnerable parameter regimes We have developed a whole‐cell model of iPSC‐CMs, composed of single exponential voltage‐dependent gating variable rate constants, parameterized to fit experimental iPSC‐CM outputs We have utilized experimental data across multiple laboratories to model experimental variability and investigate subcellular phenotypic mechanisms in iPSC‐CMs This framework links molecular mechanisms to cellular‐level outputs by revealing unique subsets of model parameters linked to known iPSC‐CM phenotypes
Abstract There is a profound need to develop a strategy for predicting patient‐to‐patient vulnerability in the emergence of cardiac arrhythmia. A promising in vitro method to address patient‐specific proclivity to cardiac disease utilizes induced pluripotent stem cell‐derived cardiomyocytes (iPSC‐CMs). A major strength of this approach is that iPSC‐CMs contain donor genetic information and therefore capture patient‐specific genotype–phenotype relationships. A cited detriment of iPSC‐CMs is the cell‐to‐cell variability observed in electrical activity. We postulated, however, that cell‐to‐cell variability may constitute a strength when appropriately utilized in a computational framework to build cell populations that can be employed to identify phenotypic mechanisms and pinpoint key sensitive parameters. Thus, we have exploited variation in experimental data across multiple laboratories to develop a computational framework for investigating subcellular phenotypic mechanisms. We have developed a whole‐cell model of iPSC‐CMs composed of simple model components comprising ion channel models with single exponential voltage‐dependent gating variable rate constants, parameterized to fit experimental iPSC‐CM data for all major ionic currents. By optimizing ionic current model parameters to multiple experimental datasets, we incorporate experimentally‐observed variability in the ionic currents. The resulting population of cellular models predicts robust inter‐subject variability in iPSC‐CMs. This approach links molecular mechanisms to known cellular‐level iPSC‐CM phenotypes, as shown by comparing immature and mature subpopulations of models to analyse the contributing factors underlying each phenotype. In the future, the presented models can be readily expanded to include genetic mutations and pharmacological interventions for studying the mechanisms of rare events, such as arrhythmia triggers. Induced pluripotent stem cell‐derived cardiomyocytes (iPSC‐CMs) capture patient‐specific genotype–phenotype relationships, as well as cell‐to‐cell variability of cardiac electrical activity Computational modelling and simulation provide a high throughput approach to reconcile multiple datasets describing physiological variability, and also identify vulnerable parameter regimes We have developed a whole‐cell model of iPSC‐CMs, composed of single exponential voltage‐dependent gating variable rate constants, parameterized to fit experimental iPSC‐CM outputs We have utilized experimental data across multiple laboratories to model experimental variability and investigate subcellular phenotypic mechanisms in iPSC‐CMs This framework links molecular mechanisms to cellular‐level outputs by revealing unique subsets of model parameters linked to known iPSC‐CM phenotypes
Collapse
Affiliation(s)
- Divya C Kernik
- Department of Physiology and Membrane Biology, Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, USA
| | - Stefano Morotti
- Department of Pharmacology, School of Medicine, University of California, Davis, CA, USA
| | - HaoDi Wu
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Priyanka Garg
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Henry J Duff
- Libin Cardiovascular Institute of Alberta, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| | - Junko Kurokawa
- Department of Bio-Informational Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - José Jalife
- Department of Internal Medicine, Center for Arrhythmia Research, Cardiovascular Research Center, University of Michigan, Ann Arbor, MI, USA.,Centro Nacional de Investigaciones Cardiovasculares (CNIC), and CIBERV, Madrid, Spain
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Eleonora Grandi
- Department of Pharmacology, School of Medicine, University of California, Davis, CA, USA
| | - Colleen E Clancy
- Department of Physiology and Membrane Biology, Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, USA
| |
Collapse
|
41
|
Han D, Xue X, Yan Y, Li G. Dysfunctional Cav1.2 channel in Timothy syndrome, from cell to bedside. Exp Biol Med (Maywood) 2019; 244:960-971. [PMID: 31324123 DOI: 10.1177/1535370219863149] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Timothy syndrome is a rare disorder caused by CACNA1C gene mutations and characterized by multi-organ system dysfunctions, including ventricular arrhythmias, syndactyly, dysmorphic facial features, intermittent hypoglycemia, immunodeficiency, developmental delay, and autism. Because of the low morbidity and high mortality at a young age, it remains a huge challenge to establish a diagnosis and treatment system to manage Timothy syndrome patients. Here, we aim to provide a detailed review of Timothy syndrome, discuss the mechanisms underlying dysfunctional Cav1.2 due to CACNA1C mutations, and provide some new emerging evidences in treating Timothy syndrome from cell to bedside, promoting the management of this rare disease. Impact statement The knowledge of Timothy syndrome (TS) caused by dysfunctional Cav1.2 channel due to CACNA1C mutations is rapidly evolving as novel technologies of electrophysiology are introduced and our understanding of the mechanisms of TS develops. In this review, we focus on the TS-related dysfunctional Cav1.2 and the underlying mechanisms. We update TS-related CACNA1C mutations in a precise way over the past 20 years and summarize all reported TS patients based on their clinical presentations and molecular mechanisms, respectively. We hope this review will provide a new comprehensive way to better understand the electrophysiological mechanisms underlying TS from cell to bedside, promoting the management of TS in practice.
Collapse
Affiliation(s)
- Dan Han
- 1 Department of Cardiology, the First Affiliated Hospital of Xi'an Jiaotong University, Shaanxi 710061, P. R. China.,2 Department of Cardiovascular Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Shaanxi 710061, P.R. China*These authors contributed equally to this work and should be considered to share first authorship
| | - Xiaolin Xue
- 1 Department of Cardiology, the First Affiliated Hospital of Xi'an Jiaotong University, Shaanxi 710061, P. R. China
| | - Yang Yan
- 2 Department of Cardiovascular Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Shaanxi 710061, P.R. China*These authors contributed equally to this work and should be considered to share first authorship
| | - Guoliang Li
- 1 Department of Cardiology, the First Affiliated Hospital of Xi'an Jiaotong University, Shaanxi 710061, P. R. China
| |
Collapse
|
42
|
Pfeiffer-Kaushik ER, Smith GL, Cai B, Dempsey GT, Hortigon-Vinagre MP, Zamora V, Feng S, Ingermanson R, Zhu R, Hariharan V, Nguyen C, Pierson J, Gintant GA, Tung L. Electrophysiological characterization of drug response in hSC-derived cardiomyocytes using voltage-sensitive optical platforms. J Pharmacol Toxicol Methods 2019; 99:106612. [PMID: 31319140 DOI: 10.1016/j.vascn.2019.106612] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 06/30/2019] [Accepted: 07/10/2019] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Voltage-sensitive optical (VSO) sensors offer a minimally invasive method to study the time course of repolarization of the cardiac action potential (AP). This Comprehensive in vitro Proarrhythmia Assay (CiPA) cross-platform study investigates protocol design and measurement variability of VSO sensors for preclinical cardiac electrophysiology assays. METHODS Three commercial and one academic laboratory completed a limited study of the effects of 8 blinded compounds on the electrophysiology of 2 commercial lines of human induced pluripotent stem-cell derived cardiomyocytes (hSC-CMs). Acquisition technologies included CMOS camera and photometry; fluorescent voltage sensors included di-4-ANEPPS, FluoVolt and genetically encoded QuasAr2. The experimental protocol was standardized with respect to cell lines, plating and maintenance media, blinded compounds, and action potential parameters measured. Serum-free media was used to study the action of drugs, but the exact composition and the protocols for cell preparation and drug additions varied among sites. RESULTS Baseline AP waveforms differed across platforms and between cell types. Despite these differences, the relative responses to four selective ion channel blockers (E-4031, nifedipine, mexiletine, and JNJ 303 blocking IKr, ICaL, INa, and IKs, respectively) were similar across all platforms and cell lines although the absolute changes differed. Similarly, four mixed ion channel blockers (flecainide, moxifloxacin, quinidine, and ranolazine) had comparable effects in all platforms. Differences in repolarisation time course and response to drugs could be attributed to cell type and experimental method differences such as composition of the assay media, stimulated versus spontaneous activity, and single versus cumulative compound addition. DISCUSSION In conclusion, VSOs represent a powerful and appropriate method to assess the electrophysiological effects of drugs on iPSC-CMs for the evaluation of proarrhythmic risk. Protocol considerations and recommendations are provided toward standardizing conditions to reduce variability of baseline AP waveform characteristics and drug responses.
Collapse
Affiliation(s)
| | - Godfrey L Smith
- Clyde Biosciences Ltd, BioCity Scotland, Bo'Ness Road, Newhouse, Lanarkshire, Scotland ML1 5UH, United Kingdom; Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Science, University of Glasgow, 126 University Place, Glasgow G12 8TA, United Kingdom
| | - Beibei Cai
- Vala Sciences Inc., 6370 Nancy Ridge Drive, Suite 106, San Diego, CA 92121, USA
| | - Graham T Dempsey
- Q-State Biosciences Inc., 179 Sidney Street, Cambridge, MA 02139, USA
| | - Maria P Hortigon-Vinagre
- Clyde Biosciences Ltd, BioCity Scotland, Bo'Ness Road, Newhouse, Lanarkshire, Scotland ML1 5UH, United Kingdom; Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Science, University of Glasgow, 126 University Place, Glasgow G12 8TA, United Kingdom
| | - Victor Zamora
- Clyde Biosciences Ltd, BioCity Scotland, Bo'Ness Road, Newhouse, Lanarkshire, Scotland ML1 5UH, United Kingdom; Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Science, University of Glasgow, 126 University Place, Glasgow G12 8TA, United Kingdom
| | - Shuyun Feng
- Vala Sciences Inc., 6370 Nancy Ridge Drive, Suite 106, San Diego, CA 92121, USA
| | - Randall Ingermanson
- Vala Sciences Inc., 6370 Nancy Ridge Drive, Suite 106, San Diego, CA 92121, USA
| | - Renjun Zhu
- Department of Biomedical Engineering, The Johns Hopkins University, 720 Rutland Ave., Baltimore, MD 21205, USA
| | - Venkatesh Hariharan
- Department of Biomedical Engineering, The Johns Hopkins University, 720 Rutland Ave., Baltimore, MD 21205, USA
| | - Cuong Nguyen
- Q-State Biosciences Inc., 179 Sidney Street, Cambridge, MA 02139, USA
| | - Jennifer Pierson
- Health and Environmental Sciences Institute, Washington, D.C. 20009, USA.
| | - Gary A Gintant
- AbbVie, 1 North Waukegan Road, Department ZR-13, Building AP-9A, North Chicago, IL 60064-6119, USA
| | - Leslie Tung
- Department of Biomedical Engineering, The Johns Hopkins University, 720 Rutland Ave., Baltimore, MD 21205, USA
| |
Collapse
|
43
|
Wang Y, Zhu R, Tung L. Contribution of potassium channels to action potential repolarization of human embryonic stem cell-derived cardiomyocytes. Br J Pharmacol 2019; 176:2780-2794. [PMID: 31074016 DOI: 10.1111/bph.14704] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 03/11/2019] [Accepted: 03/29/2019] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND AND PURPOSE The electrophysiological properties of human pluripotent stem cell-derived cardiomyocytes (CMs) have not yet been characterized in a syncytial context. This study systematically characterized the contributions of different repolarizing potassium currents in human embryonic stem cell-derived CMs (hESC-CMs) during long-term culture as cell monolayers. EXPERIMENTAL APPROACH The H9 hESC line was differentiated to CMs and plated to form confluent cell monolayers. Optical mapping was used to record the action potentials (APs) and conduction velocity (CV) during electrophysiological and pharmacological experiments. RT-PCR and Western blot were used to detect the presence and expression levels of ion channel subunits. KEY RESULTS Long-term culture of hESC-CMs led to shortened AP duration (APD), faster repolarization rate, and increased CV. Selective block of IKr , IKs , IK1 , and IKur significantly affected AP repolarization and APD in a concentration- and culture time-dependent manner. Baseline variations in APD led to either positive or negative APD dependence of drug response. Chromanol 293B produced greater relative AP prolongation in mid- and late-stage cultures, while DPO-1 had more effect in early-stage cultures. CV in cell monolayers in early- and late-stage cultures was most susceptible to slowing by E-4031 and BaCl2 respectively. CONCLUSIONS AND IMPLICATIONS IKr , IKs , IK1 , and IKur all play an essential role in the regulation of APD and CV in hESC-CMs. During time in culture, increased expression of IKr and IK1 helps to accelerate repolarization, shorten APD, and increase CV. We identified a new pro-arrhythmic parameter, positive APD dependence of ion channel block, which can increase APD and repolarization gradients.
Collapse
Affiliation(s)
- Yin Wang
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, USA.,Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Renjun Zhu
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Leslie Tung
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
44
|
Mann SA, Heide J, Knott T, Airini R, Epureanu FB, Deftu AF, Deftu AT, Radu BM, Amuzescu B. Recording of multiple ion current components and action potentials in human induced pluripotent stem cell-derived cardiomyocytes via automated patch-clamp. J Pharmacol Toxicol Methods 2019; 100:106599. [PMID: 31228558 DOI: 10.1016/j.vascn.2019.106599] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/06/2019] [Accepted: 06/14/2019] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The Comprehensive in vitro Proarrhythmia Assay (CiPA) initiative proposes a three-step approach to evaluate proarrhythmogenic liability of drug candidates: effects on individual ion channels in heterologous expression systems, integrating these data into in-silico models of the electrical activity of human cardiomyocytes, and comparison with experiments on human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM). Here we introduce patch-clamp electrophysiology techniques on hiPSC-CM to combine two of the CiPA steps in one assay. METHODS We performed automated patch-clamp experiments on hiPSC-CM (Cor.4U®, Ncardia) using the CytoPatch™2 platform in ruptured whole-cell and β-escin-perforated-patch configurations. A combination of three voltage-clamp protocols allowed recording of five distinct ion current components (voltage-gated Na+ current, L-type Ca2+ current, transient outward K+ current, delayed rectifier K+ current, and "funny" hyperpolarization-activated current) from the same cell. We proved their molecular identity by either Na+ replacement with choline or by applying specific blockers: nifedipine, cisapride, chromanol 293B, phrixotoxin-1, ZD7288. We developed a C++ script for automated analysis of voltage-clamp recordings and computation of ion current/conductance surface density for these five cardiac ion currents. RESULTS The distributions from n = 54 hiPSC-CM in "ruptured" patch-clamp vs. n = 35 hiPSC-CM in β-escin-perforated patch-clamp were similar for membrane capacitance, access resistance, and ion current/conductance surface densities. The β-escin-perforated configuration resulted in improved stability of action potential (AP) shape and duration over a 10-min interval, with APD90 decay rate 0.7 ± 1.6%/min (mean ± SD, n = 4) vs. 4.6 ± 1.1%/min. (n = 3) for "ruptured" approach (p = 0.0286, one-tailed Mann-Whitney test). DISCUSSION The improved stability obtained here will allow development of CiPA-compliant automated patch-clamp assays on hiPSC-CM. Future applications include the study of multi ion-channel blocking properties of drugs using dynamic-clamp protocols, adding a valuable new tool to the arsenal of safety-pharmacology.
Collapse
Affiliation(s)
- Stefan A Mann
- Cytocentrics Bioscience GmbH, Nattermannallee 1, 50829 Cologne, Germany
| | - Juliane Heide
- Cytocentrics Bioscience GmbH, Nattermannallee 1, 50829 Cologne, Germany
| | - Thomas Knott
- CytoBioScience Inc., 3463 Magic Drive, San Antonio, TX 78229, USA
| | - Razvan Airini
- Dept. Biophysics & Physiology, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania
| | - Florin Bogdan Epureanu
- Dept. Biophysics & Physiology, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania
| | - Alexandru-Florian Deftu
- Dept. Biophysics & Physiology, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania
| | - Antonia-Teona Deftu
- Dept. Biophysics & Physiology, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania
| | - Beatrice Mihaela Radu
- Dept. Biophysics & Physiology, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania
| | - Bogdan Amuzescu
- Dept. Biophysics & Physiology, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania.
| |
Collapse
|
45
|
Salvarani N, Crasto S, Miragoli M, Bertero A, Paulis M, Kunderfranco P, Serio S, Forni A, Lucarelli C, Dal Ferro M, Larcher V, Sinagra G, Vezzoni P, Murry CE, Faggian G, Condorelli G, Di Pasquale E. The K219T-Lamin mutation induces conduction defects through epigenetic inhibition of SCN5A in human cardiac laminopathy. Nat Commun 2019; 10:2267. [PMID: 31118417 PMCID: PMC6531493 DOI: 10.1038/s41467-019-09929-w] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 04/06/2019] [Indexed: 12/14/2022] Open
Abstract
Mutations in LMNA, which encodes the nuclear proteins Lamin A/C, can cause cardiomyopathy and conduction disorders. Here, we employ induced pluripotent stem cells (iPSCs) generated from human cells carrying heterozygous K219T mutation on LMNA to develop a disease model. Cardiomyocytes differentiated from these iPSCs, and which thus carry K219T-LMNA, have altered action potential, reduced peak sodium current and diminished conduction velocity. Moreover, they have significantly downregulated Nav1.5 channel expression and increased binding of Lamin A/C to the promoter of SCN5A, the channel's gene. Coherently, binding of the Polycomb Repressive Complex 2 (PRC2) protein SUZ12 and deposition of the repressive histone mark H3K27me3 are increased at SCN5A. CRISPR/Cas9-mediated correction of the mutation re-establishes sodium current density and SCN5A expression. Thus, K219T-LMNA cooperates with PRC2 in downregulating SCN5A, leading to decreased sodium current density and slower conduction velocity. This mechanism may underlie the conduction abnormalities associated with LMNA-cardiomyopathy.
Collapse
Affiliation(s)
- Nicolò Salvarani
- Institute of Genetic and Biomedical Research (IRGB), UOS of Milan, National Research Council of Italy, Milan, 20138, Italy
- Department of Cardiovascular Medicine and Laboratory of Medical Biotechnology, Humanitas Clinical and Research Center - IRCCS, Rozzano (MI), 20089, Italy
| | - Silvia Crasto
- Institute of Genetic and Biomedical Research (IRGB), UOS of Milan, National Research Council of Italy, Milan, 20138, Italy
- Department of Cardiovascular Medicine and Laboratory of Medical Biotechnology, Humanitas Clinical and Research Center - IRCCS, Rozzano (MI), 20089, Italy
| | - Michele Miragoli
- Institute of Genetic and Biomedical Research (IRGB), UOS of Milan, National Research Council of Italy, Milan, 20138, Italy
- Department of Cardiovascular Medicine and Laboratory of Medical Biotechnology, Humanitas Clinical and Research Center - IRCCS, Rozzano (MI), 20089, Italy
- Department of Medicine and Surgery, University of Parma, Parma, 43121, Italy
| | - Alessandro Bertero
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, 98109, WA, USA
| | - Marianna Paulis
- Institute of Genetic and Biomedical Research (IRGB), UOS of Milan, National Research Council of Italy, Milan, 20138, Italy
- Department of Cardiovascular Medicine and Laboratory of Medical Biotechnology, Humanitas Clinical and Research Center - IRCCS, Rozzano (MI), 20089, Italy
| | - Paolo Kunderfranco
- Department of Cardiovascular Medicine and Laboratory of Medical Biotechnology, Humanitas Clinical and Research Center - IRCCS, Rozzano (MI), 20089, Italy
| | - Simone Serio
- Department of Cardiovascular Medicine and Laboratory of Medical Biotechnology, Humanitas Clinical and Research Center - IRCCS, Rozzano (MI), 20089, Italy
| | - Alberto Forni
- Division of Cardiac Surgery, University of Verona, Verona, 37129, Italy
| | - Carla Lucarelli
- Division of Cardiac Surgery, University of Verona, Verona, 37129, Italy
| | - Matteo Dal Ferro
- Cardiovascular Department, "Ospedali Riuniti" and University of Trieste, Trieste, 34129, Italy
| | - Veronica Larcher
- Department of Cardiovascular Medicine and Laboratory of Medical Biotechnology, Humanitas Clinical and Research Center - IRCCS, Rozzano (MI), 20089, Italy
| | - Gianfranco Sinagra
- Cardiovascular Department, "Ospedali Riuniti" and University of Trieste, Trieste, 34129, Italy
| | - Paolo Vezzoni
- Institute of Genetic and Biomedical Research (IRGB), UOS of Milan, National Research Council of Italy, Milan, 20138, Italy
- Department of Cardiovascular Medicine and Laboratory of Medical Biotechnology, Humanitas Clinical and Research Center - IRCCS, Rozzano (MI), 20089, Italy
| | - Charles E Murry
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, 98109, WA, USA
| | - Giuseppe Faggian
- Division of Cardiac Surgery, University of Verona, Verona, 37129, Italy
| | - Gianluigi Condorelli
- Institute of Genetic and Biomedical Research (IRGB), UOS of Milan, National Research Council of Italy, Milan, 20138, Italy.
- Department of Cardiovascular Medicine and Laboratory of Medical Biotechnology, Humanitas Clinical and Research Center - IRCCS, Rozzano (MI), 20089, Italy.
- Humanitas University, Rozzano (MI), 20089, Italy.
| | - Elisa Di Pasquale
- Institute of Genetic and Biomedical Research (IRGB), UOS of Milan, National Research Council of Italy, Milan, 20138, Italy.
- Department of Cardiovascular Medicine and Laboratory of Medical Biotechnology, Humanitas Clinical and Research Center - IRCCS, Rozzano (MI), 20089, Italy.
| |
Collapse
|
46
|
Grubb S, Vestergaard ML, Andersen AS, Rasmussen KK, Mamsen LS, Tuckute G, Grunnet-Lauridsen K, Møllgård K, Ernst E, Christensen ST, Calloe K, Andersen CY. Comparison of Cultured Human Cardiomyocyte Clusters Obtained from Embryos/Fetuses or Derived from Human Embryonic Stem Cells. Stem Cells Dev 2019; 28:608-619. [PMID: 30755084 DOI: 10.1089/scd.2018.0231] [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: 01/05/2023] Open
Abstract
Cardiomyocytes (CMs) derived from human embryonic stem cells (hESCs) or induced pluripotent stem cells (iPSCs) are used to study cardiogenesis and mechanisms of heart disease, and are being used in methods for toxiological screening of drugs. The phenotype of stem-cell-derived CMs should ideally resemble native CMs. Here, we compare embryonic/fetal CMs with hESC-derived CMs according to function and morphology. CM clusters were obtained from human embryonic/fetal hearts from elective terminated pregnancies before gestational week 12, and separated into atrial and ventricular tissues. Specific markers for embryonic CMs and primary cilia were visualized using immunofluorescence microscopy analysis. Contracting human embryonic cardiomyocyte (hECM) clusters morphologically and phenotypically resemble CMs in the embryonic/fetal heart. In addition, the contracting hECM clusters expressed primary cilia similar to that of cells in the embryonic/fetal heart. The electrophysiological characteristics of atrial and ventricular CMs were established by recording action potentials (APs) using sharp electrodes. In contrast to ventricular APs, atrial APs displayed a marked early repolarization followed by a plateau phase. hESC-CMs displayed a continuum of AP shapes. In all embryonic/fetal clusters, both atrial and ventricular, AP duration was prolonged by exposure to the KV11.1 channel inhibitor dofetilide (50 nM); however, the prolongation was not significant, possibly due to the relatively small number of experiments. This study provides novel information on APs and functional characteristics of atrial and ventricular CMs in first trimester hearts, and demonstrates that Kv11.1 channels play a functional role already at these early stages. These results provide information needed to validate methods being developed on the basis of in vitro-derived CMs from either hESC or iPSC, and although there was a good correlation between the morphology of the two types of CMs, differences in electrophysiological characteristics exist.
Collapse
Affiliation(s)
- Søren Grubb
- 1 Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maj Linea Vestergaard
- 2 Laboratory of Reproductive Biology, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Astrid Sten Andersen
- 2 Laboratory of Reproductive Biology, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Karen Koefoed Rasmussen
- 3 Section of Cell and Developmental Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Linn Salto Mamsen
- 2 Laboratory of Reproductive Biology, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Greta Tuckute
- 2 Laboratory of Reproductive Biology, University Hospital of Copenhagen, Copenhagen, Denmark
| | | | - Kjeld Møllgård
- 4 Institute for Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Erik Ernst
- 5 The Department of Gynecology and Obstetrics, University Hospital of Aarhus, Aarhus, Denmark
| | - Søren Tvorup Christensen
- 3 Section of Cell and Developmental Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kirstine Calloe
- 1 Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Claus Yding Andersen
- 2 Laboratory of Reproductive Biology, University Hospital of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
47
|
Wang X, Han Z, Yu Y, Xu Z, Cai B, Yuan Y. Potential Applications of Induced Pluripotent Stem Cells for Cardiovascular Diseases. Curr Drug Targets 2018; 20:763-774. [PMID: 30539693 DOI: 10.2174/1389450120666181211164147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/05/2018] [Accepted: 12/05/2018] [Indexed: 12/13/2022]
Abstract
Owning the high incidence and disability rate in the past decades, to be expected, cardiovascular diseases (CVDs) have become one of the leading death causes worldwide. Currently, induced pluripotent stem cells (iPSCs), with the potential to form fresh myocardium and improve the functions of damaged hearts, have been studied widely in experimental CVD therapy. Moreover, iPSC-derived cardiomyocytes (CMs), as novel disease models, play a significant role in drug screening, drug safety assessment, along with the exploration of pathological mechanisms of diseases. Furthermore, a lot of studies have been carried out to clarify the biological basis of iPSCs and its derived cells in the treatment of CVDs. Their molecular mechanisms were associated with release of paracrine factors, regulation of miRNAs, mechanical support of new tissues, activation of specific pathways and specific enzymes, etc. In addition, a few small chemical molecules and suitable biological scaffolds play positive roles in enhancing the efficiency of iPSC transplantation. This article reviews the development and limitations of iPSCs in CVD therapy, and summarizes the latest research achievements regarding the application of iPSCs in CVDs.
Collapse
Affiliation(s)
- Xiaotong Wang
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Zhenbo Han
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Ying Yu
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Zihang Xu
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Benzhi Cai
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Ye Yuan
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| |
Collapse
|
48
|
Quach B, Krogh-Madsen T, Entcheva E, Christini DJ. Light-Activated Dynamic Clamp Using iPSC-Derived Cardiomyocytes. Biophys J 2018; 115:2206-2217. [PMID: 30447994 PMCID: PMC6289097 DOI: 10.1016/j.bpj.2018.10.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 08/23/2018] [Accepted: 10/02/2018] [Indexed: 01/31/2023] Open
Abstract
iPSC-derived cardiomyocytes (iPSC-CMs) are a potentially advantageous platform for drug screening because they provide a renewable source of human cardiomyocytes. One obstacle to their implementation is their immature electrophysiology, which reduces relevance to adult arrhythmogenesis. To address this, dynamic clamp is used to inject current representing the insufficient potassium current, IK1, thereby producing more adult-like electrophysiology. However, dynamic clamp requires patch clamp and is therefore low throughput and ill-suited for large-scale drug screening. Here, we use optogenetics to generate such a dynamic-clamp current. The optical dynamic clamp (ODC) uses outward-current-generating opsin, ArchT, to mimic IK1, resulting in more adult-like action potential morphology, similar to IK1 injection via classic dynamic clamp. Furthermore, in the presence of an IKr blocker, ODC revealed expected action potential prolongation and reduced spontaneous excitation. The ODC presented here still requires an electrode to measure Vm but provides a first step toward contactless dynamic clamp, which will not only enable high-throughput screening but may also allow control within multicellular iPSC-CM formats to better recapitulate adult in vivo physiology.
Collapse
Affiliation(s)
- Bonnie Quach
- Cardiovascular Research Institute, New York, New York; Weill Cornell Medicine, New York, New York
| | - Trine Krogh-Madsen
- Cardiovascular Research Institute, New York, New York; Weill Cornell Medicine, New York, New York
| | - Emilia Entcheva
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia
| | - David J Christini
- Cardiovascular Research Institute, New York, New York; Weill Cornell Medicine, New York, New York.
| |
Collapse
|
49
|
de Boer TP. Using Light to Endow Stem-Cell-Derived Cardiomyocytes With Virtual I K1 Conductances. Biophys J 2018; 115:2079-2080. [PMID: 30442328 DOI: 10.1016/j.bpj.2018.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 10/19/2018] [Accepted: 10/24/2018] [Indexed: 11/17/2022] Open
Affiliation(s)
- Teun P de Boer
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
50
|
Moreau A, Chahine M. A New Cardiac Channelopathy: From Clinical Phenotypes to Molecular Mechanisms Associated With Na v1.5 Gating Pores. Front Cardiovasc Med 2018; 5:139. [PMID: 30356750 PMCID: PMC6189448 DOI: 10.3389/fcvm.2018.00139] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/19/2018] [Indexed: 12/19/2022] Open
Abstract
Voltage gated sodium channels (NaV) are broadly expressed in the human body. They are responsible for the initiation of action potentials in excitable cells. They also underlie several physiological processes such as cognitive, sensitive, motor, and cardiac functions. The NaV1.5 channel is the main NaV expressed in the heart. A dysfunction of this channel is usually associated with the development of pure electrical disorders such as long QT syndrome, Brugada syndrome, sinus node dysfunction, atrial fibrillation, and cardiac conduction disorders. However, mutations of Nav1.5 have recently been linked to the development of an atypical clinical entity combining complex arrhythmias and dilated cardiomyopathy. Although several Nav1.5 mutations have been linked to dilated cardiomyopathy phenotypes, their pathogenic mechanisms remain to be elucidated. The gating pore may constitute a common biophysical defect for all NaV1.5 mutations located in the channel's VSDs. The creation of such a gating pore may disrupt the ionic homeostasis of cardiomyocytes, affecting electrical signals, cell morphology, and cardiac myocyte function. The main objective of this article is to review the concept of gating pores and their role in structural heart diseases and to discuss potential pharmacological treatments.
Collapse
Affiliation(s)
- Adrien Moreau
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Mohamed Chahine
- CERVO Research Centre, Institut Universitaire en Santé Mentale de Québec, Quebec City, QC, Canada.,Department of Medicine, Université Laval, Quebec City, QC, Canada
| |
Collapse
|