101
|
Mitrečić D, Hribljan V, Jagečić D, Isaković J, Lamberto F, Horánszky A, Zana M, Foldes G, Zavan B, Pivoriūnas A, Martinez S, Mazzini L, Radenovic L, Milasin J, Chachques JC, Buzanska L, Song MS, Dinnyés A. Regenerative Neurology and Regenerative Cardiology: Shared Hurdles and Achievements. Int J Mol Sci 2022; 23:855. [PMID: 35055039 PMCID: PMC8776151 DOI: 10.3390/ijms23020855] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/24/2021] [Accepted: 01/09/2022] [Indexed: 02/05/2023] Open
Abstract
From the first success in cultivation of cells in vitro, it became clear that developing cell and/or tissue specific cultures would open a myriad of new opportunities for medical research. Expertise in various in vitro models has been developing over decades, so nowadays we benefit from highly specific in vitro systems imitating every organ of the human body. Moreover, obtaining sufficient number of standardized cells allows for cell transplantation approach with the goal of improving the regeneration of injured/disease affected tissue. However, different cell types bring different needs and place various types of hurdles on the path of regenerative neurology and regenerative cardiology. In this review, written by European experts gathered in Cost European action dedicated to neurology and cardiology-Bioneca, we present the experience acquired by working on two rather different organs: the brain and the heart. When taken into account that diseases of these two organs, mostly ischemic in their nature (stroke and heart infarction), bring by far the largest burden of the medical systems around Europe, it is not surprising that in vitro models of nervous and heart muscle tissue were in the focus of biomedical research in the last decades. In this review we describe and discuss hurdles which still impair further progress of regenerative neurology and cardiology and we detect those ones which are common to both fields and some, which are field-specific. With the goal to elucidate strategies which might be shared between regenerative neurology and cardiology we discuss methodological solutions which can help each of the fields to accelerate their development.
Collapse
Affiliation(s)
- Dinko Mitrečić
- Laboratory for Stem Cells, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
- Department of Histology and Embryology, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Valentina Hribljan
- Laboratory for Stem Cells, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
- Department of Histology and Embryology, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Denis Jagečić
- Laboratory for Stem Cells, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
- Department of Histology and Embryology, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | | | - Federica Lamberto
- BioTalentum Ltd., Aulich Lajos Str. 26, 2100 Gordillo, Hungary
- Department of Physiology and Animal Health, Institute of Physiology and Animal Health, Hungarian University of Agriculture and Life Sciences, Páter Károly Str. 1, 2100 Godollo, Hungary
| | - Alex Horánszky
- BioTalentum Ltd., Aulich Lajos Str. 26, 2100 Gordillo, Hungary
- Department of Physiology and Animal Health, Institute of Physiology and Animal Health, Hungarian University of Agriculture and Life Sciences, Páter Károly Str. 1, 2100 Godollo, Hungary
| | - Melinda Zana
- BioTalentum Ltd., Aulich Lajos Str. 26, 2100 Gordillo, Hungary
| | - Gabor Foldes
- Heart and Vascular Center, Semmelweis University, 1122 Budapest, Hungary
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Barbara Zavan
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Augustas Pivoriūnas
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania
| | - Salvador Martinez
- Instituto de Neurociencias UMH-CSIC, 03550 San Juan de Alicante, Spain
| | - Letizia Mazzini
- ALS Center, Department of Neurology, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy
| | - Lidija Radenovic
- Center for Laser Microscopy, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
| | - Jelena Milasin
- Laboratory for Stem Cell Research, School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Juan Carlos Chachques
- Laboratory of Biosurgical Research, Pompidou Hospital, University of Paris, 75006 Paris, France
| | - Leonora Buzanska
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Min Suk Song
- Omnion Research International Ltd., 10000 Zagreb, Croatia
| | - András Dinnyés
- BioTalentum Ltd., Aulich Lajos Str. 26, 2100 Gordillo, Hungary
- Department of Physiology and Animal Health, Institute of Physiology and Animal Health, Hungarian University of Agriculture and Life Sciences, Páter Károly Str. 1, 2100 Godollo, Hungary
- HCEMM-USZ Stem Cell Research Group, Department of Cell Biology and Molecular Medicine, University of Szeged, 6720 Szeged, Hungary
- College of Life Sciences, Sichuan University, Chengdu 610064, China
| |
Collapse
|
102
|
MicroRNA-365 regulates human cardiac action potential duration. Nat Commun 2022; 13:220. [PMID: 35017523 PMCID: PMC8752767 DOI: 10.1038/s41467-021-27856-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 12/16/2021] [Indexed: 12/21/2022] Open
Abstract
Abnormalities of ventricular action potential cause malignant cardiac arrhythmias and sudden cardiac death. Here, we aim to identify microRNAs that regulate the human cardiac action potential and ask whether their manipulation allows for therapeutic modulation of action potential abnormalities. Quantitative analysis of the microRNA targetomes in human cardiac myocytes identifies miR-365 as a primary microRNA to regulate repolarizing ion channels. Action potential recordings in patient-specific induced pluripotent stem cell-derived cardiac myocytes show that elevation of miR-365 significantly prolongs action potential duration in myocytes derived from a Short-QT syndrome patient, whereas specific inhibition of miR-365 normalizes pathologically prolonged action potential in Long-QT syndrome myocytes. Transcriptome analyses in these cells at bulk and single-cell level corroborate the key cardiac repolarizing channels as direct targets of miR-365, together with functionally synergistic regulation of additional action potential-regulating genes by this microRNA. Whole-cell patch-clamp experiments confirm miR-365-dependent regulation of repolarizing ionic current Iks. Finally, refractory period measurements in human myocardial slices substantiate the regulatory effect of miR-365 on action potential in adult human myocardial tissue. Our results delineate miR-365 to regulate human cardiac action potential duration by targeting key factors of cardiac repolarization.
Collapse
|
103
|
Bourque K, Hawey C, Jiang A, Mazarura GR, Hébert TE. Biosensor-based profiling to track cellular signalling in patient-derived models of dilated cardiomyopathy. Cell Signal 2022; 91:110239. [PMID: 34990783 DOI: 10.1016/j.cellsig.2021.110239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/06/2021] [Accepted: 12/29/2021] [Indexed: 12/18/2022]
Abstract
Dilated cardiomyopathies (DCM) represent a diverse group of cardiovascular diseases impacting the structure and function of the myocardium. To better treat these diseases, we need to understand the impact of such cardiomyopathies on critical signalling pathways that drive disease progression downstream of receptors we often target therapeutically. Our understanding of cellular signalling events has progressed substantially in the last few years, in large part due to the design, validation and use of biosensor-based approaches to studying such events in cells, tissues and in some cases, living animals. Another transformative development has been the use of human induced pluripotent stem cells (hiPSCs) to generate disease-relevant models from individual patients. We highlight the importance of going beyond monocellular cultures to incorporate the influence of paracrine signalling mediators. Finally, we discuss the recent coalition of these approaches in the context of DCM. We discuss recent work in generating patient-derived models of cardiomyopathies and the utility of using signalling biosensors to track disease progression and test potential therapeutic strategies that can be later used to inform treatment options in patients.
Collapse
Affiliation(s)
- Kyla Bourque
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Cara Hawey
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Alyson Jiang
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Grace R Mazarura
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| |
Collapse
|
104
|
Tiburcy M, Meyer T, Satin PL, Zimmermann WH. Defined Engineered Human Myocardium for Disease Modeling, Drug Screening, and Heart Repair. Methods Mol Biol 2022; 2485:213-225. [PMID: 35618908 DOI: 10.1007/978-1-0716-2261-2_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Different engineered heart muscle formats have been developed for applications in disease modeling, drug screening, and heart repair. The advantage of 3D engineered versus 2D monolayer and 3D aggregate cardiomyocyte cultures is a clearly advanced degree of maturation, which in many aspects resembles the postnatal rather than the embryonic or fetal heart, in the most advanced 3D culture formats. According to the desired in vitro (disease modeling or drug screening) and in vivo (heart repair) application, scale and geometry of tissue engineered heart muscle must be adapted. In this updated methods paper, we report a simple and scalable (up and down) collagen-based protocol for the construction of Engineered Human Myocardium (EHM) under defined, serum-free conditions.
Collapse
Affiliation(s)
- Malte Tiburcy
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Tim Meyer
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Pierre-Luc Satin
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, 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.
- Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany.
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Göttingen, Germany.
| |
Collapse
|
105
|
Barbarics B, Eildermann K, Kaderali L, Cyganek L, Plessmann U, Bodemeyer J, Paul T, Ströbel P, Urlaub H, Tirilomis T, Lenz C, Bohnenberger H. Proteomic mapping of atrial and ventricular heart tissue in patients with aortic valve stenosis. Sci Rep 2021; 11:24389. [PMID: 34937869 PMCID: PMC8695579 DOI: 10.1038/s41598-021-03907-3] [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: 08/18/2021] [Accepted: 12/06/2021] [Indexed: 11/20/2022] Open
Abstract
Aortic valve stenosis (AVS) is one of the most common valve diseases in the world. However, detailed biological understanding of the myocardial changes in AVS hearts on the proteome level is still lacking. Proteomic studies using high-resolution mass spectrometry of formalin-fixed and paraffin-embedded (FFPE) human myocardial tissue of AVS-patients are very rare due to methodical issues. To overcome these issues this study used high resolution mass spectrometry in combination with a stem cell-derived cardiac specific protein quantification-standard to profile the proteomes of 17 atrial and 29 left ventricular myocardial FFPE human myocardial tissue samples from AVS-patients. In our proteomic analysis we quantified a median of 1980 (range 1495–2281) proteins in every single sample and identified significant upregulation of 239 proteins in atrial and 54 proteins in ventricular myocardium. We compared the proteins with published data. Well studied proteins reflect disease-related changes in AVS, such as cardiac hypertrophy, development of fibrosis, impairment of mitochondria and downregulated blood supply. In summary, we provide both a workflow for quantitative proteomics of human FFPE heart tissue and a comprehensive proteomic resource for AVS induced changes in the human myocardium.
Collapse
|
106
|
Lee SJ, Kim HA, Kim SJ, Lee HA. Improving Generation of Cardiac Organoids from Human Pluripotent Stem Cells Using the Aurora Kinase Inhibitor ZM447439. Biomedicines 2021; 9:biomedicines9121952. [PMID: 34944767 PMCID: PMC8698385 DOI: 10.3390/biomedicines9121952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/16/2021] [Accepted: 12/19/2021] [Indexed: 01/21/2023] Open
Abstract
Drug-induced cardiotoxicity reduces the success rates of drug development. Thus, the limitations of current evaluation methods must be addressed. Human cardiac organoids (hCOs) derived from induced pluripotent stem cells (hiPSCs) are useful as an advanced drug-testing model; they demonstrate similar electrophysiological functionality and drug reactivity as the heart. How-ever, similar to other organoid models, they have immature characteristics compared to adult hearts, and exhibit batch-to-batch variation. As the cell cycle is important for the mesodermal differentiation of stem cells, we examined the effect of ZM447439, an aurora kinase inhibitor that regulates the cell cycle, on cardiogenic differentiation. We determined the optimal concentration and timing of ZM447439 for the differentiation of hCOs from hiPSCs and developed a novel protocol for efficiently and reproducibly generating beating hCOs with improved electrophysiological functionality, contractility, and yield. We validated their maturity through electro-physiological- and image-based functional assays and gene profiling with next-generation sequencing, and then applied these cells to multi-electrode array platforms to monitor the cardio-toxicity of drugs related to cardiac arrhythmia; the results confirmed the drug reactivity of hCOs. These findings may enable determination of the regulatory mechanism of cell cycles underlying the generation of iPSC-derived hCOs, providing a valuable drug testing platform.
Collapse
Affiliation(s)
- Su-Jin Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology (KIT), Daejeon 34114, Korea; (S.-J.L.); (H.-A.K.)
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hyeon-A Kim
- Department of Predictive Toxicology, Korea Institute of Toxicology (KIT), Daejeon 34114, Korea; (S.-J.L.); (H.-A.K.)
| | - Sung-Joon Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
- Correspondence: (S.-J.K.); (H.-A.L.); Tel.: +82-2-740-8230 (S.-J.K.); +82-42-610-8093 (H.-A.L.)
| | - Hyang-Ae Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology (KIT), Daejeon 34114, Korea; (S.-J.L.); (H.-A.K.)
- Correspondence: (S.-J.K.); (H.-A.L.); Tel.: +82-2-740-8230 (S.-J.K.); +82-42-610-8093 (H.-A.L.)
| |
Collapse
|
107
|
Human Induced Pluripotent Stem Cell as a Disease Modeling and Drug Development Platform-A Cardiac Perspective. Cells 2021; 10:cells10123483. [PMID: 34943991 PMCID: PMC8699880 DOI: 10.3390/cells10123483] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 02/07/2023] Open
Abstract
A comprehensive understanding of the pathophysiology and cellular responses to drugs in human heart disease is limited by species differences between humans and experimental animals. In addition, isolation of human cardiomyocytes (CMs) is complicated because cells obtained by biopsy do not proliferate to provide sufficient numbers of cells for preclinical studies in vitro. Interestingly, the discovery of human-induced pluripotent stem cell (hiPSC) has opened up the possibility of generating and studying heart disease in a culture dish. The combination of reprogramming and genome editing technologies to generate a broad spectrum of human heart diseases in vitro offers a great opportunity to elucidate gene function and mechanisms. However, to exploit the potential applications of hiPSC-derived-CMs for drug testing and studying adult-onset cardiac disease, a full functional characterization of maturation and metabolic traits is required. In this review, we focus on methods to reprogram somatic cells into hiPSC and the solutions for overcome immaturity of the hiPSC-derived-CMs to mimic the structure and physiological properties of the adult human CMs to accurately model disease and test drug safety. Finally, we discuss how to improve the culture, differentiation, and purification of CMs to obtain sufficient numbers of desired types of hiPSC-derived-CMs for disease modeling and drug development platform.
Collapse
|
108
|
Martínez-Falguera D, Iborra-Egea O, Gálvez-Montón C. iPSC Therapy for Myocardial Infarction in Large Animal Models: Land of Hope and Dreams. Biomedicines 2021; 9:1836. [PMID: 34944652 PMCID: PMC8698445 DOI: 10.3390/biomedicines9121836] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023] Open
Abstract
Myocardial infarction is the main driver of heart failure due to ischemia and subsequent cell death, and cell-based strategies have emerged as promising therapeutic methods to replace dead tissue in cardiovascular diseases. Research in this field has been dramatically advanced by the development of laboratory-induced pluripotent stem cells (iPSCs) that harbor the capability to become any cell type. Like other experimental strategies, stem cell therapy must meet multiple requirements before reaching the clinical trial phase, and in vivo models are indispensable for ensuring the safety of such novel therapies. Specifically, translational studies in large animal models are necessary to fully evaluate the therapeutic potential of this approach; to empirically determine the optimal combination of cell types, supplementary factors, and delivery methods to maximize efficacy; and to stringently assess safety. In the present review, we summarize the main strategies employed to generate iPSCs and differentiate them into cardiomyocytes in large animal species; the most critical differences between using small versus large animal models for cardiovascular studies; and the strategies that have been pursued regarding implanted cells' stage of differentiation, origin, and technical application.
Collapse
Affiliation(s)
- Daina Martínez-Falguera
- Faculty of Medicine, University of Barcelona (UB), 08036 Barcelona, Spain;
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Can Ruti Campus, 08916 Badalona, Spain;
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
| | - Oriol Iborra-Egea
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Can Ruti Campus, 08916 Badalona, Spain;
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
| | - Carolina Gálvez-Montón
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Can Ruti Campus, 08916 Badalona, Spain;
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| |
Collapse
|
109
|
Matsui T, Shinozawa T. Human Organoids for Predictive Toxicology Research and Drug Development. Front Genet 2021; 12:767621. [PMID: 34790228 PMCID: PMC8591288 DOI: 10.3389/fgene.2021.767621] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/11/2021] [Indexed: 12/11/2022] Open
Abstract
Organoids are three-dimensional structures fabricated in vitro from pluripotent stem cells or adult tissue stem cells via a process of self-organization that results in the formation of organ-specific cell types. Human organoids are expected to mimic complex microenvironments and many of the in vivo physiological functions of relevant tissues, thus filling the translational gap between animals and humans and increasing our understanding of the mechanisms underlying disease and developmental processes. In the last decade, organoid research has attracted increasing attention in areas such as disease modeling, drug development, regenerative medicine, toxicology research, and personalized medicine. In particular, in the field of toxicology, where there are various traditional models, human organoids are expected to blaze a new path in future research by overcoming the current limitations, such as those related to differences in drug responses among species. Here, we discuss the potential usefulness, limitations, and future prospects of human liver, heart, kidney, gut, and brain organoids from the viewpoints of predictive toxicology research and drug development, providing cutting edge information on their fabrication methods and functional characteristics.
Collapse
Affiliation(s)
- Toshikatsu Matsui
- Drug Safety Research and Evaluation, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Tadahiro Shinozawa
- Drug Safety Research and Evaluation, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| |
Collapse
|
110
|
Soepriatna AH, Kim TY, Daley MC, Song E, Choi BR, Coulombe KLK. Human Atrial Cardiac Microtissues for Chamber-Specific Arrhythmic Risk Assessment. Cell Mol Bioeng 2021; 14:441-457. [PMID: 34777603 DOI: 10.1007/s12195-021-00703-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 09/02/2021] [Indexed: 01/10/2023] Open
Abstract
Introduction Although atrial fibrillation is the most prevalent disorder of electrical conduction, the mechanisms behind atrial arrhythmias remain elusive. To address this challenge, we developed a robust in vitro model of 3D atrial microtissue from human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes and evaluated chamber-specific chemical responses experimentally and computationally. Methods We differentiated atrial and ventricular cardiomyocytes (aCMs/vCMs) from GCaMP6f-expressing hiPSCs and assessed spontaneous AP activity using fluorescence imaging. Self-assembling 3D microtissues were formed with lactate purified CMs and 5% human cardiac fibroblasts and electrically stimulated for one week before high resolution action potential (AP) optical mapping. AP responses to the atrial-specific potassium repolarizing current I Kur-blocker 4-Aminopyridine (4-AP) and funny current I f-blocker Ivabradine were characterized within their therapeutic window. Finally, we expanded upon a published hiPSC-CM computational model by incorporating the atrial-specific I Kur current, modifying ion channel conductances to match the AP waveforms of our microtissues, and employing the updated model to reinforce our experimental findings. Results High purity CMs (> 75% cTnT+) demonstrated subtype specification by MLC2v expression. Spontaneous beating rates significantly decreased following 3D microtissue formation, with atrial microtissues characterized by their faster spontaneous beating rate, slower AP rise time, and shorter AP duration (APD) compared to ventricular microtissues. We measured atrial-specific responses, including dose-dependent APD prolongation with 4-AP treatment and dose-dependent reduction in spontaneous activity post-Ivabradine treatment. Conclusion The presented in vitro platform for screening atrial-specific responses is both robust and sensitive, with high throughput, enabling studies focused at elucidating the mechanisms underlying atrial arrhythmias. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-021-00703-x.
Collapse
Affiliation(s)
- Arvin H Soepriatna
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI USA
| | - Tae Yun Kim
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI USA
| | - Mark C Daley
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI USA
| | - Elena Song
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI USA
| | - Bum-Rak Choi
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI USA
| | - Kareen L K Coulombe
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI USA
| |
Collapse
|
111
|
Wiesinger A, Boink GJJ, Christoffels VM, Devalla HD. Retinoic acid signaling in heart development: Application in the differentiation of cardiovascular lineages from human pluripotent stem cells. Stem Cell Reports 2021; 16:2589-2606. [PMID: 34653403 PMCID: PMC8581056 DOI: 10.1016/j.stemcr.2021.09.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 11/29/2022] Open
Abstract
Retinoic acid (RA) signaling plays an important role during heart development in establishing anteroposterior polarity, formation of inflow and outflow tract progenitors, and growth of the ventricular compact wall. RA is also utilized as a key ingredient in protocols designed for generating cardiac cell types from pluripotent stem cells (PSCs). This review discusses the role of RA in cardiogenesis, currently available protocols that employ RA for differentiation of various cardiovascular lineages, and plausible transcriptional mechanisms underlying this fate specification. These insights will inform further development of desired cardiac cell types from human PSCs and their application in preclinical and clinical research.
Collapse
Affiliation(s)
- Alexandra Wiesinger
- Department of Medical Biology, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Gerard J J Boink
- Department of Medical Biology, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Department of Cardiology, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Harsha D Devalla
- Department of Medical Biology, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands.
| |
Collapse
|
112
|
Odening KE, Gomez AM, Dobrev D, Fabritz L, Heinzel FR, Mangoni ME, Molina CE, Sacconi L, Smith G, Stengl M, Thomas D, Zaza A, Remme CA, Heijman J. ESC working group on cardiac cellular electrophysiology position paper: relevance, opportunities, and limitations of experimental models for cardiac electrophysiology research. Europace 2021; 23:1795-1814. [PMID: 34313298 PMCID: PMC11636574 DOI: 10.1093/europace/euab142] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/19/2021] [Indexed: 12/19/2022] Open
Abstract
Cardiac arrhythmias are a major cause of death and disability. A large number of experimental cell and animal models have been developed to study arrhythmogenic diseases. These models have provided important insights into the underlying arrhythmia mechanisms and translational options for their therapeutic management. This position paper from the ESC Working Group on Cardiac Cellular Electrophysiology provides an overview of (i) currently available in vitro, ex vivo, and in vivo electrophysiological research methodologies, (ii) the most commonly used experimental (cellular and animal) models for cardiac arrhythmias including relevant species differences, (iii) the use of human cardiac tissue, induced pluripotent stem cell (hiPSC)-derived and in silico models to study cardiac arrhythmias, and (iv) the availability, relevance, limitations, and opportunities of these cellular and animal models to recapitulate specific acquired and inherited arrhythmogenic diseases, including atrial fibrillation, heart failure, cardiomyopathy, myocarditis, sinus node, and conduction disorders and channelopathies. By promoting a better understanding of these models and their limitations, this position paper aims to improve the quality of basic research in cardiac electrophysiology, with the ultimate goal to facilitate the clinical translation and application of basic electrophysiological research findings on arrhythmia mechanisms and therapies.
Collapse
Affiliation(s)
- Katja E Odening
- Translational Cardiology, Department of Cardiology, Inselspital, Bern University Hospital, Bern, Switzerland
- Institute of Physiology, University of Bern, Bern, Switzerland
| | - Ana-Maria Gomez
- Signaling and cardiovascular pathophysiology—UMR-S 1180, Inserm, Université Paris-Saclay, 92296 Châtenay-Malabry, France
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Larissa Fabritz
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- Department of Cardiology, University Hospital Birmingham NHS Trust, Birmingham, UK
| | - Frank R Heinzel
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Matteo E Mangoni
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Cristina E Molina
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site, Hamburg/Kiel/Lübeck, Germany
| | - Leonardo Sacconi
- National Institute of Optics and European Laboratory for Non Linear Spectroscopy, Italy
- Institute for Experimental Cardiovascular Medicine, University Freiburg, Germany
| | - Godfrey Smith
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | - Milan Stengl
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Dierk Thomas
- Department of Cardiology, University Hospital Heidelberg, Heidelberg, Germany; Heidelberg Center for Heart Rhythm Disorders (HCR), University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site, Heidelberg/Mannheim, Germany
| | - Antonio Zaza
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milano, Italy
| | - Carol Ann Remme
- Department of Experimental Cardiology, Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| | - Jordi Heijman
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
113
|
Fomin A, Gärtner A, Cyganek L, Tiburcy M, Tuleta I, Wellers L, Folsche L, Hobbach AJ, von Frieling-Salewsky M, Unger A, Hucke A, Koser F, Kassner A, Sielemann K, Streckfuß-Bömeke K, Hasenfuss G, Goedel A, Laugwitz KL, Moretti A, Gummert JF, Dos Remedios CG, Reinecke H, Knöll R, van Heesch S, Hubner N, Zimmermann WH, Milting H, Linke WA. Truncated titin proteins and titin haploinsufficiency are targets for functional recovery in human cardiomyopathy due to TTN mutations. Sci Transl Med 2021; 13:eabd3079. [PMID: 34731013 DOI: 10.1126/scitranslmed.abd3079] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Andrey Fomin
- Clinic for Cardiology and Pneumology, University Medical Center, 37075 Göttingen, Germany.,German Centre for Cardiovascular Research, 10785 Berlin, partner site Göttingen, Germany
| | - Anna Gärtner
- Erich and Hanna Klessmann Institute, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
| | - Lukas Cyganek
- Clinic for Cardiology and Pneumology, University Medical Center, 37075 Göttingen, Germany.,German Centre for Cardiovascular Research, 10785 Berlin, partner site Göttingen, Germany.,Stem Cell Unit, University Medical Center, 37075 Göttingen, Germany.,Institute of Pharmacology and Toxicology, University Medical Center, 37075 Göttingen, Germany
| | - Malte Tiburcy
- German Centre for Cardiovascular Research, 10785 Berlin, partner site Göttingen, Germany.,Institute of Pharmacology and Toxicology, University Medical Center, 37075 Göttingen, Germany
| | - Izabela Tuleta
- Department of Cardiology I, Coronary, Peripheral Vascular Disease and Heart Failure, 48149 University Hospital Münster, Münster, Germany
| | - Luisa Wellers
- Institute of Physiology II, University of Münster, 48149 Münster, Germany
| | - Lina Folsche
- Institute of Physiology II, University of Münster, 48149 Münster, Germany
| | - Anastasia J Hobbach
- Department of Cardiology I, Coronary, Peripheral Vascular Disease and Heart Failure, 48149 University Hospital Münster, Münster, Germany
| | | | - Andreas Unger
- Institute of Physiology II, University of Münster, 48149 Münster, Germany
| | - Anna Hucke
- Institute of Physiology II, University of Münster, 48149 Münster, Germany
| | - Franziska Koser
- Institute of Physiology II, University of Münster, 48149 Münster, Germany
| | - Astrid Kassner
- Erich and Hanna Klessmann Institute, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
| | - Katharina Sielemann
- Erich and Hanna Klessmann Institute, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
| | - Katrin Streckfuß-Bömeke
- Clinic for Cardiology and Pneumology, University Medical Center, 37075 Göttingen, Germany.,German Centre for Cardiovascular Research, 10785 Berlin, partner site Göttingen, Germany
| | - Gerd Hasenfuss
- Clinic for Cardiology and Pneumology, University Medical Center, 37075 Göttingen, Germany.,German Centre for Cardiovascular Research, 10785 Berlin, partner site Göttingen, Germany
| | - Alexander Goedel
- First Medical Department, Cardiology, Technical University of Munich, 81675 Munich, Germany.,German Centre for Cardiovascular Research, 10785 Berlin, partner site Munich, Germany.,Department of Cell and Molecular Biology, Karolinska Institute, S-17177 Stockholm, Sweden
| | - Karl-Ludwig Laugwitz
- First Medical Department, Cardiology, Technical University of Munich, 81675 Munich, Germany.,German Centre for Cardiovascular Research, 10785 Berlin, partner site Munich, Germany.,Munich Heart Alliance, 80802 Munich, Germany
| | - Alessandra Moretti
- First Medical Department, Cardiology, Technical University of Munich, 81675 Munich, Germany.,German Centre for Cardiovascular Research, 10785 Berlin, partner site Munich, Germany.,Munich Heart Alliance, 80802 Munich, Germany
| | - Jan F Gummert
- Erich and Hanna Klessmann Institute, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany.,Department of Cardio-Thoracic Surgery, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
| | | | - Holger Reinecke
- Department of Cardiology I, Coronary, Peripheral Vascular Disease and Heart Failure, 48149 University Hospital Münster, Münster, Germany
| | - Ralph Knöll
- Department of Medicine, Integrated Cardio Metabolic Centre (ICMC), Heart and Vascular Theme, Karolinska Institute, S-17177 Stockholm, Sweden.,Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 43183 Gothenburg, Sweden
| | - Sebastiaan van Heesch
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,German Centre for Cardiovascular Research, 10785 Berlin, partner site Berlin, Germany.,Princess Máxima Center for Pediatric Oncology, 3584 CT Utrecht, Netherlands
| | - Norbert Hubner
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,German Centre for Cardiovascular Research, 10785 Berlin, partner site Berlin, Germany.,Charité-Universitätsmedizin, 10117 Berlin, Germany.,Berlin Institute of Health, 10178 Berlin, Germany
| | - Wolfram H Zimmermann
- German Centre for Cardiovascular Research, 10785 Berlin, partner site Göttingen, Germany.,Institute of Pharmacology and Toxicology, University Medical Center, 37075 Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells," University of Göttingen, 37073 Göttingen, Germany
| | - Hendrik Milting
- Erich and Hanna Klessmann Institute, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
| | - Wolfgang A Linke
- Clinic for Cardiology and Pneumology, University Medical Center, 37075 Göttingen, Germany.,German Centre for Cardiovascular Research, 10785 Berlin, partner site Göttingen, Germany.,Institute of Physiology II, University of Münster, 48149 Münster, Germany
| |
Collapse
|
114
|
Turhan AG, Hwang JW, Chaker D, Tasteyre A, Latsis T, Griscelli F, Desterke C, Bennaceur-Griscelli A. iPSC-Derived Organoids as Therapeutic Models in Regenerative Medicine and Oncology. Front Med (Lausanne) 2021; 8:728543. [PMID: 34722569 PMCID: PMC8548367 DOI: 10.3389/fmed.2021.728543] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/17/2021] [Indexed: 01/22/2023] Open
Abstract
Progress made during the last decade in stem cell biology allows currently an unprecedented potential to translate these advances into the clinical applications and to shape the future of regenerative medicine. Organoid technology is amongst these major developments, derived from primary tissues or more recently, from induced pluripotent stem cells (iPSC). The use of iPSC technology offers the possibility of cancer modeling especially in hereditary cancers with germline oncogenic mutations. Similarly, it has the advantage to be amenable to genome editing with introduction of specific oncogenic alterations using CRISPR-mediated gene editing. In the field of regenerative medicine, iPSC-derived organoids hold promise for the generation of future advanced therapeutic medicinal products (ATMP) for organ repair. Finally, it appears that they can be of highly useful experimental tools to determine cell targets of SARS-Cov-2 infections allowing to test anti-Covid drugs. Thus, with the possibilities of genomic editing and the development of new protocols for differentiation toward functional tissues, it is expected that iPSC-derived organoid technology will represent also a therapeutic tool in all areas of medicine.
Collapse
Affiliation(s)
- Ali G Turhan
- INSERM UA/09 UMR-S 935, Université Paris Saclay, Villejuif, France.,ESTeam Paris Sud, Université Paris Saclay, Villejuif, France.,APHP Paris Saclay, Department of Hematology, Hopital Bicetre and Paul Brousse, Villejuif, France.,INGESTEM National iPSC Infrastructure, Villejuif, France.,CITHERA, Centre for IPSC Therapies, INSERM UMS-45, Genopole, Evry, France
| | - Jinwook W Hwang
- INSERM UA/09 UMR-S 935, Université Paris Saclay, Villejuif, France.,ESTeam Paris Sud, Université Paris Saclay, Villejuif, France
| | - Diana Chaker
- INSERM UA/09 UMR-S 935, Université Paris Saclay, Villejuif, France.,ESTeam Paris Sud, Université Paris Saclay, Villejuif, France.,INGESTEM National iPSC Infrastructure, Villejuif, France.,CITHERA, Centre for IPSC Therapies, INSERM UMS-45, Genopole, Evry, France
| | - Albert Tasteyre
- INSERM UA/09 UMR-S 935, Université Paris Saclay, Villejuif, France.,ESTeam Paris Sud, Université Paris Saclay, Villejuif, France.,INGESTEM National iPSC Infrastructure, Villejuif, France.,CITHERA, Centre for IPSC Therapies, INSERM UMS-45, Genopole, Evry, France
| | - Theodoros Latsis
- INSERM UA/09 UMR-S 935, Université Paris Saclay, Villejuif, France.,ESTeam Paris Sud, Université Paris Saclay, Villejuif, France.,INGESTEM National iPSC Infrastructure, Villejuif, France
| | - Frank Griscelli
- INSERM UA/09 UMR-S 935, Université Paris Saclay, Villejuif, France.,ESTeam Paris Sud, Université Paris Saclay, Villejuif, France.,INGESTEM National iPSC Infrastructure, Villejuif, France.,CITHERA, Centre for IPSC Therapies, INSERM UMS-45, Genopole, Evry, France.,Université Paris Descartes, Faculté Sorbonne Paris Cité, Faculté des Sciences Pharmaceutiques et Biologiques, Paris, France
| | - Christophe Desterke
- INSERM UA/09 UMR-S 935, Université Paris Saclay, Villejuif, France.,ESTeam Paris Sud, Université Paris Saclay, Villejuif, France.,INGESTEM National iPSC Infrastructure, Villejuif, France
| | - Annelise Bennaceur-Griscelli
- INSERM UA/09 UMR-S 935, Université Paris Saclay, Villejuif, France.,ESTeam Paris Sud, Université Paris Saclay, Villejuif, France.,APHP Paris Saclay, Department of Hematology, Hopital Bicetre and Paul Brousse, Villejuif, France.,INGESTEM National iPSC Infrastructure, Villejuif, France.,CITHERA, Centre for IPSC Therapies, INSERM UMS-45, Genopole, Evry, France
| |
Collapse
|
115
|
Blebbistatin protects iPSC-CMs from hypercontraction and facilitates automated patch-clamp based electrophysiological study. Stem Cell Res 2021; 56:102565. [PMID: 34638057 DOI: 10.1016/j.scr.2021.102565] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/01/2021] [Accepted: 10/06/2021] [Indexed: 01/11/2023] Open
Abstract
Recently, there have been great advances in cardiovascular channelopathy modeling and drug safety pharmacology using human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). The automated patch-clamp (APC) technique overcomes the disadvantages of the manual patch-clamp (MPC) technique, which is labor intensive and gives low output. However, the application of the APC platform is still limited in iPSC-CM based research, due to the difficulty in maintaining the high quality of single iPSC-CMs during dissociation and recording. In this study, we improved the method for single iPSC-CM preparation by applying 2.5 µM blebbistatin (BB, an excitation-contraction coupling uncoupler) throughout APC procedures (dissociation, filtration, storage, and recording). Under non-BB buffered condition, iPSC-CMs in suspension showed a severe bleb-like morphology. However, BB-supplement led to significant improvements in morphology and INa recording, and we even obtained several CMs that showed spontaneous action potentials with typical morphology. Furthermore, APC faithfully recapitulated the single-cell electrophysiological phenotypes of iPSC-CMs derived from Brugada syndrome patients, as detected with MPC. Our study indicates that APC is capable of replacing MPC in the modeling of cardiac channelopathies using human iPSC-CMs by providing high-quality data with higher throughput.
Collapse
|
116
|
Approaches to Optimize Stem Cell-Derived Cardiomyocyte Maturation and Function. CURRENT STEM CELL REPORTS 2021. [DOI: 10.1007/s40778-021-00197-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
117
|
Stein JM, Mummery CL, Bellin M. Engineered models of the human heart: Directions and challenges. Stem Cell Reports 2021; 16:2049-2057. [PMID: 33338434 PMCID: PMC8452488 DOI: 10.1016/j.stemcr.2020.11.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 02/07/2023] Open
Abstract
Human heart (patho)physiology is now widely studied using human pluripotent stem cells, but the immaturity of derivative cardiomyocytes has largely limited disease modeling to conditions associated with mutations in cardiac ion channel genes. Recent advances in tissue engineering and organoids have, however, created new opportunities to study diseases beyond "channelopathies." These synthetic cardiac structures allow quantitative measurement of contraction, force, and other biophysical parameters in three-dimensional configurations, in which the cardiomyocytes in addition become more mature. Multiple cardiac-relevant cell types are also often combined to form organized cardiac tissue mimetic constructs, where cell-cell, cell-extracellular matrix, and paracrine interactions can be mimicked. In this review, we provide an overview of some of the most promising technologies being implemented specifically in personalized heart-on-a-chip models and explore their applications, drawbacks, and potential for future development.
Collapse
Affiliation(s)
- Jeroen M Stein
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333ZA, the Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333ZA, the Netherlands; Department of Applied Stem Cell Technologies, University of Twente, Enschede 7500AE, the Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333ZA, the Netherlands; Department of Biology, University of Padua, Padua 35131, Italy; Veneto Institute of Molecular Medicine, Padua 35129, Italy.
| |
Collapse
|
118
|
Yigit G, Wollnik B. Cellular models and therapeutic perspectives in hypertrophic cardiomyopathy. MED GENET-BERLIN 2021; 33:235-243. [PMID: 38835701 PMCID: PMC11006313 DOI: 10.1515/medgen-2021-2094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/28/2021] [Indexed: 06/06/2024]
Abstract
Hypertrophic cardiomyopathy (HCM) is a clinically heterogeneous cardiac disease that is mainly characterized by left ventricular hypertrophy in the absence of any additional cardiac or systemic disease. HCM is genetically heterogeneous, inherited mainly in an autosomal dominant pattern, and so far pathogenic variants have been identified in more than 20 genes, mostly encoding proteins of the cardiac sarcomere. Based on its variable penetrance and expressivity, pathogenicity of newly identified variants often remains unsolved, underlining the importance of cellular and tissue-based models that help to uncover causative genetic alterations and, additionally, provide appropriate systems for the analysis of disease hallmarks as well as for the design and application of new therapeutic strategies like drug screenings and genome/base editing approaches. Here, we review the current state of cellular and tissue-engineered models and provide future perspectives for personalized therapeutic strategies of HCM.
Collapse
Affiliation(s)
- Gökhan Yigit
- Institute of Human Genetics, University Medical Center Göttingen, Heinrich-Düker-Weg 12, 37073 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| | - Bernd Wollnik
- Institute of Human Genetics, University Medical Center Göttingen, Heinrich-Düker-Weg 12, 37073 Göttingen, Germany
- DZHK (German Centre 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
|
119
|
Kamga MVK, Reppel M, Hescheler J, Nguemo F. Modeling genetic cardiac channelopathies using induced pluripotent stem cells - Status quo from an electrophysiological perspective. Biochem Pharmacol 2021; 192:114746. [PMID: 34461117 DOI: 10.1016/j.bcp.2021.114746] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/24/2021] [Accepted: 08/24/2021] [Indexed: 12/15/2022]
Abstract
Long QT syndrome (LQTS), Brugada syndrome (BrS), and catecholaminergic polymorphic ventricular tachycardia (CPVT) are genetic diseases of the heart caused by mutations in specific cardiac ion channels and are characterized by paroxysmal arrhythmias, which can deteriorate into ventricular fibrillation. In LQTS3 and BrS different mutations in the SCN5A gene lead to a gain-or a loss-of-function of the voltage-gated sodium channel Nav1.5, respectively. Although sharing the same gene mutation, these syndromes are characterized by different clinical manifestations and functional perturbations and in some cases even present an overlapping clinical phenotype. Several studies have shown that Na+ current abnormalities in LQTS3 and BrS can also cause Ca2+-signaling aberrancies in cardiomyocytes (CMs). Abnormal Ca2+ homeostasis is also the main feature of CPVT which is mostly caused by heterozygous mutations in the RyR2 gene. Large numbers of disease-causing mutations were identified in RyR2 and SCN5A but it is not clear how different variants in the SCN5A gene produce different clinical syndromes and if in CPVT Ca2+ abnormalities and drug sensitivities vary depending on the mutation site in the RyR2. These questions can now be addressed by using patient-specific in vitro models of these diseases based on induced pluripotent stem cells (iPSCs). In this review, we summarize different insights gained from these models with a focus on electrophysiological perturbations caused by different ion channel mutations and discuss how will this knowledge help develop better stratification and more efficient personalized therapies for these patients.
Collapse
Affiliation(s)
- Michelle Vanessa Kapchoup Kamga
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Michael Reppel
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, 50931 Cologne, Germany; Praxis für Kardiologie und Angiologie, Landsberg am Lech, Germany
| | - Jürgen Hescheler
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Filomain Nguemo
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
120
|
Isolation of Cardiomyocytes Derived from Human Pluripotent Stem Cells Using miRNA Switches. Methods Mol Biol 2021. [PMID: 34302646 DOI: 10.1007/978-1-0716-1484-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The most common method for isolating cells of interest is an antibody method that recognizes cell surface antigens. However, specific surface antigens for many cell types have not been identified. We have developed the microRNA (miRNA)-responsive synthetic mRNA systems (miRNA switches), which isolate target cells based on endogenous miRNA activity. In this chapter, we describe protocols for isolating human pluripotent stem cell (hPSC)-derived cardiomyocytes using miRNA switches with or without cell sorting.
Collapse
|
121
|
Andrysiak K, Stępniewski J, Dulak J. Human-induced pluripotent stem cell-derived cardiomyocytes, 3D cardiac structures, and heart-on-a-chip as tools for drug research. Pflugers Arch 2021; 473:1061-1085. [PMID: 33629131 PMCID: PMC8245367 DOI: 10.1007/s00424-021-02536-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/01/2021] [Accepted: 02/03/2021] [Indexed: 12/13/2022]
Abstract
Development of new drugs is of high interest for the field of cardiac and cardiovascular diseases, which are a dominant cause of death worldwide. Before being allowed to be used and distributed, every new potentially therapeutic compound must be strictly validated during preclinical and clinical trials. The preclinical studies usually involve the in vitro and in vivo evaluation. Due to the increasing reporting of discrepancy in drug effects in animal and humans and the requirement to reduce the number of animals used in research, improvement of in vitro models based on human cells is indispensable. Primary cardiac cells are difficult to access and maintain in cell culture for extensive experiments; therefore, the human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) became an excellent alternative. This technology enables a production of high number of patient- and disease-specific cardiomyocytes and other cardiac cell types for a large-scale research. The drug effects can be extensively evaluated in the context of electrophysiological responses with a use of well-established tools, such as multielectrode array (MEA), patch clamp, or calcium ion oscillation measurements. Cardiotoxicity, which is a common reason for withdrawing drugs from marketing or rejection at final stages of clinical trials, can be easily verified with a use of hiPSC-CM model providing a prediction of human-specific responses and higher safety of clinical trials involving patient cohort. Abovementioned studies can be performed using two-dimensional cell culture providing a high-throughput and relatively lower costs. On the other hand, more complex structures, such as engineered heart tissue, organoids, or spheroids, frequently applied as co-culture systems, represent more physiological conditions and higher maturation rate of hiPSC-derived cells. Furthermore, heart-on-a-chip technology has recently become an increasingly popular tool, as it implements controllable culture conditions, application of various stimulations and continuous parameters read-out. This paper is an overview of possible use of cardiomyocytes and other cardiac cell types derived from hiPSC as in vitro models of heart in drug research area prepared on the basis of latest scientific reports and providing thorough discussion regarding their advantages and limitations.
Collapse
Affiliation(s)
- Kalina Andrysiak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Jacek Stępniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
122
|
From Stem Cells to Populations-Using hiPSC, Next-Generation Sequencing, and GWAS to Explore the Genetic and Molecular Mechanisms of Congenital Heart Defects. Genes (Basel) 2021; 12:genes12060921. [PMID: 34208537 PMCID: PMC8235101 DOI: 10.3390/genes12060921] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/01/2021] [Accepted: 06/12/2021] [Indexed: 01/16/2023] Open
Abstract
Congenital heart defects (CHD) are developmental malformations affecting the heart and the great vessels. Early heart development requires temporally regulated crosstalk between multiple cell types, signaling pathways, and mechanical forces of early blood flow. While both genetic and environmental factors have been recognized to be involved, identifying causal genes in non-syndromic CHD has been difficult. While variants following Mendelian inheritance have been identified by linkage analysis in a few families with multiple affected members, the inheritance pattern in most familial cases is complex, with reduced penetrance and variable expressivity. Furthermore, most non-syndromic CHD are sporadic. Improved sequencing technologies and large biobank collections have enabled genome-wide association studies (GWAS) in non-syndromic CHD. The ability to generate human to create human induced pluripotent stem cells (hiPSC) and further differentiate them to organotypic cells enables further exploration of genotype–phenotype correlations in patient-derived cells. Here we review how these technologies can be used in unraveling the genetics and molecular mechanisms of heart development.
Collapse
|
123
|
Leopold JA, Kawut SM, Aldred MA, Archer SL, Benza RL, Bristow MR, Brittain EL, Chesler N, DeMan FS, Erzurum SC, Gladwin MT, Hassoun PM, Hemnes AR, Lahm T, Lima JA, Loscalzo J, Maron BA, Rosa LM, Newman JH, Redline S, Rich S, Rischard F, Sugeng L, Tang WHW, Tedford RJ, Tsai EJ, Ventetuolo CE, Zhou Y, Aggarwal NR, Xiao L. Diagnosis and Treatment of Right Heart Failure in Pulmonary Vascular Diseases: A National Heart, Lung, and Blood Institute Workshop. Circ Heart Fail 2021; 14:e007975. [PMID: 34422205 PMCID: PMC8375628 DOI: 10.1161/circheartfailure.120.007975] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Right ventricular dysfunction is a hallmark of advanced pulmonary vascular, lung parenchymal, and left heart disease, yet the underlying mechanisms that govern (mal)adaptation remain incompletely characterized. Owing to the knowledge gaps in our understanding of the right ventricle (RV) in health and disease, the National Heart, Lung, and Blood Institute (NHLBI) commissioned a working group to identify current challenges in the field. These included a need to define and standardize normal RV structure and function in populations; access to RV tissue for research purposes and the development of complex experimental platforms that recapitulate the in vivo environment; and the advancement of imaging and invasive methodologies to study the RV within basic, translational, and clinical research programs. Specific recommendations were provided, including a call to incorporate precision medicine and innovations in prognosis, diagnosis, and novel RV therapeutics for patients with pulmonary vascular disease.
Collapse
Affiliation(s)
- Jane A. Leopold
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Steven M. Kawut
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Micheala A. Aldred
- Division of Pulmonary, Critical Care, Sleep & Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, IN
| | - Stephen L. Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Ray L. Benza
- Department of Medicine, Allegheny General Hospital, Pittsburgh, PA
| | | | - Evan L. Brittain
- Division of Cardiovascular Medicine and Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University Medical Center, Nashville, TN
| | - Naomi Chesler
- Department of Biomedical Engineering, University of Wisconsin-Madison College of Engineering, Madison, WI
| | - Frances S. DeMan
- Department of Pulmonary Medicine, PHEniX laboratory, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | - Mark T. Gladwin
- Department of Medicine, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, UPMC and the University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Paul M. Hassoun
- Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Anna R. Hemnes
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Tim Lahm
- Division of Pulmonary, Critical Care, Sleep & Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, IN
| | - Joao A.C. Lima
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Bradley A. Maron
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital and Harvard Medical School and Department of Cardiology, Boston VA Healthcare System, West Roxbury, MA
| | - Laura Mercer Rosa
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - John H. Newman
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Susan Redline
- Departments of Medicine and Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Stuart Rich
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Franz Rischard
- Department of Medicine, University of Arizona- Tucson, Tucson, AZ
| | - Lissa Sugeng
- Department of Medicine, Yale School of Medicine, New Haven, CT
| | - W. H. Wilson Tang
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH
| | - Ryan J. Tedford
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC
| | - Emily J. Tsai
- Division of Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY
| | - Corey E. Ventetuolo
- Department of Medicine, Alpert Medical School of Brown University, Department of Health Services, Policy and Practice, Brown University School of Public Health, Providence, RI
| | - YouYang Zhou
- Departments of Pediatrics (Division of Critical Care), Pharmacology, and Medicine, Northwestern University Feinberg School of Medicine. Chicago, Illinois
| | - Neil R. Aggarwal
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD
| | - Lei Xiao
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD
| |
Collapse
|
124
|
Rafatian N, Vizely K, Al Asafen H, Korolj A, Radisic M. Drawing Inspiration from Developmental Biology for Cardiac Tissue Engineers. Adv Biol (Weinh) 2021; 5:e2000190. [PMID: 34008910 DOI: 10.1002/adbi.202000190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 12/21/2020] [Indexed: 12/17/2022]
Abstract
A sound understanding of developmental biology is part of the foundation of effective stem cell-derived tissue engineering. Here, the key concepts of cardiac development that are successfully applied in a bioinspired approach to growing engineered cardiac tissues, are reviewed. The native cardiac milieu is studied extensively from embryonic to adult phenotypes, as it provides a resource of factors, mechanisms, and protocols to consider when working toward establishing living tissues in vitro. It begins with the various cell types that constitute the cardiac tissue. It is discussed how myocytes interact with other cell types and their microenvironment and how they change over time from the embryonic to the adult states, with a view on how such changes affect the tissue function and may be used in engineered tissue models. Key embryonic signaling pathways that have been leveraged in the design of culture media and differentiation protocols are presented. The cellular microenvironment, from extracellular matrix chemical and physical properties, to the dynamic mechanical and electrical forces that are exerted on tissues is explored. It is shown that how such microenvironmental factors can inform the design of biomaterials, scaffolds, stimulation bioreactors, and maturation readouts, and suggest considerations for ongoing biomimetic advancement of engineered cardiac tissues and regeneration strategies for the future.
Collapse
Affiliation(s)
- Naimeh Rafatian
- Toronto General Research Institute, Toronto, Ontario, M5G 2C4, Canada
| | - Katrina Vizely
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada
| | - Hadel Al Asafen
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada
| | - Anastasia Korolj
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada.,Institute of Biomaterials Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
| | - Milica Radisic
- Toronto General Research Institute, Toronto, Ontario, M5G 2C4, Canada.,Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada.,Institute of Biomaterials Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
| |
Collapse
|
125
|
Esser T, Trossmann V, Lentz S, Engel F, Scheibel T. Designing of spider silk proteins for human induced pluripotent stem cell-based cardiac tissue engineering. Mater Today Bio 2021; 11:100114. [PMID: 34169268 PMCID: PMC8209670 DOI: 10.1016/j.mtbio.2021.100114] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/01/2021] [Accepted: 05/08/2021] [Indexed: 12/25/2022] Open
Abstract
Materials made of recombinant spider silk proteins are promising candidates for cardiac tissue engineering, and their suitability has so far been investigated utilizing primary rat cardiomyocytes. Herein, we expanded the tool box of available spider silk variants and demonstrated for the first time that human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes attach, contract, and respond to pharmacological treatment using phenylephrine and verapamil on explicit spider silk films. The hiPSC-cardiomyocytes contracted for at least 14 days on films made of positively charged engineered Araneus diadematus fibroin 4 (eADF4(κ16)) and three different arginyl-glycyl-aspartic acid (RGD)-tagged spider silk variants (positively or negatively charged and uncharged). Notably, hiPSC-cardiomyocytes exhibited different morphologies depending on the spider silk variant used, with less spreading and being smaller on films made of eADF4(κ16) than on RGD-tagged spider silk films. These results indicate that spider silk engineering is a powerful tool to provide new materials suitable for hiPSC-based cardiac tissue engineering. hiPSC-cardiomyocytes attach and contract on positively charged and/or RGD-tagged spider silk variants. hiPSC-cardiomyocytes exhibit spider silk variant-dependent morphology upon adhesion. Explicit spider silk variants promote long-term contractility of hiPSC-cardiomyocytes. hiPSC-cardiomyocytes grown on spider silk materials respond to pharmacological treatment.
Collapse
Key Words
- AFM, atomic force microscopy
- APTES, (3-aminopropyl) triethoxysilane
- ATR, attenuated total reflection
- DPBS, Dulbecco's phosphate-buffered saline
- EthHD1, ethidium homodimer 1
- FT-IR, Fourier-transform infrared (spectroscopy)
- IPTG, isopropyl-β-D-thiogalactoside
- MALDI-TOF, matrix-assisted laser desorption/ionization time-of-flight
- SDS-PAGE, sodium dodecyl sulfate-polyacrylamide gel electrophoresis
- eADF4, Engineered Araneus diadematus fibroin 4
- hiPSC, human-induced pluripotent stem cell
Collapse
Affiliation(s)
- T.U. Esser
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, 91054, Germany
| | - V.T. Trossmann
- Lehrstuhl Biomaterialien, Prof.-Rüdiger-Bormann Straße 1, Bayreuth, 95447, Germany
| | - S. Lentz
- Lehrstuhl Biomaterialien, Prof.-Rüdiger-Bormann Straße 1, Bayreuth, 95447, Germany
| | - F.B. Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, 91054, Germany
- MURCE, Muscle Research Center Erlangen, Erlangen, Germany
- Corresponding author. Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, 91054, Germany.
| | - T. Scheibel
- Lehrstuhl Biomaterialien, Prof.-Rüdiger-Bormann Straße 1, Bayreuth, 95447, Germany
- Bayreuther Zentrum für Kolloide und Grenzflächen (BZKG), Bayerisches Polymerinstitut (BPI), Bayreuther Zentrum für Molekulare Biowissenschaften (BZMB), Bayreuther Materialzentrum (BayMAT), Universitätsstraße 30, Universität Bayreuth, Bayreuth, D-95447, Germany
- Corresponding author. Lehrstuhl Biomaterialien, Prof.-Rüdiger-Bormann Straße 1, Bayreuth, 95447, Germany.
| |
Collapse
|
126
|
Huang M, Liao Z, Li X, Yang Z, Fan X, Li Y, Zhao Z, Lang S, Cyganek L, Zhou X, Akin I, Borggrefe M, El-Battrawy I. Effects of Antiarrhythmic Drugs on hERG Gating in Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes From a Patient With Short QT Syndrome Type 1. Front Pharmacol 2021; 12:675003. [PMID: 34025432 PMCID: PMC8138577 DOI: 10.3389/fphar.2021.675003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/26/2021] [Indexed: 12/17/2022] Open
Abstract
Aims: The short QT syndrome type 1 (SQT1) is linked to hERG channel mutations (e.g., N588K). Drug effects on hERG channel gating kinetics in SQT1-cells have not been investigated. Methods: This study used hiPSC-CMs of a healthy donor and a SQT1-patient carrying the N588K mutation and patch clamp to examine the drug effects on hERG channel gating kinetics. Results: Ajmaline, amiodarone, ivabradine, flecainide, quinidine, mexiletine and ranolazine inhibited the hERG channel current (IKr) less strongly in hiPSC-CMs from the SQTS1-patient (SQT1-hiPSC-CMs) comparing with cells from the healthy donor (donor-hiPSC-CMs). Quinidine and mexiletine reduced, but ajmaline, amiodarone, ivabradine and ranolazine increased the time to peak of IKr similarly in SQT1-hiPSC-CMs and donor-hiPSC-CMs. Although regarding the shift of activation and inactivation curves, tested drugs showed differential effects in donor- and SQT1-hiPSC-CMs, quinidine, ajmaline, ivabradine and mexiletine but not amiodarone, flecainide and ranolazine reduced the window current in SQT1-hiPSC-CMs. Quinidine, ajmaline, ivabradine and mexiletine differentially changed the time constant of recovery from inactivation, but all of them increased the time constant of deactivation in SQT1-hiPSC-CMs. Conclusion: The window current-reducing and deactivation-slowing effects may be important for the antiarrhythmic effect of ajmaline, ivabradine, quinidine and mexiletine in SQT1-cells. This information may be helpful for selecting drugs for treating SQT1-patients with hERG channel mutation.
Collapse
Affiliation(s)
- Mengying Huang
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Zhenxing Liao
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,North Sichuan Medical College, Nanchong, China
| | - Xin Li
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhen Yang
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,North Sichuan Medical College, Nanchong, China
| | - Xuehui Fan
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Yingrui Li
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Zhihan Zhao
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Siegfried Lang
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim and Göttingen, Mannheim, Germany
| | - Lukas Cyganek
- DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim and Göttingen, Mannheim, Germany.,Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Xiaobo Zhou
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim and Göttingen, Mannheim, Germany
| | - Ibrahim Akin
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim and Göttingen, Mannheim, Germany
| | - Martin Borggrefe
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim and Göttingen, Mannheim, Germany
| | - Ibrahim El-Battrawy
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim and Göttingen, Mannheim, Germany
| |
Collapse
|
127
|
Cell surface markers for immunophenotyping human pluripotent stem cell-derived cardiomyocytes. Pflugers Arch 2021; 473:1023-1039. [PMID: 33928456 DOI: 10.1007/s00424-021-02549-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 02/09/2021] [Accepted: 02/25/2021] [Indexed: 02/08/2023]
Abstract
Human pluripotent stem cells (hPSC) self-renew and represent a potentially unlimited source for the production of cardiomyocytes (CMs) suitable for studies of human cardiac development, drug discovery, cardiotoxicity testing, and disease modelling and for cell-based therapies. However, most cardiac differentiation protocols yield mixed cultures of atrial-, ventricular-, and pacemaker-like cells at various stages of development, as well as non-CMs. The proportions and maturation states of these cell types result from disparities among differentiation protocols and time of cultivation, as well as hPSC reprogramming inconsistencies and genetic background variations. The reproducible use of hPSC-CMs for research and therapy is therefore limited by issues of cell population heterogeneity and functional states of maturation. A validated method that overcomes issues of cell heterogeneity is immunophenotyping coupled with live cell sorting, an approach that relies on accessible surface markers restricted to the desired cell type(s). Here we review current progress in unravelling heterogeneity in hPSC-cardiac cultures and in the identification of surface markers suitable for defining cardiac identity, subtype specificity, and maturation states.
Collapse
|
128
|
Yousefi R, Fornasiero EF, Cyganek L, Montoya J, Jakobs S, Rizzoli SO, Rehling P, Pacheu‐Grau D. Monitoring mitochondrial translation in living cells. EMBO Rep 2021; 22:e51635. [PMID: 33586863 PMCID: PMC8024989 DOI: 10.15252/embr.202051635] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/05/2021] [Accepted: 01/13/2021] [Indexed: 12/20/2022] Open
Abstract
Mitochondria possess a small genome that codes for core subunits of the oxidative phosphorylation system and whose expression is essential for energy production. Information on the regulation and spatial organization of mitochondrial gene expression in the cellular context has been difficult to obtain. Here we devise an imaging approach to analyze mitochondrial translation within the context of single cells, by following the incorporation of clickable non-canonical amino acids. We apply this method to multiple cell types, including specialized cells such as cardiomyocytes and neurons, and monitor with spatial resolution mitochondrial translation in axons and dendrites. We also show that translation imaging allows to monitor mitochondrial protein expression in patient fibroblasts. Approaching mitochondrial translation with click chemistry opens new avenues to understand how mitochondrial biogenesis is integrated into the cellular context and can be used to assess mitochondrial gene expression in mitochondrial diseases.
Collapse
Affiliation(s)
- Roya Yousefi
- Department of Cellular BiochemistryUniversity Medical Center GöttingenGöttingenGermany
| | - Eugenio F Fornasiero
- Department of Neuro‐ and Sensory PhysiologyUniversity Medical Center GöttingenGöttingenGermany
| | - Lukas Cyganek
- Clinic for Cardiology and PneumologyUniversity Medical Center GöttingenGöttingenGermany
- German Center for Cardiovascular Research (DZHK)GöttingenGermany
| | - Julio Montoya
- Departamento de BioquímicaBiología Molecular y CelularUniversidad de ZaragozaZaragozaSpain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER)MadridSpain
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón)Universidad de ZaragozaZaragozaSpain
| | - Stefan Jakobs
- Department of NanoBiophotonicsMax Planck Institute for Biophysical ChemistryGöttingenGermany
- Clinic of NeurologyUniversity Medical Center GöttingenGöttingenGermany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC)University of GoettingenGöttingenGermany
| | - Silvio O Rizzoli
- Department of Neuro‐ and Sensory PhysiologyUniversity Medical Center GöttingenGöttingenGermany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC)University of GoettingenGöttingenGermany
| | - Peter Rehling
- Department of Cellular BiochemistryUniversity Medical Center GöttingenGöttingenGermany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC)University of GoettingenGöttingenGermany
- Max‐Planck Institute for Biophysical ChemistryGöttingenGermany
| | - David Pacheu‐Grau
- Department of Cellular BiochemistryUniversity Medical Center GöttingenGöttingenGermany
| |
Collapse
|
129
|
Lyra-Leite DM, Fonoudi H, Gharib M, Burridge PW. An updated protocol for the cost-effective and weekend-free culture of human induced pluripotent stem cells. STAR Protoc 2021; 2:100213. [PMID: 33786455 PMCID: PMC7988236 DOI: 10.1016/j.xpro.2020.100213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The protocol provided here describes methodologies for making a highly cost-effective, chemically defined medium for culturing hiPSCs we call B8 medium. The typical cost of B8 medium is US$10 per liter, which with modifications included here is more affordable than standard media. We provide simple protocols for making B8 supplement aliquots, making the basal media DMEM/F12, Matrigel-coated plates, thawing, passaging, culturing, and cryopreserving hiPSCs. We show typical differentiation results and provide a comprehensive troubleshooting guide. For complete details on the use and execution of this protocol, please refer to Kuo et al. (2020).
Collapse
Affiliation(s)
- Davi Marco Lyra-Leite
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Hananeh Fonoudi
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Mennat Gharib
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Paul W Burridge
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
130
|
Hnatiuk AP, Briganti F, Staudt DW, Mercola M. Human iPSC modeling of heart disease for drug development. Cell Chem Biol 2021; 28:271-282. [PMID: 33740432 PMCID: PMC8054828 DOI: 10.1016/j.chembiol.2021.02.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/26/2021] [Accepted: 02/19/2021] [Indexed: 02/08/2023]
Abstract
Human induced pluripotent stem cells (hiPSCs) have emerged as a promising platform for pharmacogenomics and drug development. In cardiology, they make it possible to produce unlimited numbers of patient-specific human cells that reproduce hallmark features of heart disease in the culture dish. Their potential applications include the discovery of mechanism-specific therapeutics, the evaluation of safety and efficacy in a human context before a drug candidate reaches patients, and the stratification of patients for clinical trials. Although this new technology has the potential to revolutionize drug discovery, translational hurdles have hindered its widespread adoption for pharmaceutical development. Here we discuss recent progress in overcoming these hurdles that should facilitate the use of hiPSCs to develop new medicines and individualize therapies for heart disease.
Collapse
Affiliation(s)
- Anna P Hnatiuk
- Stanford Cardiovascular Institute, 240 Pasteur Drive, Biomedical Innovation Building, Palo Alto, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Francesca Briganti
- Stanford Cardiovascular Institute, 240 Pasteur Drive, Biomedical Innovation Building, Palo Alto, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - David W Staudt
- Stanford Cardiovascular Institute, 240 Pasteur Drive, Biomedical Innovation Building, Palo Alto, CA 94305, USA; Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Mark Mercola
- Stanford Cardiovascular Institute, 240 Pasteur Drive, Biomedical Innovation Building, Palo Alto, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
131
|
James EC, Tomaskovic-Crook E, Crook JM. Bioengineering Clinically Relevant Cardiomyocytes and Cardiac Tissues from Pluripotent Stem Cells. Int J Mol Sci 2021; 22:ijms22063005. [PMID: 33809429 PMCID: PMC8001925 DOI: 10.3390/ijms22063005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/23/2022] Open
Abstract
The regenerative capacity of cardiomyocytes is insufficient to functionally recover damaged tissue, and as such, ischaemic heart disease forms the largest proportion of cardiovascular associated deaths. Human-induced pluripotent stem cells (hiPSCs) have enormous potential for developing patient specific cardiomyocytes for modelling heart disease, patient-based cardiac toxicity testing and potentially replacement therapy. However, traditional protocols for hiPSC-derived cardiomyocytes yield mixed populations of atrial, ventricular and nodal-like cells with immature cardiac properties. New insights gleaned from embryonic heart development have progressed the precise production of subtype-specific hiPSC-derived cardiomyocytes; however, their physiological immaturity severely limits their utility as model systems and their use for drug screening and cell therapy. The long-entrenched challenges in this field are being addressed by innovative bioengingeering technologies that incorporate biophysical, biochemical and more recently biomimetic electrical cues, with the latter having the potential to be used to both direct hiPSC differentiation and augment maturation and the function of derived cardiomyocytes and cardiac tissues by mimicking endogenous electric fields.
Collapse
Affiliation(s)
- Emma Claire James
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Wollongong 2500, Australia;
| | - Eva Tomaskovic-Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Wollongong 2500, Australia;
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong 2500, Australia
- Correspondence: (E.T.-C.); (J.M.C.)
| | - Jeremy Micah Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Wollongong 2500, Australia;
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong 2500, Australia
- Department of Surgery, St Vincent’s Hospital, The University of Melbourne, Fitzroy 3065, Australia
- Correspondence: (E.T.-C.); (J.M.C.)
| |
Collapse
|
132
|
Abstract
PURPOSE OF REVIEW Heart failure is among the most prevalent disease complexes overall and is associated with high morbidity and mortality. The underlying aetiology is manifold including coronary artery disease, genetic alterations and mutations, viral infections, adverse immune responses, and cardiac toxicity. To date, no specific therapies have been developed despite notable efforts. This can especially be attributed to hurdles in translational research, mainly due to the lack of proficient models of heart failure limited translation of therapeutic approaches from bench to bedside. RECENT FINDINGS Human induced pluripotent stem cells (hiPSCs) are rising in popularity, granting the ability to divide infinitely, to hold human, patient-specific genome, and to differentiate into any human cell, including cardiomyocytes (hiPSC-CMs). This brings magnificent promise to cardiological research, providing the possibility to recapitulate cardiac diseases in a dish. Advances in yield, maturity, and in vivo resemblance due to straightforward, low-cost protocols, high-throughput approaches, and complex 3D cultures have made this tool widely applicable. In recent years, hiPSC-CMs have been used to model a wide variety of cardiac diseases, bringing along the possibility to not only elucidate molecular mechanisms but also to test novel therapeutic approaches in the dish. Within the last decade, hiPSC-CMs have been exponentially employed to model heart failure. Constant advancements are aiming at improvements of differentiation protocols, hiPSC-CM maturity, and assays to elucidate molecular mechanisms and cellular functions. However, hiPSC-CMs are remaining relatively immature, and in vitro models can only partially recapitulate the complex interactions in vivo. Nevertheless, hiPSC-CMs have evolved as an essential model system in cardiovascular research.
Collapse
Affiliation(s)
- Anton Deicher
- Department of Internal Medicine III, University Hospital Heidelberg, INF 410, 69126, Heidelberg, Germany
| | - Timon Seeger
- Department of Internal Medicine III, University Hospital Heidelberg, INF 410, 69126, Heidelberg, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
133
|
Yao Y, Marra AN, Yelon D. Pathways Regulating Establishment and Maintenance of Cardiac Chamber Identity in Zebrafish. J Cardiovasc Dev Dis 2021; 8:13. [PMID: 33572830 PMCID: PMC7912383 DOI: 10.3390/jcdd8020013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 02/07/2023] Open
Abstract
The vertebrate heart is comprised of two types of chambers-ventricles and atria-that have unique morphological and physiological properties. Effective cardiac function depends upon the distinct characteristics of ventricular and atrial cardiomyocytes, raising interest in the genetic pathways that regulate chamber-specific traits. Chamber identity seems to be specified in the early embryo by signals that establish ventricular and atrial progenitor populations and trigger distinct differentiation pathways. Intriguingly, chamber-specific features appear to require active reinforcement, even after myocardial differentiation is underway, suggesting plasticity of chamber identity within the developing heart. Here, we review the utility of the zebrafish as a model organism for studying the mechanisms that establish and maintain cardiac chamber identity. By combining genetic and embryological approaches, work in zebrafish has revealed multiple players with potent influences on chamber fate specification and commitment. Going forward, analysis of cardiomyocyte identity at the single-cell level is likely to yield a high-resolution understanding of the pathways that link the relevant players together, and these insights will have the potential to inform future strategies in cardiac tissue engineering.
Collapse
Affiliation(s)
| | | | - Deborah Yelon
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; (Y.Y.); (A.N.M.)
| |
Collapse
|
134
|
Peper J, Kownatzki-Danger D, Weninger G, Seibertz F, Pronto JRD, Sutanto H, Pacheu-Grau D, Hindmarsh R, Brandenburg S, Kohl T, Hasenfuss G, Gotthardt M, Rog-Zielinska EA, Wollnik B, Rehling P, Urlaub H, Wegener J, Heijman J, Voigt N, Cyganek L, Lenz C, Lehnart SE. Caveolin3 Stabilizes McT1-Mediated Lactate/Proton Transport in Cardiomyocytes. Circ Res 2021; 128:e102-e120. [PMID: 33486968 DOI: 10.1161/circresaha.119.316547] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Jonas Peper
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen (J.P., D.K.-D., G.W., S.B., T.K., G.H., J.W., S.E.L.), University Medical Center Göttingen.,Cardiology & Pneumology (J.P., D.K.-D., G.W., R.H., S.B., T.K., G.H., J.W., L.C., S.E.L.), University Medical Center Göttingen
| | - Daniel Kownatzki-Danger
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen (J.P., D.K.-D., G.W., S.B., T.K., G.H., J.W., S.E.L.), University Medical Center Göttingen.,Cardiology & Pneumology (J.P., D.K.-D., G.W., R.H., S.B., T.K., G.H., J.W., L.C., S.E.L.), University Medical Center Göttingen
| | - Gunnar Weninger
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen (J.P., D.K.-D., G.W., S.B., T.K., G.H., J.W., S.E.L.), University Medical Center Göttingen.,Cardiology & Pneumology (J.P., D.K.-D., G.W., R.H., S.B., T.K., G.H., J.W., L.C., S.E.L.), University Medical Center Göttingen
| | - Fitzwilliam Seibertz
- Institute of Pharmacology and Toxicology (F.S., J.R.D.P., N.V.), University Medical Center Göttingen.,DZHK (German Centre for Cardiovascular Research), partner site Göttingen (F.S., S.B., T.K., G.H., J.W., N.V., L.C., S.E.L.)
| | - Julius Ryan D Pronto
- Institute of Pharmacology and Toxicology (F.S., J.R.D.P., N.V.), University Medical Center Göttingen
| | - Henry Sutanto
- Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University (H.S., J.H.)
| | - David Pacheu-Grau
- Cellular Biochemistry, University Medical Center, Georg-August-University (D.P.G., P.R.)
| | - Robin Hindmarsh
- Cardiology & Pneumology (J.P., D.K.-D., G.W., R.H., S.B., T.K., G.H., J.W., L.C., S.E.L.), University Medical Center Göttingen
| | - Sören Brandenburg
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen (J.P., D.K.-D., G.W., S.B., T.K., G.H., J.W., S.E.L.), University Medical Center Göttingen.,Cardiology & Pneumology (J.P., D.K.-D., G.W., R.H., S.B., T.K., G.H., J.W., L.C., S.E.L.), University Medical Center Göttingen.,DZHK (German Centre for Cardiovascular Research), partner site Göttingen (F.S., S.B., T.K., G.H., J.W., N.V., L.C., S.E.L.)
| | - Tobias Kohl
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen (J.P., D.K.-D., G.W., S.B., T.K., G.H., J.W., S.E.L.), University Medical Center Göttingen.,Cardiology & Pneumology (J.P., D.K.-D., G.W., R.H., S.B., T.K., G.H., J.W., L.C., S.E.L.), University Medical Center Göttingen.,DZHK (German Centre for Cardiovascular Research), partner site Göttingen (F.S., S.B., T.K., G.H., J.W., N.V., L.C., S.E.L.)
| | - Gerd Hasenfuss
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen (J.P., D.K.-D., G.W., S.B., T.K., G.H., J.W., S.E.L.), University Medical Center Göttingen.,Cardiology & Pneumology (J.P., D.K.-D., G.W., R.H., S.B., T.K., G.H., J.W., L.C., S.E.L.), University Medical Center Göttingen.,DZHK (German Centre for Cardiovascular Research), partner site Göttingen (F.S., S.B., T.K., G.H., J.W., N.V., L.C., S.E.L.).,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen (G.H., B.W., P.R., N.V., S.E.L.)
| | - Michael Gotthardt
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin (M.G.).,Cardiology, Virchow Klinikum, Charité-University Medicine, Berlin (M.G.).,DZHK (German Center for Cardiovascular Research), partner site Berlin (M.G.)
| | - Eva A Rog-Zielinska
- University Heart Center, Faculty of Medicine, University of Freiburg (E.A.R.-Z.)
| | - Bernd Wollnik
- Institute of Human Genetics (B.W.), University Medical Center Göttingen.,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen (G.H., B.W., P.R., N.V., S.E.L.)
| | - Peter Rehling
- Cellular Biochemistry, University Medical Center, Georg-August-University (D.P.G., P.R.).,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen (G.H., B.W., P.R., N.V., S.E.L.)
| | - Henning Urlaub
- Bioanalytics, Institute of Clinical Chemistry (H.U., C.L.), University Medical Center Göttingen.,Bioanalytical Mass Spectrometry, Max Planck Institute for Biophysical Chemistry, Göttingen (H.U., C.L.)
| | - Jörg Wegener
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen (J.P., D.K.-D., G.W., S.B., T.K., G.H., J.W., S.E.L.), University Medical Center Göttingen.,Cardiology & Pneumology (J.P., D.K.-D., G.W., R.H., S.B., T.K., G.H., J.W., L.C., S.E.L.), University Medical Center Göttingen.,DZHK (German Centre for Cardiovascular Research), partner site Göttingen (F.S., S.B., T.K., G.H., J.W., N.V., L.C., S.E.L.)
| | - Jordi Heijman
- Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University (H.S., J.H.)
| | - Niels Voigt
- Institute of Pharmacology and Toxicology (F.S., J.R.D.P., N.V.), University Medical Center Göttingen.,DZHK (German Centre for Cardiovascular Research), partner site Göttingen (F.S., S.B., T.K., G.H., J.W., N.V., L.C., S.E.L.).,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen (G.H., B.W., P.R., N.V., S.E.L.)
| | - Lukas Cyganek
- DZHK (German Centre for Cardiovascular Research), partner site Göttingen (F.S., S.B., T.K., G.H., J.W., N.V., L.C., S.E.L.)
| | - Christof Lenz
- Bioanalytics, Institute of Clinical Chemistry (H.U., C.L.), University Medical Center Göttingen.,Bioanalytical Mass Spectrometry, Max Planck Institute for Biophysical Chemistry, Göttingen (H.U., C.L.)
| | - Stephan E Lehnart
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen (J.P., D.K.-D., G.W., S.B., T.K., G.H., J.W., S.E.L.), University Medical Center Göttingen.,Cardiology & Pneumology (J.P., D.K.-D., G.W., R.H., S.B., T.K., G.H., J.W., L.C., S.E.L.), University Medical Center Göttingen.,DZHK (German Centre for Cardiovascular Research), partner site Göttingen (F.S., S.B., T.K., G.H., J.W., N.V., L.C., S.E.L.).,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen (G.H., B.W., P.R., N.V., S.E.L.).,BioMET, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore (S.E.L.)
| |
Collapse
|
135
|
Arrhythmia Mechanisms in Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. J Cardiovasc Pharmacol 2020; 77:300-316. [PMID: 33323698 DOI: 10.1097/fjc.0000000000000972] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 12/08/2020] [Indexed: 12/30/2022]
Abstract
ABSTRACT Despite major efforts by clinicians and researchers, cardiac arrhythmia remains a leading cause of morbidity and mortality in the world. Experimental work has relied on combining high-throughput strategies with standard molecular and electrophysiological studies, which are, to a great extent, based on the use of animal models. Because this poses major challenges for translation, the progress in the development of novel antiarrhythmic agents and clinical care has been mostly disappointing. Recently, the advent of human induced pluripotent stem cell-derived cardiomyocytes has opened new avenues for both basic cardiac research and drug discovery; now, there is an unlimited source of cardiomyocytes of human origin, both from healthy individuals and patients with cardiac diseases. Understanding arrhythmic mechanisms is one of the main use cases of human induced pluripotent stem cell-derived cardiomyocytes, in addition to pharmacological cardiotoxicity and efficacy testing, in vitro disease modeling, developing patient-specific models and personalized drugs, and regenerative medicine. Here, we review the advances that the human induced pluripotent stem cell-derived-based modeling systems have brought so far regarding the understanding of both arrhythmogenic triggers and substrates, while also briefly speculating about the possibilities in the future.
Collapse
|
136
|
El-Battrawy I, Albers S, Cyganek L, Zhao Z, Lan H, Li X, Xu Q, Kleinsorge M, Huang M, Liao Z, Zhong R, Rudic B, Müller J, Dinkel H, Lang S, Diecke S, Zimmermann WH, Utikal J, Wieland T, Borggrefe M, Zhou X, Akin I. A cellular model of Brugada syndrome with SCN10A variants using human-induced pluripotent stem cell-derived cardiomyocytes. Europace 2020; 21:1410-1421. [PMID: 31106349 DOI: 10.1093/europace/euz122] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 04/03/2019] [Indexed: 11/15/2022] Open
Abstract
AIMS Brugada syndrome (BrS) is associated with a pronounced risk to develop sudden cardiac death (SCD). Up to 21% of patients are related to mutations in SCN5A. Studies identified SCN10A as a contributor of BrS. However, the investigation of the human cellular phenotype of BrS in the presence of SCN10A mutations remains lacking. The objective of this study was to establish a cellular model of BrS in presence of SCN10A mutations using human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). METHODS AND RESULTS Dermal fibroblasts obtained from a BrS patient suffering from SCD harbouring the SCN10A double variants (c.3803G>A and c.3749G>A) and three independent healthy control subjects were reprogrammed to hiPSCs. Human-induced pluripotent stem cells were differentiated into cardiomyocytes (hiPSC-CMs).The hiPSC-CMs from the BrS patient showed a significantly reduced peak sodium channel current (INa) and a significantly reduced ATX II (sea anemone toxin, an enhancer of late INa) sensitive as well as A-887826 (a blocker of SCN10A channel) sensitive late sodium channel current (INa) when compared with the healthy control hiPSC-CMs, indicating loss-of-function of sodium channels. Consistent with reduced INa the action potential amplitude and upstroke velocity (Vmax) were significantly reduced, which may contribute to arrhythmogenesis of BrS. Moreover, Ajmaline effects on action potentials were stronger in BrS-hiPSC-CMs than in healthy control cells. This is in agreement with the higher susceptibility of patients to sodium channel blocking drugs in unmasking BrS. CONCLUSION Patient-specific hiPSC-CMs are able to recapitulate single-cell phenotype features of BrS with SCN10A mutations and may provide novel opportunities to further elucidate the cellular disease mechanism.
Collapse
Affiliation(s)
- Ibrahim El-Battrawy
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim and Göttingen, Göttingen, Germany
| | - Sebastian Albers
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim and Göttingen, Göttingen, Germany
| | - Lukas Cyganek
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim and Göttingen, Göttingen, Germany.,Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Zhihan Zhao
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim and Göttingen, Göttingen, Germany
| | - Huan Lan
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim and Göttingen, Göttingen, Germany.,Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Xin Li
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany
| | - Qiang Xu
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany
| | - Mandy Kleinsorge
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim and Göttingen, Göttingen, Germany.,Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Mengying Huang
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany
| | - Zhenxing Liao
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany
| | - Rujia Zhong
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany
| | - Boris Rudic
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany
| | - Jonas Müller
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany
| | - Hendrik Dinkel
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany
| | - Siegfried Lang
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim and Göttingen, Göttingen, Germany
| | | | - Wolfram-Hubertus Zimmermann
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim and Göttingen, Göttingen, Germany.,Institute of Pharmacology and Toxicology, University of Göttingen, Göttingen, Germany
| | - Jochen Utikal
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim and Göttingen, Göttingen, Germany.,Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Thomas Wieland
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim and Göttingen, Göttingen, Germany.,Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Martin Borggrefe
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim and Göttingen, Göttingen, Germany
| | - Xiaobo Zhou
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim and Göttingen, Göttingen, Germany.,Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Ibrahim Akin
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim and Göttingen, Göttingen, Germany
| |
Collapse
|
137
|
Li J, Hua Y, Miyagawa S, Zhang J, Li L, Liu L, Sawa Y. hiPSC-Derived Cardiac Tissue for Disease Modeling and Drug Discovery. Int J Mol Sci 2020; 21:E8893. [PMID: 33255277 PMCID: PMC7727666 DOI: 10.3390/ijms21238893] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 12/20/2022] Open
Abstract
Relevant, predictive normal, or disease model systems are of vital importance for drug development. The difference between nonhuman models and humans could contribute to clinical trial failures despite ideal nonhuman results. As a potential substitute for animal models, human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs) provide a powerful tool for drug toxicity screening, modeling cardiovascular diseases, and drug discovery. Here, we review recent hiPSC-CM disease models and discuss the features of hiPSC-CMs, including subtype and maturation and the tissue engineering technologies for drug assessment. Updates from the international multisite collaborators/administrations for development of novel drug discovery paradigms are also summarized.
Collapse
Affiliation(s)
- Junjun Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
- Department of Cell Design for Tissue Construction, Faculty of Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ying Hua
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Jingbo Zhang
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Lingjun Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Li Liu
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
- Department of Design for Tissue Regeneration, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| |
Collapse
|
138
|
Li W, Stauske M, Luo X, Wagner S, Vollrath M, Mehnert CS, Schubert M, Cyganek L, Chen S, Hasheminasab SM, Wulf G, El-Armouche A, Maier LS, Hasenfuss G, Guan K. Disease Phenotypes and Mechanisms of iPSC-Derived Cardiomyocytes From Brugada Syndrome Patients With a Loss-of-Function SCN5A Mutation. Front Cell Dev Biol 2020; 8:592893. [PMID: 33195263 PMCID: PMC7642519 DOI: 10.3389/fcell.2020.592893] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 10/01/2020] [Indexed: 12/19/2022] Open
Abstract
Brugada syndrome (BrS) is one of the major causes of sudden cardiac death in young people, while the underlying mechanisms are not completely understood. Here, we investigated the pathophysiological phenotypes and mechanisms using induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CMs) from two BrS patients (BrS-CMs) carrying a heterozygous SCN5A mutation p.S1812X. Compared to CMs derived from healthy controls (Ctrl-CMs), BrS-CMs displayed a 50% reduction of INa density, a 69.5% reduction of NaV1.5 expression, and the impaired localization of NaV1.5 and connexin 43 (Cx43) at the cell surface. BrS-CMs exhibited reduced action potential (AP) upstroke velocity and conduction slowing. The Ito in BrS-CMs was significantly augmented, and the ICaL window current probability was increased. Our data indicate that the electrophysiological mechanisms underlying arrhythmia in BrS-CMs may involve both depolarization and repolarization disorders. Cilostazol and milrinone showed dramatic inhibitions of Ito in BrS-CMs and alleviated the arrhythmic activity, suggesting their therapeutic potential for BrS patients.
Collapse
Affiliation(s)
- Wener Li
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Michael Stauske
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Göttingen, Göttingen, Germany
| | - Xiaojing Luo
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Stefan Wagner
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,Department of Hematology and Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Meike Vollrath
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Carola S Mehnert
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Mario Schubert
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Lukas Cyganek
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Göttingen, Göttingen, Germany
| | - Simin Chen
- German Center for Cardiovascular Research (DZHK), Partner site Göttingen, Göttingen, Germany
| | - Sayed-Mohammad Hasheminasab
- Department of Dermatology, Venereology and Allergy, Charité - Universitätsmedizin Berlin, Berlin, Germany.,CCU Translational Radiation Oncology, German Cancer Consortium Core-Center Heidelberg, National Center for Tumor Diseases, Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Gerald Wulf
- Department of Hematology and Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Ali El-Armouche
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Lars S Maier
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,Clinic for Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Göttingen, Göttingen, Germany
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany.,Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Göttingen, Göttingen, Germany
| |
Collapse
|
139
|
Chen X, Zhang YS, Zhang X, Liu C. Organ-on-a-chip platforms for accelerating the evaluation of nanomedicine. Bioact Mater 2020; 6:1012-1027. [PMID: 33102943 PMCID: PMC7566214 DOI: 10.1016/j.bioactmat.2020.09.022] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/01/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Nanomedicine involves the use of engineered nanoscale materials in an extensive range of diagnostic and therapeutic applications and can be applied to the treatment of many diseases. Despite the rapid progress and tremendous potential of nanomedicine in the past decades, the clinical translational process is still quite slow, owing to the difficulty in understanding, evaluating, and predicting nanomaterial behaviors within the complex environment of human beings. Microfluidics-based organ-on-a-chip (Organ Chip) techniques offer a promising way to resolve these challenges. Sophisticatedly designed Organ Chip enable in vitro simulation of the in vivo microenvironments, thus providing robust platforms for evaluating nanomedicine. Herein, we review recent developments and achievements in Organ Chip models for nanomedicine evaluations, categorized into seven broad sections based on the target organ systems: respiratory, digestive, lymphatic, excretory, nervous, and vascular, as well as coverage on applications relating to cancer. We conclude by providing our perspectives on the challenges and potential future directions for applications of Organ Chip in nanomedicine. Microfluidics-based organ-on-a-chip (Organ Chip) techniques offer a promising way to understand, evaluate, and predict nanomedicine behaviors within the complex environment. Organ Chip models for nanomedicine evaluations are categorized into seven broad sections based on the targeted body systems. Limitations, challenges, and perspectives of Organ Chip for accelerating the assessment of nanomedicine are discussed, respectively.
Collapse
Affiliation(s)
- Xi Chen
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, United States
| | - Xinping Zhang
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| |
Collapse
|
140
|
Single-cell protein expression of hiPSC-derived cardiomyocytes using Single-Cell Westerns. J Mol Cell Cardiol 2020; 149:115-122. [PMID: 33010256 DOI: 10.1016/j.yjmcc.2020.09.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 09/21/2020] [Accepted: 09/25/2020] [Indexed: 11/22/2022]
Abstract
The ability to reprogram human somatic cells into human induced pluripotent stem cells (hiPSCs) has enabled researchers to generate cell types in vitro that have the potential to faithfully recapitulate patient-specific disease processes and phenotypes. hiPSC-derived cardiomyocytes (hiPSC-CMs) offer the promise of in vitro patient- and disease-specific models for drug testing and the discovery of novel therapeutic approaches for treating cardiovascular diseases. While methods to differentiate hiPSCs into cardiomyocytes have been demonstrated, the heterogeneity and immaturity of these differentiated populations have restricted their potential in reproducing human disease and the associated target cell phenotypes. These barriers may be overcome through comprehensive single-cell characterization to dissect the rich heterogeneity of hiPSC-CMs and to study the source of varying cell fates. In this study, we optimized and validated a new Single-Cell Western method to assess protein expression in hiPSC-CMs. To better understand distinct subpopulations generated from cardiomyocyte differentiations and to track populations at single-cell resolution over time, we measured and quantified the expression of cardiomyocyte subtype-specific proteins (MLC2V and MLC2A) using Single-Cell Westerns. By understanding their heterogeneity through single-cell protein expression and quantification, we may improve upon current cardiomyocyte differentiation protocols, generate hiPSC-CMs that are more representative of in vivo derived cardiomyocytes for disease modeling, and utilize hiPSC-CMs for regenerative medicine purposes. Single-Cell Westerns provide a robust platform for protein expression analysis at single-cell resolution.
Collapse
|
141
|
Assessment of Cardiotoxicity With Stem Cell-based Strategies. Clin Ther 2020; 42:1892-1910. [PMID: 32938533 DOI: 10.1016/j.clinthera.2020.08.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/20/2020] [Accepted: 08/25/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE Adverse cardiovascular drug effects pose a substantial medical risk and represent a common cause of drug withdrawal from the market. Thus, current in vitro assays and in vivo animal models still have shortcomings in assessing cardiotoxicity. A human model for more accurate preclinical cardiotoxicity assessment is highly desirable. Current differentiation protocols allow for the generation of human pluripotent stem cell-derived cardiomyocytes in basically unlimited numbers and offer the opportunity to study drug effects on human cardiomyocytes. The purpose of this review is to provide a brief overview of the current approaches to translate studies with pluripotent stem cell-derived cardiomyocytes from basic science to preclinical risk assessment. METHODS A review of the literature was performed to gather data on the pathophysiology of cardiotoxicity, the current cardiotoxicity screening assays, stem cell-derived cardiomyocytes, and their application in cardiotoxicity screening. FINDINGS There is increasing evidence that stem cell-derived cardiomyocytes predict arrhythmogenicity with high accuracy. Cardiomyocyte immaturity represents the major limitation so far. However, strategies are being developed to overcome this hurdle, such as tissue engineering. In addition, stem cell-based strategies offer the possibility to assess structural drug toxicity (eg, by anticancer drugs) on complex models that more closely mirror the structure of the heart and contain endothelial cells and fibroblasts. IMPLICATIONS Pluripotent stem cell-derived cardiomyocytes have the potential to substantially change how preclinical cardiotoxicity screening is performed. To which extent they will replace or complement current approaches is being evaluated.
Collapse
|
142
|
Selvakumar D, Clayton ZE, Chong JJH. Robust Cardiac Regeneration: Fulfilling the Promise of Cardiac Cell Therapy. Clin Ther 2020; 42:1857-1879. [PMID: 32943195 DOI: 10.1016/j.clinthera.2020.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/12/2020] [Accepted: 08/14/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE We review the history of cardiac cell therapy, highlighting lessons learned from initial adult stem cell (ASC) clinical trials. We present pluripotent stem cell-derived cardiomyocytes (PSC-CMs) as a leading candidate for robust regeneration of infarcted myocardium but identify several issues that must be addressed before successful clinical translation. METHODS We conducted an unstructured literature review of PubMed-listed articles, selecting the most comprehensive and relevant research articles, review articles, clinical trials, and basic or translation articles in the field of cardiac cell therapy. Articles were identified using the search terms adult stem cells, pluripotent stem cells, cardiac stem cell, and cardiac regeneration or from references of relevant articles, Articles were prioritized and selected based on their impact, originality, or potential clinical applicability. FINDINGS Since its inception, the ASC therapy field has been troubled by conflicting preclinical data, academic controversies, and inconsistent trial designs. These issues have damaged perceptions of cardiac cell therapy among investors, the academic community, health care professionals, and, importantly, patients. In hindsight, the key issue underpinning these problems was the inability of these cell types to differentiate directly into genuine cardiomyocytes, rendering them unable to replace damaged myocardium. Despite this, beneficial effects through indirect paracrine or immunomodulatory effects remain possible and continue to be investigated. However, in preclinical models, PSC-CMs have robustly remuscularized infarcted myocardium with functional, force-generating cardiomyocytes. Hence, PSC-CMs have now emerged as a leading candidate for cardiac regeneration, and unpublished reports of first-in-human delivery of these cells have recently surfaced. However, the cardiac cell therapy field's history should serve as a cautionary tale, and we identify several translational hurdles that still remain. Preclinical solutions to issues such as arrhythmogenicity, immunogenicity, and poor engraftment rates are needed, and next-generation clinical trials must draw on robust knowledge of mechanistic principles of the therapy. IMPLICATIONS The clinical transplantation of functional stem cell-derived heart tissue with seamless integration into native myocardium is a lofty goal. However, considerable advances have been made during the past 2 decades. Currently, PSC-CMs appear to be the best prospect to reach this goal, but several hurdles remain. The history of adult stem cell trials has taught us that shortcuts cannot be taken without dire consequences, and it is essential that progress not be hurried and that a worldwide, cross-disciplinary approach be used to ensure safe and effective clinical translation.
Collapse
Affiliation(s)
- Dinesh Selvakumar
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Zoe E Clayton
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - James J H Chong
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia.
| |
Collapse
|
143
|
Gunawan MG, Sangha SS, Shafaattalab S, Lin E, Heims-Waldron DA, Bezzerides VJ, Laksman Z, Tibbits GF. Drug screening platform using human induced pluripotent stem cell-derived atrial cardiomyocytes and optical mapping. Stem Cells Transl Med 2020; 10:68-82. [PMID: 32927497 PMCID: PMC7780813 DOI: 10.1002/sctm.19-0440] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 07/13/2020] [Accepted: 08/03/2020] [Indexed: 12/17/2022] Open
Abstract
Current drug development efforts for the treatment of atrial fibrillation are hampered by the fact that many preclinical models have been unsuccessful in reproducing human cardiac physiology and its response to medications. In this study, we demonstrated an approach using human induced pluripotent stem cell-derived atrial and ventricular cardiomyocytes (hiPSC-aCMs and hiPSC-vCMs, respectively) coupled with a sophisticated optical mapping system for drug screening of atrial-selective compounds in vitro. We optimized differentiation of hiPSC-aCMs by modulating the WNT and retinoid signaling pathways. Characterization of the transcriptome and proteome revealed that retinoic acid pushes the differentiation process into the atrial lineage and generated hiPSC-aCMs. Functional characterization using optical mapping showed that hiPSC-aCMs have shorter action potential durations and faster Ca2+ handling dynamics compared with hiPSC-vCMs. Furthermore, pharmacological investigation of hiPSC-aCMs captured atrial-selective effects by displaying greater sensitivity to atrial-selective compounds 4-aminopyridine, AVE0118, UCL1684, and vernakalant when compared with hiPSC-vCMs. These results established that a model system incorporating hiPSC-aCMs combined with optical mapping is well-suited for preclinical drug screening of novel and targeted atrial selective compounds.
Collapse
Affiliation(s)
- Marvin G Gunawan
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada.,Tibbits Research Team, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Sarabjit S Sangha
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada.,Tibbits Research Team, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Sanam Shafaattalab
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada.,Tibbits Research Team, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Eric Lin
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | | | | | - Zachary Laksman
- Division of Cardiology, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Heart and Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Glen F Tibbits
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada.,Tibbits Research Team, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| |
Collapse
|
144
|
Optimizing the Use of iPSC-CMs for Cardiac Regeneration in Animal Models. Animals (Basel) 2020; 10:ani10091561. [PMID: 32887495 PMCID: PMC7552322 DOI: 10.3390/ani10091561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 12/29/2022] Open
Abstract
Simple Summary In 2006, the first induced pluripotent stem cells were generated by reprogramming skin cells. Induced pluripotent stem cells undergo fast cell division, can differentiate into many different cell types, can be patient-specific, and do not raise ethical issues. Thus, they offer great promise as in vitro disease models, drug toxicity testing platforms, and for autologous tissue regeneration. Heart failure is one of the major causes of death worldwide. It occurs when the heart cannot meet the body’s metabolic demands. Induced pluripotent stem cells can be differentiated into cardiac myocytes, can form patches resembling native cardiac tissue, and can engraft to the damaged heart. However, despite correct host/graft coupling, most animal studies demonstrate an arrhythmogenicity of the engrafted tissue and variable survival. This is partially because of the heterogeneity and immaturity of the cells. New evidence suggests that by modulating induced pluripotent stem cells-cardiac myocytes (iPSC-CM) metabolism by switching substrates and changing metabolic pathways, you can decrease iPSC-CM heterogeneity and arrhythmogenicity. Novel culture methods and tissue engineering along with animal models of heart failure are needed to fully unlock the potential of cardiac myocytes derived from induced pluripotent stem cells for cardiac regeneration. Abstract Heart failure (HF) is a common disease in which the heart cannot meet the metabolic demands of the body. It mostly occurs in individuals 65 years or older. Cardiac transplantation is the best option for patients with advanced HF. High numbers of patient-specific cardiac myocytes (CMs) can be generated from induced pluripotent stem cells (iPSCs) and can possibly be used to treat HF. While some studies found iPSC-CMS can couple efficiently to the damaged heart and restore cardiac contractility, almost all found iPSC-CM transplantation is arrhythmogenic, thus hampering the use of iPSC-CMs for cardiac regeneration. Studies show that iPSC-CM cultures are highly heterogeneous containing atrial-, ventricular- and nodal-like CMs. Furthermore, they have an immature phenotype, resembling more fetal than adult CMs. There is an urgent need to overcome these issues. To this end, a novel and interesting avenue to increase CM maturation consists of modulating their metabolism. Combined with careful engineering and animal models of HF, iPSC-CMs can be assessed for their potential for cardiac regeneration and a cure for HF.
Collapse
|
145
|
Luo X, Li W, Künzel K, Henze S, Cyganek L, Strano A, Poetsch MS, Schubert M, Guan K. IP3R-Mediated Compensatory Mechanism for Calcium Handling in Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes With Cardiac Ryanodine Receptor Deficiency. Front Cell Dev Biol 2020; 8:772. [PMID: 32903370 PMCID: PMC7434870 DOI: 10.3389/fcell.2020.00772] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 07/22/2020] [Indexed: 02/03/2023] Open
Abstract
In adult cardiomyocytes (CMs), the type 2 ryanodine receptor (RYR2) is an indispensable Ca2+ release channel that ensures the integrity of excitation-contraction coupling, which is fundamental for every heartbeat. However, the role and importance of RYR2 during human embryonic cardiac development are still poorly understood. Here, we generated two human induced pluripotent stem cell (iPSC)-based RYR2 knockout (RYR2–/–) lines using the CRISPR/Cas9 gene editing technology. We found that RYR2–/–-iPSCs could differentiate into CMs with the efficiency similar to control-iPSCs (Ctrl-iPSCs); however, the survival of iPSC-CMs was markedly affected by the lack of functional RYR2. While Ctrl-iPSC-CMs exhibited regular Ca2+ handling, we observed significantly reduced frequency and intense abnormalities of Ca2+ transients in RYR2–/–-iPSC-CMs. Ctrl-iPSC-CMs displayed sensitivity to extracellular Ca2+ ([Ca2+ ]o) and caffeine in a concentration-dependent manner, while RYR2–/–-iPSC-CMs showed inconsistent reactions to [Ca2+ ]o and were insensitive to caffeine, indicating there is no RYR2-mediated Ca2+ release from the sarcoplasmic reticulum (SR). Instead, compensatory mechanism for calcium handling in RYR2–/–-iPSC-CMs is partially mediated by the inositol 1,4,5-trisphosphate receptor (IP3R). Similar to Ctrl-iPSC-CMs, SR Ca2+ refilling in RYR2–/–-iPSC-CMs is mediated by SERCA. Additionally, RYR2–/–-iPSC-CMs showed a decreased beating rate and a reduced peak amplitude of L-type Ca2+ current. These findings demonstrate that RYR2 is not required for CM lineage commitment but is important for CM survival and contractile function. IP3R-mediated Ca2+ release is one of the major compensatory mechanisms for Ca2+ cycling in human CMs with the RYR2 deficiency.
Collapse
Affiliation(s)
- Xiaojing Luo
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Wener Li
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Karolina Künzel
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Sarah Henze
- Clinic for Cardiology and Pneumology, Universitätsmedizin Göttingen, Göttingen, Germany
| | - Lukas Cyganek
- Clinic for Cardiology and Pneumology, Universitätsmedizin Göttingen, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Anna Strano
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Mareike S Poetsch
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Mario Schubert
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany.,Clinic for Cardiology and Pneumology, Universitätsmedizin Göttingen, Göttingen, Germany
| |
Collapse
|
146
|
Bektik E, Cowan DB, Wang DZ. Long Non-Coding RNAs in Atrial Fibrillation: Pluripotent Stem Cell-Derived Cardiomyocytes as a Model System. Int J Mol Sci 2020; 21:ijms21155424. [PMID: 32751460 PMCID: PMC7432754 DOI: 10.3390/ijms21155424] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) is a type of sustained arrhythmia in humans often characterized by devastating alterations to the cardiac conduction system as well as the structure of the atria. AF can lead to decreased cardiac function, heart failure, and other complications. Long non-coding RNAs (lncRNAs) have been shown to play important roles in the cardiovascular system, including AF; however, a large group of lncRNAs is not conserved between mouse and human. Furthermore, AF has complex networks showing variations in mechanisms in different species, making it challenging to utilize conventional animal models to investigate the functional roles and potential therapeutic benefits of lncRNAs for AF. Fortunately, pluripotent stem cell (PSC)-derived cardiomyocytes (CMs) offer a reliable platform to study lncRNA functions in AF because of certain electrophysiological and molecular similarities with native human CMs. In this review, we first summarize the broad aspects of lncRNAs in various heart disease settings, then focus on their potential roles in AF development and pathophysiology. We also discuss current uses of PSCs in AF research and describe how these studies could be developed into novel therapeutics for AF and other cardiovascular diseases.
Collapse
Affiliation(s)
- Emre Bektik
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood, Boston, MA 02115, USA; (E.B.); (D.B.C.)
| | - Douglas B. Cowan
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood, Boston, MA 02115, USA; (E.B.); (D.B.C.)
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood, Boston, MA 02115, USA; (E.B.); (D.B.C.)
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
- Correspondence:
| |
Collapse
|
147
|
Hilderink S, Devalla HD, Bosch L, Wilders R, Verkerk AO. Ultrarapid Delayed Rectifier K + Channelopathies in Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Front Cell Dev Biol 2020; 8:536. [PMID: 32850774 PMCID: PMC7399090 DOI: 10.3389/fcell.2020.00536] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/08/2020] [Indexed: 12/12/2022] Open
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia. About 5-15% of AF patients have a mutation in a cardiac gene, including mutations in KCNA5, encoding the Kv1.5 α-subunit of the ion channel carrying the atrial-specific ultrarapid delayed rectifier K+ current (IKur). Both loss-of-function and gain-of-function AF-related mutations in KCNA5 are known, but their effects on action potentials (APs) of human cardiomyocytes have been poorly studied. Here, we assessed the effects of wild-type and mutant IKur on APs of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). We found that atrial-like hiPSC-CMs, generated by a retinoic acid-based differentiation protocol, have APs with faster repolarization compared to ventricular-like hiPSC-CMs, resulting in shorter APs with a lower AP plateau. Native IKur, measured as current sensitive to 50 μM 4-aminopyridine, was 1.88 ± 0.49 (mean ± SEM, n = 17) and 0.26 ± 0.26 pA/pF (n = 17) in atrial- and ventricular-like hiPSC-CMs, respectively. In both atrial- and ventricular-like hiPSC-CMs, IKur blockade had minimal effects on AP parameters. Next, we used dynamic clamp to inject various amounts of a virtual IKur, with characteristics as in freshly isolated human atrial myocytes, into 11 atrial-like and 10 ventricular-like hiPSC-CMs, in which native IKur was blocked. Injection of IKur with 100% density shortened the APs, with its effect being strongest on the AP duration at 20% repolarization (APD20) of atrial-like hiPSC-CMs. At IKur densities < 100% (compared to 100%), simulating loss-of-function mutations, significant AP prolongation and raise of plateau were observed. At IKur densities > 100%, simulating gain-of-function mutations, APD20 was decreased in both atrial- and ventricular-like hiPSC-CMs, but only upon a strong increase in IKur. In ventricular-like hiPSC-CMs, lowering of the plateau resulted in AP shortening. We conclude that a decrease in IKur, mimicking loss-of-function mutations, has a stronger effect on the AP of hiPSC-CMs than an increase, mimicking gain-of-function mutations, whereas in ventricular-like hiPSC-CMs such increase results in AP shortening, causing their AP morphology to become more atrial-like. Effects of native IKur modulation on atrial-like hiPSC-CMs are less pronounced than effects of virtual IKur injection because IKur density of atrial-like hiPSC-CMs is substantially smaller than that of freshly isolated human atrial myocytes.
Collapse
Affiliation(s)
- Sarah Hilderink
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Harsha D Devalla
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Leontien Bosch
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - 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
| |
Collapse
|
148
|
Mapping signalling perturbations in myocardial fibrosis via the integrative phosphoproteomic profiling of tissue from diverse sources. Nat Biomed Eng 2020; 4:889-900. [PMID: 32661320 DOI: 10.1038/s41551-020-0585-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 06/14/2020] [Indexed: 12/13/2022]
Abstract
Study of the molecular basis of myocardial fibrosis is hampered by limited access to tissues from human patients and by confounding variables associated with sample accessibility, collection, processing and storage. Here, we report an integrative strategy based on mass spectrometry for the phosphoproteomic profiling of normal and fibrotic cardiac tissue obtained from surgical explants from patients with hypertrophic cardiomyopathy, from a transaortic-constriction mouse model of cardiac hypertrophy and fibrosis, and from a heart-on-a-chip model of cardiac fibrosis. We used the integrative approach to map the relative abundance of thousands of proteins, phosphoproteins and phosphorylation sites specific to each tissue source, to identify key signalling pathways driving fibrosis and to screen for anti-fibrotic compounds targeting glycogen synthase kinase 3, which has a consistent role as a key mediator of fibrosis in all three types of tissue specimen. The integrative disease-modelling strategy may reveal new insights into mechanisms of cardiac disease and serve as a test bed for drug screening.
Collapse
|
149
|
Hanses U, Kleinsorge M, Roos L, Yigit G, Li Y, Barbarics B, El-Battrawy I, Lan H, Tiburcy M, Hindmarsh R, Lenz C, Salinas G, Diecke S, Müller C, Adham I, Altmüller J, Nürnberg P, Paul T, Zimmermann WH, Hasenfuss G, Wollnik B, Cyganek L. Intronic CRISPR Repair in a Preclinical Model of Noonan Syndrome-Associated Cardiomyopathy. Circulation 2020; 142:1059-1076. [PMID: 32623905 DOI: 10.1161/circulationaha.119.044794] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Noonan syndrome (NS) is a multisystemic developmental disorder characterized by common, clinically variable symptoms, such as typical facial dysmorphisms, short stature, developmental delay, intellectual disability as well as cardiac hypertrophy. The underlying mechanism is a gain-of-function of the RAS-mitogen-activated protein kinase signaling pathway. However, our understanding of the pathophysiological alterations and mechanisms, especially of the associated cardiomyopathy, remains limited and effective therapeutic options are lacking. METHODS Here, we present a family with two siblings displaying an autosomal recessive form of NS with massive hypertrophic cardiomyopathy as clinically the most prevalent symptom caused by biallelic mutations within the leucine zipper-like transcription regulator 1 (LZTR1). We generated induced pluripotent stem cell-derived cardiomyocytes of the affected siblings and investigated the patient-specific cardiomyocytes on the molecular and functional level. RESULTS Patients' induced pluripotent stem cell-derived cardiomyocytes recapitulated the hypertrophic phenotype and uncovered a so-far-not-described causal link between LZTR1 dysfunction, RAS-mitogen-activated protein kinase signaling hyperactivity, hypertrophic gene response and cellular hypertrophy. Calcium channel blockade and MEK inhibition could prevent some of the disease characteristics, providing a molecular underpinning for the clinical use of these drugs in patients with NS, but might not be a sustainable therapeutic option. In a proof-of-concept approach, we explored a clinically translatable intronic CRISPR (clustered regularly interspaced short palindromic repeats) repair and demonstrated a rescue of the hypertrophic phenotype. CONCLUSIONS Our study revealed the human cardiac pathogenesis in patient-specific induced pluripotent stem cell-derived cardiomyocytes from NS patients carrying biallelic variants in LZTR1 and identified a unique disease-specific proteome signature. In addition, we identified the intronic CRISPR repair as a personalized and in our view clinically translatable therapeutic strategy to treat NS-associated hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Ulrich Hanses
- Clinic for Cardiology and Pneumology (U.H., M.K., L.R., R.H., G.H., L.C.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Mandy Kleinsorge
- Clinic for Cardiology and Pneumology (U.H., M.K., L.R., R.H., G.H., L.C.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Lennart Roos
- Clinic for Cardiology and Pneumology (U.H., M.K., L.R., R.H., G.H., L.C.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Gökhan Yigit
- Institute of Human Genetics (G.Y., Y.L., G.S., C.M., I.A., B.W.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Yun Li
- Institute of Human Genetics (G.Y., Y.L., G.S., C.M., I.A., B.W.)
| | - Boris Barbarics
- Clinic for Pediatric Cardiology and Intensive Care Medicine (B.B., T.P.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Ibrahim El-Battrawy
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.).,First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany (I.E-B., H.L.)
| | - Huan Lan
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany (I.E-B., H.L.)
| | - Malte Tiburcy
- Institute of Pharmacology and Toxicology (M.T., W-H.Z.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Robin Hindmarsh
- Clinic for Cardiology and Pneumology (U.H., M.K., L.R., R.H., G.H., L.C.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Christof Lenz
- Institute for Clinical Chemistry (C.L.), University Medical Center Göttingen, Germany.,Bioanalytical Mass Spectrometry, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany (C.L.)
| | - Gabriela Salinas
- Institute of Human Genetics (G.Y., Y.L., G.S., C.M., I.A., B.W.)
| | - Sebastian Diecke
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.).,Stem Cell Core Facility, Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.D.).,Berlin Institute of Health, Germany (S.D.)
| | - Christian Müller
- Institute of Human Genetics (G.Y., Y.L., G.S., C.M., I.A., B.W.)
| | - Ibrahim Adham
- Institute of Human Genetics (G.Y., Y.L., G.S., C.M., I.A., B.W.)
| | - Janine Altmüller
- Cologne Center for Genomics, University of Cologne, Germany (J.A., P.N.)
| | - Peter Nürnberg
- Cologne Center for Genomics, University of Cologne, Germany (J.A., P.N.)
| | - Thomas Paul
- Clinic for Pediatric Cardiology and Intensive Care Medicine (B.B., T.P.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology (M.T., W-H.Z.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.).,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W-H.Z., G.H., B.W.)
| | - Gerd Hasenfuss
- Clinic for Cardiology and Pneumology (U.H., M.K., L.R., R.H., G.H., L.C.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.).,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W-H.Z., G.H., B.W.)
| | - Bernd Wollnik
- Institute of Human Genetics (G.Y., Y.L., G.S., C.M., I.A., B.W.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.).,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W-H.Z., G.H., B.W.)
| | - Lukas Cyganek
- Clinic for Cardiology and Pneumology (U.H., M.K., L.R., R.H., G.H., L.C.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| |
Collapse
|
150
|
Induced pluripotent stem cells of patients with Tetralogy of Fallot reveal transcriptional alterations in cardiomyocyte differentiation. Sci Rep 2020; 10:10921. [PMID: 32616843 PMCID: PMC7331606 DOI: 10.1038/s41598-020-67872-z] [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: 02/26/2020] [Accepted: 06/16/2020] [Indexed: 12/27/2022] Open
Abstract
Patient-specific induced pluripotent stem cells (ps-iPSCs) and their differentiated cell types are a powerful model system to gain insight into mechanisms driving early developmental and disease-associated regulatory networks. In this study, we use ps-iPSCs to gain insights into Tetralogy of Fallot (TOF), which represents the most common cyanotic heart defect in humans. iPSCs were generated and further differentiated to cardiomyocytes (CMs) using standard methods from two well-characterized TOF patients and their healthy relatives serving as controls. Patient-specific expression patterns and genetic variability were investigated using whole genome and transcriptome sequencing data. We first studied the clonal mutational burden of the derived iPSCs. In two out of three iPSC lines of patient TOF-01, we found a somatic mutation in the DNA-binding domain of tumor suppressor P53, which was not observed in the genomic DNA from blood. Further characterization of this mutation showed its functional impact. For patient TOF-02, potential disease-relevant differential gene expression between and across cardiac differentiation was shown. Here, clear differences at the later stages of differentiation could be observed between CMs of the patient and its controls. Overall, this study provides first insights into the complex molecular mechanisms underlying iPSC-derived cardiomyocyte differentiation and its transcriptional alterations in TOF.
Collapse
|