1
|
House A, Santillan A, Correa E, Youssef V, Guvendiren M. Cellular Alignment and Matrix Stiffening Induced Changes in Human Induced Pluripotent Stem Cell Derived Cardiomyocytes. Adv Healthc Mater 2024:e2402228. [PMID: 39468891 DOI: 10.1002/adhm.202402228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/15/2024] [Indexed: 10/30/2024]
Abstract
Biological processes are inherently dynamic, necessitating biomaterial platforms capable of spatiotemporal control over cellular organization and matrix stiffness for accurate study of tissue development, wound healing, and disease. However, most in vitro platforms remain static. In this study, a dynamic biomaterial platform comprising a stiffening hydrogel is introduced and achieved through a stepwise approach of addition followed by light-mediated crosslinking, integrated with an elastomeric substrate featuring strain-responsive lamellar surface patterns. Employing this platform, the response of human induced pluripotent stem cell-derived cardiomyocytes (hIPSC-CMs) is investigated to dynamic stiffening from healthy to fibrotic tissue stiffness. The results demonstrate that culturing hIPSC-CMs on physiologically relevant healthy stiffness significantly enhances their function, as evidenced by increased sarcomere fraction, wider sarcomere width, significantly higher connexin-43 content, and elevated cell beating frequency compared to cells cultured on fibrotic matrix. Conversely, dynamic matrix stiffening negatively impacts hIPSC-CM function, with earlier stiffening events exerting a more pronounced hindering effect. These findings provide valuable insights into material-based approaches for addressing existing challenges in hIPSC-CM maturation and have broader implications across various tissue models, including muscle, tendon, nerve, and cornea, where both cellular alignment and matrix stiffening play pivotal roles in tissue development and regeneration.
Collapse
Affiliation(s)
- Andrew House
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, University Heights, Newark, NJ, 07102, USA
| | - Anjeli Santillan
- Department of Biomedical Engineering, New Jersey Institute of Technology, University Heights, Newark, NJ, 07102, USA
| | - Evan Correa
- Federated Department of Biological Sciences, New Jersey Institute of Technology, University Heights, Newark, NJ, 07102, USA
| | - Victoria Youssef
- Federated Department of Biological Sciences, New Jersey Institute of Technology, University Heights, Newark, NJ, 07102, USA
| | - Murat Guvendiren
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, University Heights, Newark, NJ, 07102, USA
- Department of Biomedical Engineering, New Jersey Institute of Technology, University Heights, Newark, NJ, 07102, USA
| |
Collapse
|
2
|
Kistamás K, Lamberto F, Vaiciuleviciute R, Leal F, Muenthaisong S, Marte L, Subías-Beltrán P, Alaburda A, Arvanitis DN, Zana M, Costa PF, Bernotiene E, Bergaud C, Dinnyés A. The Current State of Realistic Heart Models for Disease Modelling and Cardiotoxicity. Int J Mol Sci 2024; 25:9186. [PMID: 39273136 PMCID: PMC11394806 DOI: 10.3390/ijms25179186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/18/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
One of the many unresolved obstacles in the field of cardiovascular research is an uncompromising in vitro cardiac model. While primary cell sources from animal models offer both advantages and disadvantages, efforts over the past half-century have aimed to reduce their use. Additionally, obtaining a sufficient quantity of human primary cardiomyocytes faces ethical and legal challenges. As the practically unlimited source of human cardiomyocytes from induced pluripotent stem cells (hiPSC-CM) is now mostly resolved, there are great efforts to improve their quality and applicability by overcoming their intrinsic limitations. The greatest bottleneck in the field is the in vitro ageing of hiPSC-CMs to reach a maturity status that closely resembles that of the adult heart, thereby allowing for more appropriate drug developmental procedures as there is a clear correlation between ageing and developing cardiovascular diseases. Here, we review the current state-of-the-art techniques in the most realistic heart models used in disease modelling and toxicity evaluations from hiPSC-CM maturation through heart-on-a-chip platforms and in silico models to the in vitro models of certain cardiovascular diseases.
Collapse
Affiliation(s)
- Kornél Kistamás
- BioTalentum Ltd., Aulich Lajos Str 26, H-2100 Gödöllő, Hungary
| | - Federica Lamberto
- BioTalentum Ltd., Aulich Lajos Str 26, H-2100 Gödöllő, Hungary
- Department of Physiology and Animal Health, Institute of Physiology and Animal Nutrition, Hungarian University of Agriculture and Life Sciences, Páter Károly Str 1, H-2100 Gödöllő, Hungary
| | - Raminta Vaiciuleviciute
- Department of Regenerative Medicine, State Research Institute Innovative Medicine Centre, Santariskiu g. 5, LT-08406 Vilnius, Lithuania
| | - Filipa Leal
- Biofabics Lda, Rua Alfredo Allen 455, 4200-135 Porto, Portugal
| | | | - Luis Marte
- Digital Health Unit, Eurecat-Centre Tecnològic de Catalunya, 08005 Barcelona, Spain
| | - Paula Subías-Beltrán
- Digital Health Unit, Eurecat-Centre Tecnològic de Catalunya, 08005 Barcelona, Spain
| | - Aidas Alaburda
- Department of Regenerative Medicine, State Research Institute Innovative Medicine Centre, Santariskiu g. 5, LT-08406 Vilnius, Lithuania
- Institute of Biosciences, Life Sciences Center, Vilnius University, Sauletekio al. 7, LT-10257 Vilnius, Lithuania
| | - Dina N Arvanitis
- Laboratory for Analysis and Architecture of Systems-French National Centre for Scientific Research (LAAS-CNRS), 7 Avenue du Colonel Roche, F-31400 Toulouse, France
| | - Melinda Zana
- BioTalentum Ltd., Aulich Lajos Str 26, H-2100 Gödöllő, Hungary
| | - Pedro F Costa
- Biofabics Lda, Rua Alfredo Allen 455, 4200-135 Porto, Portugal
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Innovative Medicine Centre, Santariskiu g. 5, LT-08406 Vilnius, Lithuania
- Faculty of Fundamental Sciences, Vilnius Tech, Sauletekio al. 11, LT-10223 Vilnius, Lithuania
| | - Christian Bergaud
- Laboratory for Analysis and Architecture of Systems-French National Centre for Scientific Research (LAAS-CNRS), 7 Avenue du Colonel Roche, F-31400 Toulouse, France
| | - András Dinnyés
- BioTalentum Ltd., Aulich Lajos Str 26, H-2100 Gödöllő, Hungary
- Department of Physiology and Animal Health, Institute of Physiology and Animal Nutrition, Hungarian University of Agriculture and Life Sciences, Páter Károly Str 1, H-2100 Gödöllő, Hungary
| |
Collapse
|
3
|
Clancy CE, Santana LF. Advances in induced pluripotent stem cell-derived cardiac myocytes: technological breakthroughs, key discoveries and new applications. J Physiol 2024; 602:3871-3892. [PMID: 39032073 PMCID: PMC11326976 DOI: 10.1113/jp282562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 07/02/2024] [Indexed: 07/22/2024] Open
Abstract
A transformation is underway in precision and patient-specific medicine. Rapid progress has been enabled by multiple new technologies including induced pluripotent stem cell-derived cardiac myocytes (iPSC-CMs). Here, we delve into these advancements and their future promise, focusing on the efficiency of reprogramming techniques, the fidelity of differentiation into the cardiac lineage, the functional characterization of the resulting cardiac myocytes, and the many applications of in silico models to understand general and patient-specific mechanisms controlling excitation-contraction coupling in health and disease. Furthermore, we explore the current and potential applications of iPSC-CMs in both research and clinical settings, underscoring the far-reaching implications of this rapidly evolving field.
Collapse
Affiliation(s)
- Colleen E Clancy
- Department of Physiology & Membrane Biology, School of Medicine, University of California Davis, Davis, CA, USA
- Center for Precision Medicine and Data Sciences, University of California Davis, School of Medicine, Sacramento, CA, USA
| | - L Fernando Santana
- Department of Physiology & Membrane Biology, School of Medicine, University of California Davis, Davis, CA, USA
- Center for Precision Medicine and Data Sciences, University of California Davis, School of Medicine, Sacramento, CA, USA
| |
Collapse
|
4
|
Lickiss B, Hunker J, Bhagwan J, Linder P, Thomas U, Lotay H, Broadbent S, Dragicevic E, Stoelzle-Feix S, Turner J, Gossmann M. Chamber-specific contractile responses of atrial and ventricular hiPSC-cardiomyocytes to GPCR and ion channel targeting compounds: A microphysiological system for cardiac drug development. J Pharmacol Toxicol Methods 2024; 128:107529. [PMID: 38857637 DOI: 10.1016/j.vascn.2024.107529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/15/2024] [Accepted: 06/05/2024] [Indexed: 06/12/2024]
Abstract
Human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs) have found utility for conducting in vitro drug screening and disease modelling to gain crucial insights into pharmacology or disease phenotype. However, diseases such as atrial fibrillation, affecting >33 M people worldwide, demonstrate the need for cardiac subtype-specific cells. Here, we sought to investigate the base characteristics and pharmacological differences between commercially available chamber-specific atrial or ventricular hiPSC-CMs seeded onto ultra-thin, flexible PDMS membranes to simultaneously measure contractility in a 96 multi-well format. We investigated the effects of GPCR agonists (acetylcholine and carbachol), a Ca2+ channel agonist (S-Bay K8644), an HCN channel antagonist (ivabradine) and K+ channel antagonists (4-AP and vernakalant). We observed differential effects between atrial and ventricular hiPSC-CMs on contractile properties including beat rate, beat duration, contractile force and evidence of arrhythmias at a range of concentrations. As an excerpt of the compound analysis, S-Bay K8644 treatment showed an induced concentration-dependent transient increase in beat duration of atrial hiPSC-CMs, whereas ventricular cells showed a physiological increase in beat rate over time. Carbachol treatment produced marked effects on atrial cells, such as increased beat duration alongside a decrease in beat rate over time, but only minimal effects on ventricular cardiomyocytes. In the context of this chamber-specific pharmacology, we not only add to contractile characterization of hiPSC-CMs but propose a multi-well platform for medium-throughput early compound screening. Overall, these insights illustrate the key pharmacological differences between chamber-specific cardiomyocytes and their application on a multi-well contractility platform to gain insights for in vitro cardiac liability studies and disease modelling.
Collapse
Affiliation(s)
| | - Jan Hunker
- innoVitro GmbH, Artilleriestr 2, 52428 Jülich, Germany
| | - Jamie Bhagwan
- Axol Bioscience Ltd, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Peter Linder
- innoVitro GmbH, Artilleriestr 2, 52428 Jülich, Germany
| | - Ulrich Thomas
- Nanion Technologies GmbH, Ganghoferstr 70A, 80339 Munich, Germany
| | - Hardeep Lotay
- Axol Bioscience Ltd, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Steven Broadbent
- Axol Bioscience Ltd, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Elena Dragicevic
- Nanion Technologies GmbH, Ganghoferstr 70A, 80339 Munich, Germany
| | | | - Jan Turner
- Axol Bioscience Ltd, Babraham Research Campus, Cambridge CB22 3AT, UK
| | | |
Collapse
|
5
|
Yan W, Xia Y, Zhao H, Xu X, Ma X, Tao L. Stem cell-based therapy in cardiac repair after myocardial infarction: Promise, challenges, and future directions. J Mol Cell Cardiol 2024; 188:1-14. [PMID: 38246086 DOI: 10.1016/j.yjmcc.2023.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/09/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024]
Abstract
Stem cells represent an attractive resource for cardiac regeneration. However, the survival and function of transplanted stem cells is poor and remains a major challenge for the development of effective therapies. As two main cell types currently under investigation in heart repair, mesenchymal stromal cells (MSCs) indirectly support endogenous regenerative capacities after transplantation, while induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) functionally integrate into the damaged myocardium and directly contribute to the restoration of its pump function. These two cell types are exposed to a common microenvironment with many stressors in ischemic heart tissue. This review summarizes the research progress on the mechanisms and challenges of MSCs and iPSC-CMs in post-MI heart repair, introduces several randomized clinical trials with 3D-mapping-guided cell therapy, and outlines recent findings related to the factors that affect the survival and function of stem cells. We also discuss the future directions for optimization such as biomaterial utilization, cell combinations, and intravenous injection of engineered nucleus-free MSCs.
Collapse
Affiliation(s)
- Wenjun Yan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yunlong Xia
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Huishou Zhao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Xiaoming Xu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
6
|
Ronchi C, Galli C, Tullii G, Marzuoli C, Mazzola M, Malferrari M, Crasto S, Rapino S, Di Pasquale E, Antognazza MR. Nongenetic Optical Modulation of Pluripotent Stem Cells Derived Cardiomyocytes Function in the Red Spectral Range. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304303. [PMID: 37948328 PMCID: PMC10797444 DOI: 10.1002/advs.202304303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/24/2023] [Indexed: 11/12/2023]
Abstract
Optical stimulation in the red/near infrared range recently gained increasing interest, as a not-invasive tool to control cardiac cell activity and repair in disease conditions. Translation of this approach to therapy is hampered by scarce efficacy and selectivity. The use of smart biocompatible materials, capable to act as local, NIR-sensitive interfaces with cardiac cells, may represent a valuable solution, capable to overcome these limitations. In this work, a far red-responsive conjugated polymer, namely poly[2,1,3-benzothiadiazole-4,7-diyl[4,4-bis(2-ethylhexyl)-4H-cyclopenta[2,1-b:3,4-b']dithiophene-2,6-diyl]] (PCPDTBT) is proposed for the realization of photoactive interfaces with cardiomyocytes derived from pluripotent stem cells (hPSC-CMs). Optical excitation of the polymer turns into effective ionic and electrical modulation of hPSC-CMs, in particular by fastening Ca2+ dynamics, inducing action potential shortening, accelerating the spontaneous beating frequency. The involvement in the phototransduction pathway of Sarco-Endoplasmic Reticulum Calcium ATPase (SERCA) and Na+ /Ca2+ exchanger (NCX) is proven by pharmacological assays and is correlated with physical/chemical processes occurring at the polymer surface upon photoexcitation. Very interestingly, an antiarrhythmogenic effect, unequivocally triggered by polymer photoexcitation, is also observed. Overall, red-light excitation of conjugated polymers may represent an unprecedented opportunity for fine control of hPSC-CMs functionality and can be considered as a perspective, noninvasive approach to treat arrhythmias.
Collapse
Affiliation(s)
- Carlotta Ronchi
- Center for Nano Science and TechnologyIstituto Italiano di TecnologiaMilano20133Italy
| | - Camilla Galli
- Humanitas Cardio CenterIRCCS Humanitas Research HospitalVia Manzoni 56RozzanoMilan20089Italy
| | - Gabriele Tullii
- Center for Nano Science and TechnologyIstituto Italiano di TecnologiaMilano20133Italy
| | - Camilla Marzuoli
- Center for Nano Science and TechnologyIstituto Italiano di TecnologiaMilano20133Italy
- Politecnico di MilanoPhysics Dept.P.zza L. Da Vinci 32Milano20133Italy
| | - Marta Mazzola
- Humanitas Cardio CenterIRCCS Humanitas Research HospitalVia Manzoni 56RozzanoMilan20089Italy
| | - Marco Malferrari
- Department of Chemistry, University of Bologna‘‘Giacomo Ciamician,’’via Francesco Selmi 2Bologna40126Italy
| | - Silvia Crasto
- Humanitas Cardio CenterIRCCS Humanitas Research HospitalVia Manzoni 56RozzanoMilan20089Italy
| | - Stefania Rapino
- Department of Chemistry, University of Bologna‘‘Giacomo Ciamician,’’via Francesco Selmi 2Bologna40126Italy
| | - Elisa Di Pasquale
- Humanitas Cardio CenterIRCCS Humanitas Research HospitalVia Manzoni 56RozzanoMilan20089Italy
- Institute of Genetic and Biomedical Research (IRGB)UOS of Milan—National Research Council of Italy (CNR)Milan20138Italy
| | - Maria Rosa Antognazza
- Center for Nano Science and TechnologyIstituto Italiano di TecnologiaMilano20133Italy
| |
Collapse
|
7
|
Wauchop M, Rafatian N, Zhao Y, Chen W, Gagliardi M, Massé S, Cox BJ, Lai P, Liang T, Landau S, Protze S, Gao XD, Wang EY, Tung KC, Laksman Z, Lu RXZ, Keller G, Nanthakumar K, Radisic M, Backx PH. Maturation of iPSC-derived cardiomyocytes in a heart-on-a-chip device enables modeling of dilated cardiomyopathy caused by R222Q-SCN5A mutation. Biomaterials 2023; 301:122255. [PMID: 37651922 PMCID: PMC10942743 DOI: 10.1016/j.biomaterials.2023.122255] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/17/2023] [Accepted: 07/23/2023] [Indexed: 09/02/2023]
Abstract
To better understand sodium channel (SCN5A)-related cardiomyopathies, we generated ventricular cardiomyocytes from induced pluripotent stem cells obtained from a dilated cardiomyopathy patient harbouring the R222Q mutation, which is only expressed in adult SCN5A isoforms. Because the adult SCN5A isoform was poorly expressed, without functional differences between R222Q and control in both embryoid bodies and cell sheet preparations (cultured for 29-35 days), we created heart-on-a-chip biowires which promote myocardial maturation. Indeed, biowires expressed primarily adult SCN5A with R222Q preparations displaying (arrhythmogenic) short action potentials, altered Na+ channel biophysical properties and lower contractility compared to corrected controls. Comprehensive RNA sequencing revealed differential gene regulation between R222Q and control biowires in cellular pathways related to sarcoplasmic reticulum and dystroglycan complex as well as biological processes related to calcium ion regulation and action potential. Additionally, R222Q biowires had marked reductions in actin expression accompanied by profound sarcoplasmic disarray, without differences in cell composition (fibroblast, endothelial cells, and cardiomyocytes) compared to corrected biowires. In conclusion, we demonstrate that in addition to altering cardiac electrophysiology and Na+ current, the R222Q mutation also causes profound sarcomere disruptions and mechanical destabilization. Possible mechanisms for these observations are discussed.
Collapse
Affiliation(s)
- Marianne Wauchop
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Naimeh Rafatian
- Division of Cardiology and Peter Munk Cardiac Center, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Yimu Zhao
- Toronto General Hospital Research Institute, Toronto, ON, M5G 2C4, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
| | - Wenliang Chen
- Division of Cardiology and Peter Munk Cardiac Center, University Health Network, Toronto, ON, M5G 1L7, Canada; Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
| | - Mark Gagliardi
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Stéphane Massé
- Division of Cardiology and Peter Munk Cardiac Center, University Health Network, Toronto, ON, M5G 1L7, Canada; Toronto General Hospital Research Institute, Toronto, ON, M5G 2C4, Canada; The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, Toronto, ON, M5G 2C4, Canada
| | - Brian J Cox
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada; Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Patrick Lai
- Division of Cardiology and Peter Munk Cardiac Center, University Health Network, Toronto, ON, M5G 1L7, Canada; Toronto General Hospital Research Institute, Toronto, ON, M5G 2C4, Canada; The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, Toronto, ON, M5G 2C4, Canada
| | - Timothy Liang
- Division of Cardiology and Peter Munk Cardiac Center, University Health Network, Toronto, ON, M5G 1L7, Canada; Toronto General Hospital Research Institute, Toronto, ON, M5G 2C4, Canada; The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, Toronto, ON, M5G 2C4, Canada
| | - Shira Landau
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
| | - Stephanie Protze
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Xiao Dong Gao
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
| | - Erika Yan Wang
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
| | - Kelvin Chan Tung
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Zachary Laksman
- Department of Medicine, University of British Columbia, Vancouver, BC, V6E 1M7, Canada
| | - Rick Xing Ze Lu
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
| | - Gordon Keller
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada
| | - Kumaraswamy Nanthakumar
- Division of Cardiology and Peter Munk Cardiac Center, University Health Network, Toronto, ON, M5G 1L7, Canada; Toronto General Hospital Research Institute, Toronto, ON, M5G 2C4, Canada; The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, Toronto, ON, M5G 2C4, Canada.
| | - Milica Radisic
- Toronto General Hospital Research Institute, Toronto, ON, M5G 2C4, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada, M5S 3E5.
| | - Peter H Backx
- Division of Cardiology and Peter Munk Cardiac Center, University Health Network, Toronto, ON, M5G 1L7, Canada; Department of Biology, York University, Toronto, ON, M3J 1P3, Canada; Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1, Canada.
| |
Collapse
|
8
|
Wang K, Schriver BJ, Aschar-Sobbi R, Yi AY, Feric NT, Graziano MP. Human engineered cardiac tissue model of hypertrophic cardiomyopathy recapitulates key hallmarks of the disease and the effect of chronic mavacamten treatment. Front Bioeng Biotechnol 2023; 11:1227184. [PMID: 37771571 PMCID: PMC10523579 DOI: 10.3389/fbioe.2023.1227184] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/28/2023] [Indexed: 09/30/2023] Open
Abstract
Introduction: The development of patient-specific induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) offers an opportunity to study genotype-phenotype correlation of hypertrophic cardiomyopathy (HCM), one of the most common inherited cardiac diseases. However, immaturity of the iPSC-CMs and the lack of a multicellular composition pose concerns over its faithfulness in disease modeling and its utility in developing mechanism-specific treatment. Methods: The Biowire platform was used to generate 3D engineered cardiac tissues (ECTs) using HCM patient-derived iPSC-CMs carrying a β-myosin mutation (MYH7-R403Q) and its isogenic control (WT), withal ECTs contained healthy human cardiac fibroblasts. ECTs were subjected to electro-mechanical maturation for 6 weeks before being used in HCM phenotype studies. Results: Both WT and R403Q ECTs exhibited mature cardiac phenotypes, including a lack of automaticity and a ventricular-like action potential (AP) with a resting membrane potential < -75 mV. Compared to WT, R403Q ECTs demonstrated many HCM-associated pathological changes including increased tissue size and cell volume, shortened sarcomere length and disorganized sarcomere structure. In functional assays, R403Q ECTs showed increased twitch amplitude, slower contractile kinetics, a less pronounced force-frequency relationship, a smaller post-rest potentiation, prolonged AP durations, and slower Ca2+ transient decay time. Finally, we observed downregulation of calcium handling genes and upregulation of NPPB in R403Q vs. WT ECTs. In an HCM phenotype prevention experiment, ECTs were treated for 5-weeks with 250 nM mavacamten or a vehicle control. We found that chronic mavacamten treatment of R403Q ECTs: (i) shortened relaxation time, (ii) reduced APD90 prolongation, (iii) upregulated ADRB2, ATP2A2, RYR2, and CACNA1C, (iv) decreased B-type natriuretic peptide (BNP) mRNA and protein expression levels, and (v) increased sarcomere length and reduced sarcomere disarray. Discussion: Taken together, we demonstrated R403Q ECTs generated in the Biowire platform recapitulated many cardiac hypertrophy phenotypes and that chronic mavacamten treatment prevented much of the pathology. This demonstrates that the Biowire ECTs are well-suited to phenotypic-based drug discovery in a human-relevant disease model.
Collapse
Affiliation(s)
- Kai Wang
- Valo Health, Inc., Department of Discovery Research, New York, NY, United States
| | | | | | | | | | | |
Collapse
|
9
|
Pezhouman A, Nguyen NB, Kay M, Kanjilal B, Noshadi I, Ardehali R. Cardiac regeneration - Past advancements, current challenges, and future directions. J Mol Cell Cardiol 2023; 182:75-85. [PMID: 37482238 DOI: 10.1016/j.yjmcc.2023.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
Cardiovascular disease is the leading cause of mortality and morbidity worldwide. Despite improvements in the standard of care for patients with heart diseases, including innovation in pharmacotherapy and surgical interventions, none have yet been proven effective to prevent the progression to heart failure. Cardiac transplantation is the last resort for patients with severe heart failure, but donor shortages remain a roadblock. Cardiac regenerative strategies include cell-based therapeutics, gene therapy, direct reprogramming of non-cardiac cells, acellular biologics, and tissue engineering methods to restore damaged hearts. Significant advancements have been made over the past several decades within each of these fields. This review focuses on the advancements of: 1) cell-based cardiac regenerative therapies, 2) the use of noncoding RNA to induce endogenous cell proliferation, and 3) application of bioengineering methods to promote retention and integration of engrafted cells. Different cell sources have been investigated, including adult stem cells derived from bone marrow and adipose cells, cardiosphere-derived cells, skeletal myoblasts, and pluripotent stem cells. In addition to cell-based transplantation approaches, there have been accumulating interest over the past decade in inducing endogenous CM proliferation for heart regeneration, particularly with the use of noncoding RNAs such as miRNAs and lncRNAs. Bioengineering applications have focused on combining cell-transplantation approaches with fabrication of a porous, vascularized scaffold using biomaterials and advanced bio-fabrication techniques that may offer enhanced retention of transplanted cells, with the hope that these cells would better engraft with host tissue to improve cardiac function. This review summarizes the present status and future challenges of cardiac regenerative therapies.
Collapse
Affiliation(s)
- Arash Pezhouman
- Baylor College of Medicine, Department of Medicine, Division of Cardiology, Houston, Texas 77030, United States; Texas Heart Institute, Houston, Texas 77030, United States
| | - Ngoc B Nguyen
- Baylor College of Medicine, Department of Internal Medicine, Houston, Texas 77030, United States
| | - Maryam Kay
- Department of Medicine, Division of Cardiology, University of California, Los Angeles, CA 90095, United States
| | - Baishali Kanjilal
- Department of Bioengineering, University of California, Riverside, Riverside, CA 92521, United States
| | - Iman Noshadi
- Department of Bioengineering, University of California, Riverside, Riverside, CA 92521, United States
| | - Reza Ardehali
- Baylor College of Medicine, Department of Medicine, Division of Cardiology, Houston, Texas 77030, United States; Texas Heart Institute, Houston, Texas 77030, United States.
| |
Collapse
|
10
|
Cofiño-Fabres C, Passier R, Schwach V. Towards Improved Human In Vitro Models for Cardiac Arrhythmia: Disease Mechanisms, Treatment, and Models of Atrial Fibrillation. Biomedicines 2023; 11:2355. [PMID: 37760796 PMCID: PMC10525681 DOI: 10.3390/biomedicines11092355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/18/2023] [Accepted: 08/19/2023] [Indexed: 09/29/2023] Open
Abstract
Heart rhythm disorders, arrhythmias, place a huge economic burden on society and have a large impact on the quality of life of a vast number of people. Arrhythmias can have genetic causes but primarily arise from heart tissue remodeling during aging or heart disease. As current therapies do not address the causes of arrhythmias but only manage the symptoms, it is of paramount importance to generate innovative test models and platforms for gaining knowledge about the underlying disease mechanisms which are compatible with drug screening. In this review, we outline the most important features of atrial fibrillation (AFib), the most common cardiac arrhythmia. We will discuss the epidemiology, risk factors, underlying causes, and present therapies of AFib, as well as the shortcomings and opportunities of current models for cardiac arrhythmia, including animal models, in silico and in vitro models utilizing human pluripotent stem cell (hPSC)-derived cardiomyocytes.
Collapse
Affiliation(s)
- Carla Cofiño-Fabres
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Drienerlolaan 5, 7500 AE Enschede, The Netherlands;
| | - Robert Passier
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Drienerlolaan 5, 7500 AE Enschede, The Netherlands;
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2300 RC Leiden, The Netherlands
| | - Verena Schwach
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Drienerlolaan 5, 7500 AE Enschede, The Netherlands;
| |
Collapse
|
11
|
Galow AM, Brenmoehl J, Hoeflich A. Synergistic effects of hormones on structural and functional maturation of cardiomyocytes and implications for heart regeneration. Cell Mol Life Sci 2023; 80:240. [PMID: 37541969 PMCID: PMC10403476 DOI: 10.1007/s00018-023-04894-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/18/2023] [Accepted: 07/22/2023] [Indexed: 08/06/2023]
Abstract
The limited endogenous regenerative capacity of the human heart renders cardiovascular diseases a major health threat, thus motivating intense research on in vitro heart cell generation and cell replacement therapies. However, so far, in vitro-generated cardiomyocytes share a rather fetal phenotype, limiting their utility for drug testing and cell-based heart repair. Various strategies to foster cellular maturation provide some success, but fully matured cardiomyocytes are still to be achieved. Today, several hormones are recognized for their effects on cardiomyocyte proliferation, differentiation, and function. Here, we will discuss how the endocrine system impacts cardiomyocyte maturation. After detailing which features characterize a mature phenotype, we will contemplate hormones most promising to induce such a phenotype, the routes of their action, and experimental evidence for their significance in this process. Due to their pleiotropic effects, hormones might be not only valuable to improve in vitro heart cell generation but also beneficial for in vivo heart regeneration. Accordingly, we will also contemplate how the presented hormones might be exploited for hormone-based regenerative therapies.
Collapse
Affiliation(s)
- Anne-Marie Galow
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany.
| | - Julia Brenmoehl
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| | - Andreas Hoeflich
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| |
Collapse
|
12
|
Allan A, Creech J, Hausner C, Krajcarski P, Gunawan B, Poulin N, Kozlowski P, Clark CW, Dow R, Saraithong P, Mair DB, Block T, Monteiro da Rocha A, Kim DH, Herron TJ. High-throughput longitudinal electrophysiology screening of mature chamber-specific hiPSC-CMs using optical mapping. iScience 2023; 26:107142. [PMID: 37416454 PMCID: PMC10320609 DOI: 10.1016/j.isci.2023.107142] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 06/01/2023] [Accepted: 06/12/2023] [Indexed: 07/08/2023] Open
Abstract
hiPSC-CMs are being considered by the Food and Drug Administration and other regulatory agencies for in vitro cardiotoxicity screening to provide human-relevant safety data. Widespread adoption of hiPSC-CMs in regulatory and academic science is limited by the immature, fetal-like phenotype of the cells. Here, to advance the maturation state of hiPSC-CMs, we developed and validated a human perinatal stem cell-derived extracellular matrix coating applied to high-throughput cell culture plates. We also present and validate a cardiac optical mapping device designed for high-throughput functional assessment of mature hiPSC-CM action potentials using voltage-sensitive dye and calcium transients using calcium-sensitive dyes or genetically encoded calcium indicators (GECI, GCaMP6). We utilize the optical mapping device to provide new biological insight into mature chamber-specific hiPSC-CMs, responsiveness to cardioactive drugs, the effect of GCaMP6 genetic variants on electrophysiological function, and the effect of daily β-receptor stimulation on hiPSC-CM monolayer function and SERCA2a expression.
Collapse
Affiliation(s)
- Andrew Allan
- Cairn Research, Graveney Road, Faversham, Kent ME13 8UP UK
| | - Jeffery Creech
- University of Michigan, Frankel Cardiovascular Regeneration Core Laboratory, Ann Arbor, MI 48109, USA
| | - Christian Hausner
- University of Michigan, Frankel Cardiovascular Regeneration Core Laboratory, Ann Arbor, MI 48109, USA
| | - Peyton Krajcarski
- University of Michigan, Frankel Cardiovascular Regeneration Core Laboratory, Ann Arbor, MI 48109, USA
| | - Bianca Gunawan
- University of Michigan, Frankel Cardiovascular Regeneration Core Laboratory, Ann Arbor, MI 48109, USA
| | - Noah Poulin
- University of Michigan, Frankel Cardiovascular Regeneration Core Laboratory, Ann Arbor, MI 48109, USA
| | - Paul Kozlowski
- Michigan Medicine, Internal Medicine-Cardiology, Ann Arbor, MI 48109, USA
| | - Christopher Wayne Clark
- University of Michigan, School of Public Health, Department of Environmental Health Sciences, Ann Arbor, MI 48109, USA
| | - Rachel Dow
- University of Michigan, Frankel Cardiovascular Regeneration Core Laboratory, Ann Arbor, MI 48109, USA
| | - Prakaimuk Saraithong
- University of Michigan, Frankel Cardiovascular Regeneration Core Laboratory, Ann Arbor, MI 48109, USA
- Michigan Medicine, Internal Medicine-Cardiology, Ann Arbor, MI 48109, USA
| | - Devin B. Mair
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Travis Block
- StemBioSys, Inc, 3463 Magic Drive, Suite 110, San Antonio, TX 78229, USA
| | - Andre Monteiro da Rocha
- University of Michigan, Frankel Cardiovascular Regeneration Core Laboratory, Ann Arbor, MI 48109, USA
- Michigan Medicine, Internal Medicine-Cardiology, Ann Arbor, MI 48109, USA
| | - Deok-Ho Kim
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Todd J. Herron
- University of Michigan, Frankel Cardiovascular Regeneration Core Laboratory, Ann Arbor, MI 48109, USA
- Michigan Medicine, Internal Medicine-Cardiology, Ann Arbor, MI 48109, USA
- Michigan Medicine, Molecular & Integrative Physiology, Ann Arbor, MI 48109, USA
| |
Collapse
|
13
|
Patino-Guerrero A, Esmaeili H, Migrino RQ, Nikkhah M. Nanoengineering of gold nanoribbon-embedded isogenic stem cell-derived cardiac organoids. RSC Adv 2023; 13:16985-17000. [PMID: 37288383 PMCID: PMC10243308 DOI: 10.1039/d3ra01811c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 05/29/2023] [Indexed: 06/09/2023] Open
Abstract
Cardiac tissue engineering is an emerging field providing tools to treat and study cardiovascular diseases (CVDs). In the past years, the integration of stem cell technologies with micro- and nanoengineering techniques has enabled the creation of novel engineered cardiac tissues (ECTs) with potential applications in disease modeling, drug screening, and regenerative medicine. However, a major unaddressed limitation of stem cell-derived ECTs is their immature state, resembling a neonatal phenotype and genotype. The modulation of the cellular microenvironment within the ECTs has been proposed as an efficient mechanism to promote cellular maturation and improve features such as cellular coupling and synchronization. The integration of biological and nanoscale cues in the ECTs could serve as a tool for the modification and control of the engineered tissue microenvironment. Here we present a proof-of-concept study for the integration of biofunctionalized gold nanoribbons (AuNRs) with hiPSC-derived isogenic cardiac organoids to enhance tissue function and maturation. We first present extensive characterization of the synthesized AuNRs, their PEGylation and cytotoxicity evaluation. We then evaluated the functional contractility and transcriptomic profile of cardiac organoids fabricated with hiPSC-derived cardiomyocytes (mono-culture) as well as with hiPSC-derived cardiomyocytes and cardiac fibroblasts (co-culture). We demonstrated that PEGylated AuNRs are biocompatible and do not induce cell death in hiPSC-derived cardiac cells and organoids. We also found an improved transcriptomic profile of the co-cultured organoids indicating maturation of the hiPSC-derived cardiomyocytes in the presence of cardiac fibroblasts. Overall, we present for the first time the integration of AuNRs into cardiac organoids, showing promising results for improved tissue function.
Collapse
Affiliation(s)
| | - Hamid Esmaeili
- School of Biological and Health Systems Engineering, Arizona State University Tempe AZ 8528 USA
| | - Raymond Q Migrino
- Phoenix Veterans Affairs Health Care System Phoenix AZ 85012 USA
- University of Arizona College of Medicine Phoenix AZ 85004 USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering, Arizona State University Tempe AZ 8528 USA
- Center for Personalized Diagnostics Biodesign Institute, Arizona State University Tempe AZ 85281 USA
| |
Collapse
|
14
|
Kumar A, He S, Mali P. Systematic discovery of transcription factors that improve hPSC-derived cardiomyocyte maturation via temporal analysis of bioengineered cardiac tissues. APL Bioeng 2023; 7:026109. [PMID: 37252678 PMCID: PMC10219684 DOI: 10.1063/5.0137458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 05/09/2023] [Indexed: 05/31/2023] Open
Abstract
Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have the potential to become powerful tools for disease modeling, drug testing, and transplantation; however, their immaturity limits their applications. Transcription factor (TF) overexpression can improve hPSC-CM maturity, but identifying these TFs has been elusive. Toward this, we establish here an experimental framework for systematic identification of maturation enhancing factors. Specifically, we performed temporal transcriptome RNAseq analyses of progressively matured hPSC-derived cardiomyocytes across 2D and 3D differentiation systems and further compared these bioengineered tissues to native fetal and adult-derived tissues. These analyses revealed 22 TFs whose expression did not increase in 2D differentiation systems but progressively increased in 3D culture systems and adult mature cell types. Individually overexpressing each of these TFs in immature hPSC-CMs identified five TFs (KLF15, ZBTB20, ESRRA, HOPX, and CAMTA2) as regulators of calcium handling, metabolic function, and hypertrophy. Notably, the combinatorial overexpression of KLF15, ESRRA, and HOPX improved all three maturation parameters simultaneously. Taken together, we introduce a new TF cocktail that can be used in solo or in conjunction with other strategies to improve hPSC-CM maturation and anticipate that our generalizable methodology can also be implemented to identify maturation-associated TFs for other stem cell progenies.
Collapse
Affiliation(s)
- Aditya Kumar
- Department of Bioengineering, University of California, San Diego, California 92093, USA
| | - Starry He
- Department of Bioengineering, University of California, San Diego, California 92093, USA
| | - Prashant Mali
- Department of Bioengineering, University of California, San Diego, California 92093, USA
| |
Collapse
|
15
|
Campostrini G, Kosmidis G, Ward-van Oostwaard D, Davis RP, Yiangou L, Ottaviani D, Veerman CC, Mei H, Orlova VV, Wilde AAM, Bezzina CR, Verkerk AO, Mummery CL, Bellin M. Maturation of hiPSC-derived cardiomyocytes promotes adult alternative splicing of SCN5A and reveals changes in sodium current associated with cardiac arrhythmia. Cardiovasc Res 2023; 119:167-182. [PMID: 35394010 PMCID: PMC10022870 DOI: 10.1093/cvr/cvac059] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/07/2022] [Accepted: 03/23/2022] [Indexed: 11/13/2022] Open
Abstract
AIMS Human-induced pluripotent stem cell-cardiomyocytes (hiPSC-CMs) are widely used to study arrhythmia-associated mutations in ion channels. Among these, the cardiac sodium channel SCN5A undergoes foetal-to-adult isoform switching around birth. Conventional hiPSC-CM cultures, which are phenotypically foetal, have thus far been unable to capture mutations in adult gene isoforms. Here, we investigated whether tri-cellular cross-talk in a three-dimensional (3D) cardiac microtissue (MT) promoted post-natal SCN5A maturation in hiPSC-CMs. METHODS AND RESULTS We derived patient hiPSC-CMs carrying compound mutations in the adult SCN5A exon 6B and exon 4. Electrophysiological properties of patient hiPSC-CMs in monolayer were not altered by the exon 6B mutation compared with isogenic controls since it is not expressed; further, CRISPR/Cas9-mediated excision of the foetal exon 6A did not promote adult SCN5A expression. However, when hiPSC-CMs were matured in 3D cardiac MTs, SCN5A underwent isoform switch and the functional consequences of the mutation located in exon 6B were revealed. Up-regulation of the splicing factor muscleblind-like protein 1 (MBNL1) drove SCN5A post-natal maturation in microtissues since its overexpression in hiPSC-CMs was sufficient to promote exon 6B inclusion, whilst knocking-out MBNL1 failed to foster isoform switch. CONCLUSIONS Our study shows that (i) the tri-cellular cardiac microtissues promote post-natal SCN5A isoform switch in hiPSC-CMs, (ii) adult splicing of SCN5A is driven by MBNL1 in these tissues, and (iii) this model can be used for examining post-natal cardiac arrhythmias due to mutations in the exon 6B. TRANSLATIONAL PERSPECTIVE The cardiac sodium channel is essential for conducting the electrical impulse in the heart. Postnatal alternative splicing regulation causes mutual exclusive inclusion of fetal or adult exons of the corresponding gene, SCN5A. Typically, immature hiPSCCMs fall short in studying the effect of mutations located in the adult exon. We describe here that an innovative tri-cellular three-dimensional cardiac microtissue culture promotes hiPSC-CMs maturation through upregulation of MBNL1, thus revealing the effect of a pathogenic genetic variant located in the SCN5A adult exon. These results help advancing the use of hiPSC-CMs in studying adult heart disease and for developing personalized medicine applications.
Collapse
Affiliation(s)
- Giulia Campostrini
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Georgios Kosmidis
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Dorien Ward-van Oostwaard
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Richard Paul Davis
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Loukia Yiangou
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Daniele Ottaviani
- Department of Biology, University of Padua, 35121 Padua, Italy
- Veneto Institute of Molecular Medicine, 35129 Padua, Italy
| | - Christiaan Cornelis Veerman
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam University Medical Centre, location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Leiden University Medical Center, 2333 Leiden, The Netherlands
| | - Valeria Viktorovna Orlova
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Arthur Arnold Maria Wilde
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam University Medical Centre, location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Connie Rose Bezzina
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam University Medical Centre, location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Arie Otto Verkerk
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam University Medical Centre, location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Christine Lindsay Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
- Department of Applied Stem Cell Technologies, University of Twente, 7500 AE, Enschede, The Netherlands
| | | |
Collapse
|
16
|
Patino-Guerrero A, Ponce Wong RD, Kodibagkar VD, Zhu W, Migrino RQ, Graudejus O, Nikkhah M. Development and Characterization of Isogenic Cardiac Organoids from Human-Induced Pluripotent Stem Cells Under Supplement Starvation Regimen. ACS Biomater Sci Eng 2023; 9:944-958. [PMID: 36583992 DOI: 10.1021/acsbiomaterials.2c01290] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The prevalence of cardiovascular risk factors is expected to increase the occurrence of cardiovascular diseases (CVDs) worldwide. Cardiac organoids are promising candidates for bridging the gap between in vitro experimentation and translational applications in drug development and cardiac repair due to their attractive features. Here we present the fabrication and characterization of isogenic scaffold-free cardiac organoids derived from human induced pluripotent stem cells (hiPSCs) formed under a supplement-deprivation regimen that allows for metabolic synchronization and maturation of hiPSC-derived cardiac cells. We propose the formation of coculture cardiac organoids that include hiPSC-derived cardiomyocytes and hiPSC-derived cardiac fibroblasts (hiPSC-CMs and hiPSC-CFs, respectively). The cardiac organoids were characterized through extensive morphological assessment, evaluation of cellular ultrastructures, and analysis of transcriptomic and electrophysiological profiles. The morphology and transcriptomic profile of the organoids were improved by coculture of hiPSC-CMs with hiPSC-CFs. Specifically, upregulation of Ca2+ handling-related genes, such as RYR2 and SERCA, and structure-related genes, such as TNNT2 and MYH6, was observed. Additionally, the electrophysiological characterization of the organoids under supplement deprivation shows a trend for reduced conduction velocity for coculture organoids. These studies help us gain a better understanding of the role of other isogenic cells such as hiPSC-CFs in the formation of mature cardiac organoids, along with the introduction of exogenous chemical cues, such as supplement starvation.
Collapse
Affiliation(s)
- Alejandra Patino-Guerrero
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona8528, United States
| | | | - Vikram D Kodibagkar
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona8528, United States
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Mayo Clinic Arizona, Scottsdale, Arizona85259, United States
| | - Raymond Q Migrino
- Phoenix Veterans Affairs Health Care System, Phoenix, Arizona85012, United States.,University of Arizona College of Medicine, Phoenix, Arizona85004, United States
| | - Oliver Graudejus
- BMSEED, Mesa, Arizona85201, United States.,School of Molecular Sciences, Arizona State University, Tempe, Arizona85287, United States
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona8528, United States.,Center for Personalized Diagnostics Biodesign Institute, Arizona State University, Tempe, Arizona85281, United States
| |
Collapse
|
17
|
The Exciting Realities and Possibilities of iPS-Derived Cardiomyocytes. Bioengineering (Basel) 2023; 10:bioengineering10020237. [PMID: 36829731 PMCID: PMC9952364 DOI: 10.3390/bioengineering10020237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/03/2023] [Accepted: 02/09/2023] [Indexed: 02/12/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) have become a prevalent topic after their discovery, advertised as an ethical alternative to embryonic stem cells (ESCs). Due to their ability to differentiate into several kinds of cells, including cardiomyocytes, researchers quickly realized the potential for differentiated cardiomyocytes to be used in the treatment of heart failure, a research area with few alternatives. This paper discusses the differentiation process for human iPSC-derived cardiomyocytes and the possible applications of said cells while answering some questions regarding ethical issues.
Collapse
|
18
|
Wu P, Sai X, Li Z, Ye X, Jin L, Liu G, Li G, Yang P, Zhao M, Zhu S, Liu N, Zhu P. Maturation of induced pluripotent stem cell-derived cardiomyocytes and its therapeutic effect on myocardial infarction in mouse. Bioact Mater 2023; 20:286-305. [PMID: 35702609 PMCID: PMC9167678 DOI: 10.1016/j.bioactmat.2022.05.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 11/30/2022] Open
Abstract
Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) have an irreplaceable role in the treatment of myocardial infarction (MI), which can be injected into the transplanted area with new cardiomyocytes (Cardiomyocytes, CMs), and improve myocardial function. However, the immaturity of the structure and function of iPSC-CMs is the main bottleneck at present. Since collagen participates in the formation of extracellular matrix (ECM), we synthesized nano colloidal gelatin (Gel) with collagen as the main component, and confirmed that the biomaterial has good biocompatibility and is suitable for cellular in vitro growth. Subsequently, we combined the PI3K/AKT/mTOR pathway inhibitor BEZ-235 with Gel and found that the two combined increased the sarcomere length and action potential amplitude (APA) of iPSC-CMs, and improved the Ca2+ processing ability, the maturation of mitochondrial morphological structure and metabolic function. Not only that, Gel can also prolong the retention rate of iPSC-CMs in the myocardium and increase the expression of Cx43 and angiogenesis in the transplanted area of mature iPSC-CMs, which also provides a reliable basis for the subsequent treatment of mature iPSC-CMs. BEZ-235 + Gel promotes the maturation of sarcomere structure in iPSC-CMs. BEZ-235 + Gel promotes electrophysiological maturation of iPSC-CMs. BEZ-235 + Gel increases mitochondrial respiration in iPSC-CMs. Gel loaded with mature iPSC-CMs enhanced angiogenesis and gap junction formation at the injection site.
Collapse
|
19
|
Ernst P, Bidwell PA, Dora M, Thomas DD, Kamdar F. Cardiac calcium regulation in human induced pluripotent stem cell cardiomyocytes: Implications for disease modeling and maturation. Front Cell Dev Biol 2023; 10:986107. [PMID: 36742199 PMCID: PMC9889838 DOI: 10.3389/fcell.2022.986107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 12/23/2022] [Indexed: 01/19/2023] Open
Abstract
Human induced pluripotent stem cell cardiomyocytes (hiPSC-CMs) are based on ground-breaking technology that has significantly impacted cardiovascular research. They provide a renewable source of human cardiomyocytes for a variety of applications including in vitro disease modeling and drug toxicity testing. Cardiac calcium regulation plays a critical role in the cardiomyocyte and is often dysregulated in cardiovascular disease. Due to the limited availability of human cardiac tissue, calcium handling and its regulation have most commonly been studied in the context of animal models. hiPSC-CMs can provide unique insights into human physiology and pathophysiology, although a remaining limitation is the relative immaturity of these cells compared to adult cardiomyocytes Therefore, this field is rapidly developing techniques to improve the maturity of hiPSC-CMs, further establishing their place in cardiovascular research. This review briefly covers the basics of cardiomyocyte calcium cycling and hiPSC technology, and will provide a detailed description of our current understanding of calcium in hiPSC-CMs.
Collapse
Affiliation(s)
- Patrick Ernst
- Cardiovascular Division, University of Minnesota, Minneapolis, MN, United States
| | - Philip A. Bidwell
- Cardiovascular Division, University of Minnesota, Minneapolis, MN, United States
| | - Michaela Dora
- College of Biological Sciences, University of Minnesota, Minneapolis, MN, United States
| | - David D. Thomas
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - Forum Kamdar
- Cardiovascular Division, University of Minnesota, Minneapolis, MN, United States,*Correspondence: Forum Kamdar,
| |
Collapse
|
20
|
Abstract
Long QT syndrome (LQTS) is a detrimental arrhythmia syndrome mainly caused by dysregulated expression or aberrant function of ion channels. The major clinical symptoms of ventricular arrhythmia, palpitations and syncope vary among LQTS subtypes. Susceptibility to malignant arrhythmia is a result of delayed repolarisation of the cardiomyocyte action potential (AP). There are 17 distinct subtypes of LQTS linked to 15 autosomal dominant genes with monogenic mutations. However, due to the presence of modifier genes, the identical mutation may result in completely different clinical manifestations in different carriers. In this review, we describe the roles of various ion channels in orchestrating APs and discuss molecular aetiologies of various types of LQTS. We highlight the usage of patient-specific induced pluripotent stem cell (iPSC) models in characterising fundamental mechanisms associated with LQTS. To mitigate the outcomes of LQTS, treatment strategies are initially focused on small molecules targeting ion channel activities. Next-generation treatments will reap the benefits from development of LQTS patient-specific iPSC platform, which is bolstered by the state-of-the-art technologies including whole-genome sequencing, CRISPR genome editing and machine learning. Deep phenotyping and high-throughput drug testing using LQTS patient-specific cardiomyocytes herald the upcoming precision medicine in LQTS.
Collapse
|
21
|
Martyniak A, Jeż M, Dulak J, Stępniewski J. Adaptation of cardiomyogenesis to the generation and maturation of cardiomyocytes from human pluripotent stem cells. IUBMB Life 2023; 75:8-29. [PMID: 36263833 DOI: 10.1002/iub.2685] [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: 06/08/2022] [Accepted: 10/05/2022] [Indexed: 12/29/2022]
Abstract
The advent of methods for efficient generation and cardiac differentiation of pluripotent stem cells opened new avenues for disease modelling, drug testing, and cell therapies of the heart. However, cardiomyocytes (CM) obtained from such cells demonstrate an immature, foetal-like phenotype that involves spontaneous contractions, irregular morphology, expression of embryonic isoforms of sarcomere components, and low level of ion channels. These and other features may affect cellular response to putative therapeutic compounds and the efficient integration into the host myocardium after in vivo delivery. Therefore, novel strategies to increase the maturity of pluripotent stem cell-derived CM are of utmost importance. Several approaches have already been developed relying on molecular changes that occur during foetal and postnatal maturation of the heart, its electromechanical activity, and the cellular composition. As a better understanding of these determinants may facilitate the generation of efficient protocols for in vitro acquisition of an adult-like phenotype by immature CM, this review summarizes the most important molecular factors that govern CM during embryonic development, postnatal changes that trigger heart maturation, as well as protocols that are currently used to generate mature pluripotent stem cell-derived cardiomyocytes.
Collapse
Affiliation(s)
- Alicja Martyniak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Mateusz Jeż
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Jacek Stępniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
22
|
Chirico N, Kessler EL, Maas RGC, Fang J, Qin J, Dokter I, Daniels M, Šarić T, Neef K, Buikema JW, Lei Z, Doevendans PA, Sluijter JPG, van Mil A. Small molecule-mediated rapid maturation of human induced pluripotent stem cell-derived cardiomyocytes. Stem Cell Res Ther 2022; 13:531. [PMID: 36575473 PMCID: PMC9795728 DOI: 10.1186/s13287-022-03209-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 12/01/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (iPSC-CMs) do not display all hallmarks of mature primary cardiomyocytes, especially the ability to use fatty acids (FA) as an energy source, containing high mitochondrial mass, presenting binucleation and increased DNA content per nuclei (polyploidism), and synchronized electrical conduction. This immaturity represents a bottleneck to their application in (1) disease modelling-as most cardiac (genetic) diseases have a middle-age onset-and (2) clinically relevant models, where integration and functional coupling are key. So far, several methods have been reported to enhance iPSC-CM maturation; however, these protocols are laborious, costly, and not easily scalable. Therefore, we developed a simple, low-cost, and rapid protocol to promote cardiomyocyte maturation using two small molecule activators of the peroxisome proliferator-activated receptor β/δ and gamma coactivator 1-alpha (PPAR/PGC-1α) pathway: asiatic acid (AA) and GW501516 (GW). METHODS AND RESULTS: Monolayers of iPSC-CMs were incubated with AA or GW every other day for ten days resulting in increased expression of FA metabolism-related genes and markers for mitochondrial activity. AA-treated iPSC-CMs responsiveness to the mitochondrial respiratory chain inhibitors increased and exhibited higher flexibility in substrate utilization. Additionally, structural maturity improved after treatment as demonstrated by an increase in mRNA expression of sarcomeric-related genes and higher nuclear polyploidy in AA-treated samples. Furthermore, treatment led to increased ion channel gene expression and protein levels. CONCLUSIONS Collectively, we developed a fast, easy, and economical method to induce iPSC-CMs maturation via PPAR/PGC-1α activation. Treatment with AA or GW led to increased metabolic, structural, functional, and electrophysiological maturation, evaluated using a multiparametric quality assessment.
Collapse
Affiliation(s)
- Nino Chirico
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Elise L. Kessler
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Renée G. C. Maas
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Juntao Fang
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jiabin Qin
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Inge Dokter
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mark Daniels
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tomo Šarić
- grid.6190.e0000 0000 8580 3777Center for Physiology and Pathophysiology, Institute for Neurophysiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Klaus Neef
- grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.491096.3Department of Cardiology, Amsterdam Medical Centre, 1105 AZ Amsterdam, The Netherlands
| | - Jan-Willem Buikema
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Zhiyong Lei
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Pieter A. Doevendans
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.411737.7Netherlands Heart Institute, Utrecht, The Netherlands
| | - Joost P. G. Sluijter
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Alain van Mil
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
23
|
Mostert D, Groenen B, Klouda L, Passier R, Goumans MJ, Kurniawan NA, Bouten CVC. Human pluripotent stem cell-derived cardiomyocytes align under cyclic strain when guided by cardiac fibroblasts. APL Bioeng 2022; 6:046108. [PMID: 36567768 PMCID: PMC9771596 DOI: 10.1063/5.0108914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022] Open
Abstract
The myocardium is a mechanically active tissue typified by anisotropy of the resident cells [cardiomyocytes (CMs) and cardiac fibroblasts (cFBs)] and the extracellular matrix (ECM). Upon ischemic injury, the anisotropic tissue is replaced by disorganized scar tissue, resulting in loss of coordinated contraction. Efforts to re-establish tissue anisotropy in the injured myocardium are hampered by a lack of understanding of how CM and/or cFB structural organization is affected by the two major physical cues inherent in the myocardium: ECM organization and cyclic mechanical strain. Herein, we investigate the singular and combined effect of ECM (dis)organization and cyclic strain in a two-dimensional human in vitro co-culture model of the myocardial microenvironment. We show that (an)isotropic ECM protein patterning can guide the orientation of CMs and cFBs, both in mono- and co-culture. Subsequent application of uniaxial cyclic strain-mimicking the local anisotropic deformation of beating myocardium-causes no effect when applied parallel to the anisotropic ECM. However, when cultured on isotropic substrates, cFBs, but not CMs, orient away from the direction of cyclic uniaxial strain (strain avoidance). In contrast, CMs show strain avoidance via active remodeling of their sarcomeres only when co-cultured with at least 30% cFBs. Paracrine signaling or N-cadherin-mediated communication between CMs and cFBs was no contributing factor. Our findings suggest that the mechanoresponsive cFBs provide structural guidance for CM orientation and elongation. Our study, therefore, highlights a synergistic mechanobiological interplay between CMs and cFBs in shaping tissue organization, which is of relevance for regenerating functionally organized myocardium.
Collapse
Affiliation(s)
| | - Bart Groenen
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| | - Leda Klouda
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| | | | - Marie-Jose Goumans
- Department of Cell and Chemical Biology and Center for Biomedical Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | | | | |
Collapse
|
24
|
Verkerk AO, Doszpod IJ, Mengarelli I, Magyar T, Polyák A, Pászti B, Efimov IR, Wilders R, Koncz I. Acetylcholine Reduces L-Type Calcium Current without Major Changes in Repolarization of Canine and Human Purkinje and Ventricular Tissue. Biomedicines 2022; 10:biomedicines10112987. [PMID: 36428555 PMCID: PMC9687254 DOI: 10.3390/biomedicines10112987] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/11/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
Vagal nerve stimulation (VNS) holds a strong basis as a potentially effective treatment modality for chronic heart failure, which explains why a multicenter VNS study in heart failure with reduced ejection fraction is ongoing. However, more detailed information is required on the effect of acetylcholine (ACh) on repolarization in Purkinje and ventricular cardiac preparations to identify the advantages, risks, and underlying cellular mechanisms of VNS. Here, we studied the effect of ACh on the action potential (AP) of canine Purkinje fibers (PFs) and several human ventricular preparations. In addition, we characterized the effects of ACh on the L-type Ca2+ current (ICaL) and AP of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and performed computer simulations to explain the observed effects. Using microelectrode recordings, we found a small but significant AP prolongation in canine PFs. In the human myocardium, ACh slightly prolonged the AP in the midmyocardium but resulted in minor AP shortening in subepicardial tissue. Perforated patch-clamp experiments on hiPSC-CMs demonstrated that 5 µM ACh caused an ≈15% decrease in ICaL density without changes in gating properties. Using dynamic clamp, we found that under blocked K+ currents, 5 µM ACh resulted in an ≈23% decrease in AP duration at 90% of repolarization in hiPSC-CMs. Computer simulations using the O'Hara-Rudy human ventricular cell model revealed that the overall effect of ACh on AP duration is a tight interplay between the ACh-induced reduction in ICaL and ACh-induced changes in K+ currents. In conclusion, ACh results in minor changes in AP repolarization and duration of canine PFs and human ventricular myocardium due to the concomitant inhibition of inward ICaL and outward K+ currents, which limits changes in net repolarizing current and thus prevents major changes in AP repolarization.
Collapse
Affiliation(s)
- Arie O. Verkerk
- Department of Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Illés J. Doszpod
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary
| | - Isabella Mengarelli
- Department of Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Tibor Magyar
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary
| | - Alexandra Polyák
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary
| | - Bence Pászti
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary
| | - Igor R. Efimov
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA
- Department of Biomedical Engineering, Northwestern University, Chicago, IL 60611, USA
- Department of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Correspondence: (R.W.); (I.K.)
| | - István Koncz
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA
- Correspondence: (R.W.); (I.K.)
| |
Collapse
|
25
|
Minter-Dykhouse K, Nelson TJ, Folmes CD. Uncoupling of Proliferative Capacity from Developmental Stage During Directed Cardiac Differentiation of Pluripotent Stem Cells. Stem Cells Dev 2022; 31:521-528. [PMID: 35726436 PMCID: PMC9641990 DOI: 10.1089/scd.2022.0041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 06/17/2022] [Indexed: 11/13/2022] Open
Abstract
Lineage-specific differentiation of human-induced pluripotent stem cells (hiPSCs) into cardiomyocytes (CMs) offers a patient-specific model to dissect development and disease pathogenesis in a dish. However, challenges exist with this model system, such as the relative immaturity of iPSC-derived CMs, which evoke the question of whether this model faithfully recapitulates in vivo cardiac development. As in vivo cardiac developmental stage is intimately linked with the proliferative capacity (or maturation is inversely correlated to proliferative capacity), we sought to understand how proliferation is regulated during hiPSC CM differentiation and how it compares with in vivo mouse cardiac development. Using standard Chemically Defined Media 3 differentiation, gene expression profiles demonstrate that hiPSC-derived cardiomyocytes (hiPSC-CMs) do not progress past the equivalent of embryonic day 14.5 of murine cardiac development. Throughout differentiation, overall DNA synthesis rapidly declines with <5% of hiPSC-CMs actively synthesizing DNA at the end of the differentiation period despite their immaturity. Bivariate cell cycle analysis demonstrated that hiPSC-CMs have a cell cycle profile distinct from their non-cardiac counterparts from the same differentiation, with significantly fewer cells within G1 and a marked accumulation of cells in G2/M than their non-cardiac counterparts throughout differentiation. Pulse-chase analysis demonstrated that non-cardiac cells progressed completely through the cell cycle within a 24-h period, whereas hiPSC-CMs had restricted progression with only a small proportion of cells undergoing cytokinesis with the remainder stalling in late S-phase or G2/M. This cell cycle arrest phenotype is associated with abbreviated expression of cell cycle promoting genes compared with expression throughout murine embryonic cardiac development. In summary, directed differentiation of hiPSCs into CMs uncouples the developmental stage from cell cycle regulation compared with in vivo mouse cardiac development, leading to a premature exit of hiPSC-CMs from the cell cycle despite their relative immaturity.
Collapse
Affiliation(s)
- Katherine Minter-Dykhouse
- Stem Cell and Regenerative Metabolism Laboratory, Departments of Cardiovascular Diseases, Biochemistry and Molecular Biology, and Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona, USA
| | - Timothy J. Nelson
- Todd and Karen Wanek Family Program for Hypoplastic Left Heart Syndrome, Departments of General Internal Medicine and Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Clifford D.L. Folmes
- Address correspondence to: Clifford D. L. Folmes, PhD, Stem Cell and Regenerative Metabolism Laboratory, Departments of Cardiovascular Diseases, Biochemistry and Molecular Biology, Mayo Clinic, 13400 E Shea Boulevard, Scottsdale, AZ 85259, USA
| |
Collapse
|
26
|
Zhang M, Xu Y, Chen Y, Yan Q, Li X, Ding L, Wei T, Zeng D. Three-Dimensional Poly-(ε-Caprolactone) Nanofibrous Scaffolds Promote the Maturation of Human Pluripotent Stem Cells-Induced Cardiomyocytes. Front Cell Dev Biol 2022; 10:875278. [PMID: 35979378 PMCID: PMC9377449 DOI: 10.3389/fcell.2022.875278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/25/2022] [Indexed: 11/30/2022] Open
Abstract
Although pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) have been proved to be a new platform for heart regeneration, the lack of maturity significantly hinders the clinic application. Recent researches indicate that the function of stem cell is associated with the nanoscale geometry/topography of the extracellular matrix (ECM). However, the effects of 3D nanofibrous scaffolds in maturation of iPSC-CMs still remain unclear. Thus, we explored the effects of restructuring iPSC-CMs in 3D nano-scaffolds on cell morphology, cardiac-specific structural protein, gap junction and calcium transient kinetics. Using the electrospinning technology, poly-(ε-caprolactone) (PCL) nanofibrous scaffold were constructed and iPSC-CMs were seeded into these forms. As expected, strong sarcolemmal remodeling processes and myofilament reorientation were observed in 3D nano-scaffolds culture, as well as more expression of cardiac mature proteins, such as β-MHC and MLC2v. The mature morphology of 3D-shaped iPSC-CMs leaded to enhanced calcium transient kinetics, with increased calcium peak transient amplitude and the maximum upstroke velocity (Vmax). The results revealed that the maturation of iPSC-CMs was enhanced by the electrospun 3D PCL nanofibrous scaffolds treatment. These findings also proposed a feasible strategy to improve the myocardium bioengineering by combining stem cells with scaffolds.
Collapse
Affiliation(s)
- Mingming Zhang
- Department of Cardiology, Tangdu Hospital, the Fourth Military Medical University, Xi’an, China
| | - Yuerong Xu
- Department of Orthodontics, School of Stomatology, the Fourth Military Medical University, Xi’an, China
| | - Yan Chen
- Department of Cardiology, 971th Hospital, Chinese People’s Liberation Army Navy, Qingdao, China
| | - Qinru Yan
- Department of Neurological Rehabilitation, Xi ‘an International Medical Center Hospital, Xi’an, China
| | - Xiaoli Li
- Department of Cardiology, Tangdu Hospital, the Fourth Military Medical University, Xi’an, China
| | - Lu Ding
- Department of Cardiology, Tangdu Hospital, the Fourth Military Medical University, Xi’an, China
| | - Ting Wei
- Department of Cardiology, Tangdu Hospital, the Fourth Military Medical University, Xi’an, China
| | - Di Zeng
- Department of Cardiology, Tangdu Hospital, the Fourth Military Medical University, Xi’an, China
- *Correspondence: Di Zeng,
| |
Collapse
|
27
|
Sun Q, Yang X, Guo J, Zhao Y, Liu Y. CIEGAN: A Deep Learning Tool for Cell Image Enhancement. Front Genet 2022; 13:913372. [PMID: 35873483 PMCID: PMC9298179 DOI: 10.3389/fgene.2022.913372] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/25/2022] [Indexed: 11/24/2022] Open
Abstract
Long-term live-cell imaging technology has emerged in the study of cell culture and development, and it is expected to elucidate the differentiation or reprogramming morphology of cells and the dynamic process of interaction between cells. There are some advantages to this technique: it is noninvasive, high-throughput, low-cost, and it can help researchers explore phenomena that are otherwise difficult to observe. Many challenges arise in the real-time process, for example, low-quality micrographs are often obtained due to unavoidable human factors or technical factors in the long-term experimental period. Moreover, some core dynamics in the developmental process are rare and fleeting in imaging observation and difficult to recapture again. Therefore, this study proposes a deep learning method for microscope cell image enhancement to reconstruct sharp images. We combine generative adversarial nets and various loss functions to make blurry images sharp again, which is much more convenient for researchers to carry out further analysis. This technology can not only make up the blurry images of critical moments of the development process through image enhancement but also allows long-term live-cell imaging to find a balance between imaging speed and image quality. Furthermore, the scalability of this technology makes the methods perform well in fluorescence image enhancement. Finally, the method is tested in long-term live-cell imaging of human-induced pluripotent stem cell-derived cardiomyocyte differentiation experiments, and it can greatly improve the image space resolution ratio.
Collapse
Affiliation(s)
- Qiushi Sun
- Beijing Key Lab of Traffic Data Analysis and Mining, School of Computer and Information Technology, Beijing Jiaotong University, Beijing, China
| | - Xiaochun Yang
- State Key Laboratory of Natural and Biomimetic Drugs, MOE Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Jingtao Guo
- Beijing Key Lab of Traffic Data Analysis and Mining, School of Computer and Information Technology, Beijing Jiaotong University, Beijing, China
| | - Yang Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, MOE Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- *Correspondence: Yang Zhao, ; Yi Liu,
| | - Yi Liu
- Beijing Key Lab of Traffic Data Analysis and Mining, School of Computer and Information Technology, Beijing Jiaotong University, Beijing, China
- *Correspondence: Yang Zhao, ; Yi Liu,
| |
Collapse
|
28
|
Portero V, Nicol T, Podliesna S, Marchal GA, Baartscheer A, Casini S, Tadros R, Treur JL, Tanck MWT, Cox IJ, Probert F, Hough TA, Falcone S, Beekman L, Müller-Nurasyid M, Kastenmüller G, Gieger C, Peters A, Kääb S, Sinner MF, Blease A, Verkerk AO, Bezzina CR, Potter PK, Remme CA. Chronically elevated branched chain amino acid levels are pro-arrhythmic. Cardiovasc Res 2022; 118:1742-1757. [PMID: 34142125 PMCID: PMC9215196 DOI: 10.1093/cvr/cvab207] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/16/2021] [Indexed: 01/03/2023] Open
Abstract
AIMS Cardiac arrhythmias comprise a major health and economic burden and are associated with significant morbidity and mortality, including cardiac failure, stroke, and sudden cardiac death (SCD). Development of efficient preventive and therapeutic strategies is hampered by incomplete knowledge of disease mechanisms and pathways. Our aim is to identify novel mechanisms underlying cardiac arrhythmia and SCD using an unbiased approach. METHODS AND RESULTS We employed a phenotype-driven N-ethyl-N-nitrosourea mutagenesis screen and identified a mouse line with a high incidence of sudden death at young age (6-9 weeks) in the absence of prior symptoms. Affected mice were found to be homozygous for the nonsense mutation Bcat2p.Q300*/p.Q300* in the Bcat2 gene encoding branched chain amino acid transaminase 2. At the age of 4-5 weeks, Bcat2p.Q300*/p.Q300* mice displayed drastic increase of plasma levels of branch chain amino acids (BCAAs-leucine, isoleucine, valine) due to the incomplete catabolism of BCAAs, in addition to inducible arrhythmias ex vivo as well as cardiac conduction and repolarization disturbances. In line with these findings, plasma BCAA levels were positively correlated to electrocardiogram indices of conduction and repolarization in the German community-based KORA F4 Study. Isolated cardiomyocytes from Bcat2p.Q300*/p.Q300* mice revealed action potential (AP) prolongation, pro-arrhythmic events (early and late afterdepolarizations, triggered APs), and dysregulated calcium homeostasis. Incubation of human pluripotent stem cell-derived cardiomyocytes with elevated concentration of BCAAs induced similar calcium dysregulation and pro-arrhythmic events which were prevented by rapamycin, demonstrating the crucial involvement of mTOR pathway activation. CONCLUSIONS Our findings identify for the first time a causative link between elevated BCAAs and arrhythmia, which has implications for arrhythmogenesis in conditions associated with BCAA metabolism dysregulation such as diabetes, metabolic syndrome, and heart failure.
Collapse
Affiliation(s)
- Vincent Portero
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Thomas Nicol
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell, Oxfordshire, UK
| | - Svitlana Podliesna
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Gerard A Marchal
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Antonius Baartscheer
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Simona Casini
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Rafik Tadros
- Cardiovascular Genetics Center, Montreal Heart Institute and Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Jorien L Treur
- Department of Psychiatry, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | - Michael W T Tanck
- Amsterdam UMC, University of Amsterdam, Department of Epidemiology and Data Science, Amsterdam Public Health (APH), The Netherlands
| | - I Jane Cox
- Institute of Hepatology London, Foundation for Liver Research, London, UK
- Faculty of Life Sciences & Medicine, Kings College, London, UK
| | - Fay Probert
- Department of Chemistry, University of Oxford, Oxford, UK
| | - Tertius A Hough
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell, Oxfordshire, UK
| | - Sara Falcone
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell, Oxfordshire, UK
| | - Leander Beekman
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Martina Müller-Nurasyid
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- IBE, Faculty of Medicine, Ludwig Maximilian’s University (LMU) Munich, Munich, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site: Munich Heart Alliance, Munich, Germany
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site: Munich Heart Alliance, Munich, Germany
| | - Stefan Kääb
- German Centre for Cardiovascular Research (DZHK), Partner Site: Munich Heart Alliance, Munich, Germany
- Department of Medicine I (Cardiology), University Hospital, LMU Munich, Munich, Germany
| | - Moritz F Sinner
- German Centre for Cardiovascular Research (DZHK), Partner Site: Munich Heart Alliance, Munich, Germany
- Department of Medicine I (Cardiology), University Hospital, LMU Munich, Munich, Germany
| | - Andrew Blease
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell, Oxfordshire, UK
| | - Arie O Verkerk
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Connie R Bezzina
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Paul K Potter
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, UK
| | - Carol Ann Remme
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| |
Collapse
|
29
|
Quality criteria for in vitro human pluripotent stem cell-derived models of tissue-based cells. Reprod Toxicol 2022; 112:36-50. [PMID: 35697279 DOI: 10.1016/j.reprotox.2022.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/27/2022] [Accepted: 06/07/2022] [Indexed: 12/21/2022]
Abstract
The advent of the technology to isolate or generate human pluripotent stem cells provided the potential to develop a wide range of human models that could enhance understanding of mechanisms underlying human development and disease. These systems are now beginning to mature and provide the basis for the development of in vitro assays suitable to understand the biological processes involved in the multi-organ systems of the human body, and will improve strategies for diagnosis, prevention, therapies and precision medicine. Induced pluripotent stem cell lines are prone to phenotypic and genotypic changes and donor/clone dependent variability, which means that it is important to identify the most appropriate characterization markers and quality control measures when sourcing new cell lines and assessing differentiated cell and tissue culture preparations for experimental work. This paper considers those core quality control measures for human pluripotent stem cell lines and evaluates the state of play in the development of key functional markers for their differentiated cell derivatives to promote assurance of reproducibility of scientific data derived from pluripotent stem cell-based systems.
Collapse
|
30
|
Song Y, Zheng Z, Lian J. Deciphering Common Long QT Syndrome Using CRISPR/Cas9 in Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Front Cardiovasc Med 2022; 9:889519. [PMID: 35647048 PMCID: PMC9136094 DOI: 10.3389/fcvm.2022.889519] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
From carrying potentially pathogenic genes to severe clinical phenotypes, the basic research in the inherited cardiac ion channel disease such as long QT syndrome (LQTS) has been a significant challenge in explaining gene-phenotype heterogeneity. These have opened up new pathways following the parallel development and successful application of stem cell and genome editing technologies. Stem cell-derived cardiomyocytes and subsequent genome editing have allowed researchers to introduce desired genes into cells in a dish to replicate the disease features of LQTS or replace causative genes to normalize the cellular phenotype. Importantly, this has made it possible to elucidate potential genetic modifiers contributing to clinical heterogeneity and hierarchically manage newly identified variants of uncertain significance (VUS) and more therapeutic options to be tested in vitro. In this paper, we focus on and summarize the recent advanced application of human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) combined with clustered regularly interspaced short palindromic repeats/CRISPR-associated system 9 (CRISPR/Cas9) in the interpretation for the gene-phenotype relationship of the common LQTS and presence challenges, increasing our understanding of the effects of mutations and the physiopathological mechanisms in the field of cardiac arrhythmias.
Collapse
Affiliation(s)
- Yongfei Song
- Department of Cardiovascular, Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo, China
- Yongfei Song
| | - Zequn Zheng
- Department of Cardiovascular, Medical College, Ningbo University, Ningbo, China
- Department of Cardiovascular, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| | - Jiangfang Lian
- Department of Cardiovascular, Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo, China
- Department of Cardiovascular, Medical College, Ningbo University, Ningbo, China
- Department of Cardiovascular, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
- *Correspondence: Jiangfang Lian
| |
Collapse
|
31
|
Querceto S, Santoro R, Gowran A, Grandinetti B, Pompilio G, Regnier M, Tesi C, Poggesi C, Ferrantini C, Pioner JM. The harder the climb the better the view: The impact of substrate stiffness on cardiomyocyte fate. J Mol Cell Cardiol 2022; 166:36-49. [PMID: 35139328 PMCID: PMC11270945 DOI: 10.1016/j.yjmcc.2022.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/22/2021] [Accepted: 02/02/2022] [Indexed: 12/27/2022]
Abstract
The quest for novel methods to mature human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) for cardiac regeneration, modelling and drug testing has emphasized a need to create microenvironments with physiological features. Many studies have reported on how cardiomyocytes sense substrate stiffness and adapt their morphological and functional properties. However, these observations have raised new biological questions and a shared vision to translate it into a tissue or organ context is still elusive. In this review, we will focus on the relevance of substrates mimicking cardiac extracellular matrix (cECM) rigidity for the understanding of the biomechanical crosstalk between the extracellular and intracellular environment. The ability to opportunely modulate these pathways could be a key to regulate in vitro hiPSC-CM maturation. Therefore, both hiPSC-CM models and substrate stiffness appear as intriguing tools for the investigation of cECM-cell interactions. More understanding of these mechanisms may provide novel insights on how cECM affects cardiac cell function in the context of genetic cardiomyopathies.
Collapse
Affiliation(s)
- Silvia Querceto
- Division of Physiology, Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| | - Rosaria Santoro
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy; Department of Electronics, Information and Biomedical Engineering, Politecnico di Milano, Milan, Italy
| | - Aoife Gowran
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Bruno Grandinetti
- European Laboratory for Non-Linear Spectroscopy (LENS), Sesto Fiorentino, FI, Italy
| | - Giulio Pompilio
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy; Department of Biomedical, Surgical and Dental Sciences, University of Milan, Italy
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Chiara Tesi
- Division of Physiology, Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| | - Corrado Poggesi
- Division of Physiology, Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| | - Cecilia Ferrantini
- Division of Physiology, Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| | - Josè Manuel Pioner
- Department of Biology, Università degli Studi di Firenze, Florence, Italy.
| |
Collapse
|
32
|
Varzideh F, Mone P, Santulli G. Bioengineering Strategies to Create 3D Cardiac Constructs from Human Induced Pluripotent Stem Cells. Bioengineering (Basel) 2022; 9:168. [PMID: 35447728 PMCID: PMC9028595 DOI: 10.3390/bioengineering9040168] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 12/12/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) can be used to generate various cell types in the human body. Hence, hiPSC-derived cardiomyocytes (hiPSC-CMs) represent a significant cell source for disease modeling, drug testing, and regenerative medicine. The immaturity of hiPSC-CMs in two-dimensional (2D) culture limit their applications. Cardiac tissue engineering provides a new promise for both basic and clinical research. Advanced bioengineered cardiac in vitro models can create contractile structures that serve as exquisite in vitro heart microtissues for drug testing and disease modeling, thereby promoting the identification of better treatments for cardiovascular disorders. In this review, we will introduce recent advances of bioengineering technologies to produce in vitro cardiac tissues derived from hiPSCs.
Collapse
Affiliation(s)
- Fahimeh Varzideh
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
- Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Pasquale Mone
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
| | - Gaetano Santulli
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
- Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
33
|
Losartan protects human stem cell-derived cardiomyocytes from angiotensin II-induced alcoholic cardiotoxicity. Cell Death Dis 2022; 8:134. [PMID: 35347130 PMCID: PMC8960777 DOI: 10.1038/s41420-022-00945-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 02/18/2022] [Accepted: 03/14/2022] [Indexed: 11/08/2022]
Abstract
Alcoholic cardiomyopathy (ACM) is a myocardial injury caused by long-term heavy drinking. Existing evidence indicates that high levels of oxidative stress are the key to pathological cardiomyopathy caused by long-term exposure to high concentrations of alcohol, while angiotensin II (AngII) and its type 1 receptor (AT1R) play an important role in excessive drinking. Whether oxidative stress-induced damage in ACM is related to AngII and AT1R is unclear, and the effects of alcohol on the electrophysiology of myocardial cells have not been reported. Most existing studies have used animal models. This study established an in vitro model of ACM based on human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). The transcriptional profiling of alcohol treatment was performed by RNA-seq analysis. The role of oxidative stress, the expression of nicotinamide adenine dinucleotide phosphate oxidase (NOX), and the role of AngII and AT1R in the overactivation of oxidative stress were studied using fluorescent labeling, Western blotting, and high-content quantitative analysis. Real-time cell analysis(RTCA) and microelectrode array (MEA) were used to continuously monitor myocardial beating, observe the effects of alcohol on myocardial electrophysiological activity, and clarify the protective effects of the AT1R blocker losartan on ACM. We found that AngII and AT1R contribute to the effects of alcohol on the myocardium through oxidative stress damage, the mechanism of which may be achieved by regulating NOX.
Collapse
|
34
|
Progress in Bioengineering Strategies for Heart Regenerative Medicine. Int J Mol Sci 2022; 23:ijms23073482. [PMID: 35408844 PMCID: PMC8998628 DOI: 10.3390/ijms23073482] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 02/05/2023] Open
Abstract
The human heart has the least regenerative capabilities among tissues and organs, and heart disease continues to be a leading cause of mortality in the industrialized world with insufficient therapeutic options and poor prognosis. Therefore, developing new therapeutic strategies for heart regeneration is a major goal in modern cardiac biology and medicine. Recent advances in stem cell biology and biotechnologies such as human pluripotent stem cells (hPSCs) and cardiac tissue engineering hold great promise for opening novel paths to heart regeneration and repair for heart disease, although these areas are still in their infancy. In this review, we summarize and discuss the recent progress in cardiac tissue engineering strategies, highlighting stem cell engineering and cardiomyocyte maturation, development of novel functional biomaterials and biofabrication tools, and their therapeutic applications involving drug discovery, disease modeling, and regenerative medicine for heart disease.
Collapse
|
35
|
Barc J, Tadros R, Glinge C, Chiang DY, Jouni M, Simonet F, Jurgens SJ, Baudic M, Nicastro M, Potet F, Offerhaus JA, Walsh R, Choi SH, Verkerk AO, Mizusawa Y, Anys S, Minois D, Arnaud M, Duchateau J, Wijeyeratne YD, Muir A, Papadakis M, Castelletti S, Torchio M, Ortuño CG, Lacunza J, Giachino DF, Cerrato N, Martins RP, Campuzano O, Van Dooren S, Thollet A, Kyndt F, Mazzanti A, Clémenty N, Bisson A, Corveleyn A, Stallmeyer B, Dittmann S, Saenen J, Noël A, Honarbakhsh S, Rudic B, Marzak H, Rowe MK, Federspiel C, Le Page S, Placide L, Milhem A, Barajas-Martinez H, Beckmann BM, Krapels IP, Steinfurt J, Winkel BG, Jabbari R, Shoemaker MB, Boukens BJ, Škorić-Milosavljević D, Bikker H, Manevy FC, Lichtner P, Ribasés M, Meitinger T, Müller-Nurasyid M, Veldink JH, van den Berg LH, Van Damme P, Cusi D, Lanzani C, Rigade S, Charpentier E, Baron E, Bonnaud S, Lecointe S, Donnart A, Le Marec H, Chatel S, Karakachoff M, Bézieau S, London B, Tfelt-Hansen J, Roden D, Odening KE, Cerrone M, Chinitz LA, Volders PG, van de Berg MP, Laurent G, Faivre L, Antzelevitch C, Kääb S, Arnaout AA, Dupuis JM, Pasquie JL, Billon O, Roberts JD, Jesel L, Borggrefe M, Lambiase PD, Mansourati J, Loeys B, Leenhardt A, Guicheney P, Maury P, Schulze-Bahr E, Robyns T, Breckpot J, Babuty D, Priori SG, Napolitano C, de Asmundis C, Brugada P, Brugada R, Arbelo E, Brugada J, Mabo P, Behar N, Giustetto C, Molina MS, Gimeno JR, Hasdemir C, Schwartz PJ, Crotti L, McKeown PP, Sharma S, Behr ER, Haissaguerre M, Sacher F, Rooryck C, Tan HL, Remme CA, Postema PG, Delmar M, Ellinor PT, Lubitz SA, Gourraud JB, Tanck MW, George AL, MacRae CA, Burridge PW, Dina C, Probst V, Wilde AA, Schott JJ, Redon R, Bezzina CR. Genome-wide association analyses identify new Brugada syndrome risk loci and highlight a new mechanism of sodium channel regulation in disease susceptibility. Nat Genet 2022; 54:232-239. [PMID: 35210625 DOI: 10.1038/s41588-021-01007-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 12/13/2021] [Indexed: 12/19/2022]
Abstract
Brugada syndrome (BrS) is a cardiac arrhythmia disorder associated with sudden death in young adults. With the exception of SCN5A, encoding the cardiac sodium channel NaV1.5, susceptibility genes remain largely unknown. Here we performed a genome-wide association meta-analysis comprising 2,820 unrelated cases with BrS and 10,001 controls, and identified 21 association signals at 12 loci (10 new). Single nucleotide polymorphism (SNP)-heritability estimates indicate a strong polygenic influence. Polygenic risk score analyses based on the 21 susceptibility variants demonstrate varying cumulative contribution of common risk alleles among different patient subgroups, as well as genetic associations with cardiac electrical traits and disorders in the general population. The predominance of cardiac transcription factor loci indicates that transcriptional regulation is a key feature of BrS pathogenesis. Furthermore, functional studies conducted on MAPRE2, encoding the microtubule plus-end binding protein EB2, point to microtubule-related trafficking effects on NaV1.5 expression as a new underlying molecular mechanism. Taken together, these findings broaden our understanding of the genetic architecture of BrS and provide new insights into its molecular underpinnings.
Collapse
Affiliation(s)
- Julien Barc
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France. .,European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart, .
| | - Rafik Tadros
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Medicine, Cardiovascular Genetics Center, Montreal Heart Institute and Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Charlotte Glinge
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,The Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - David Y Chiang
- Medicine, Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Mariam Jouni
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Floriane Simonet
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Sean J Jurgens
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Manon Baudic
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Michele Nicastro
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Franck Potet
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Joost A Offerhaus
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Roddy Walsh
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Arie O Verkerk
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Medical Biology, University of Amsterdam, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Yuka Mizusawa
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart.,Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Soraya Anys
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Damien Minois
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Marine Arnaud
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Josselin Duchateau
- IHU Liryc, Electrophysiology and Heart Modeling Institute, fondation Bordeaux Université, Pessac-Bordeaux, France.,Université Bordeaux, Centre de recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,INSERM, Centre de recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,Electrophysiology and Ablation Unit, Bordeaux University Hospital (CHU), Pessac, France
| | - Yanushi D Wijeyeratne
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart.,Molecular and Clinical Sciences Research Institute, St. George's, University of London, London, UK.,Cardiology Clinical Academic Group, St. George's University Hospitals' NHS Foundation Trust, London, UK
| | - Alison Muir
- Cardiology, Belfast Health and Social Care Trust and Queen's University Belfast, Belfast, UK
| | - Michael Papadakis
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, London, UK.,Cardiology Clinical Academic Group, St. George's University Hospitals' NHS Foundation Trust, London, UK
| | - Silvia Castelletti
- Center for Cardiac Arrhythmias of Genetic Origin, Istituto Auxologico Italiano IRCCS, Milan, Italy
| | - Margherita Torchio
- Laboratory of Cardiovascular Genetics, Istituto Auxologico Italiano IRCCS, Cusano Milanino, Italy
| | - Cristina Gil Ortuño
- Cardiogenetic, Unidad de Cardiopatías Familiares, Instituto Murciano de Investigación Biosanitaria, Universidad de Murcia, Murcia, Spain
| | - Javier Lacunza
- Cardiology, Unidad de Cardiopatías Familiares, Hospital Universitario Virgen de la Arrixaca, Universidad de Murcia, Murcia, Spain
| | - Daniela F Giachino
- Clinical and Biological Sciences, Medical Genetics, University of Torino, Orbassano, Italy.,Medical Genetics, San Luigi Gonzaga University Hospital, Orbassano, Italy
| | - Natascia Cerrato
- Medical Sciences, Cardiology, University of Torino, Torino, Italy
| | - Raphaël P Martins
- Cardiologie et Maladies vasculaires, Université Rennes1 - CHU Rennes, Rennes, France
| | - Oscar Campuzano
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain.,Medical Science Department, University of Girona, Girona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Biochemistry and Molecular Genetics Department, Hospital Clinic, University of Barcelona-IDIBAPS, Barcelona, Spain
| | - Sonia Van Dooren
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart.,Centre for Medical Genetics, research group Reproduction and Genetics, research cluster Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Aurélie Thollet
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Florence Kyndt
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Andrea Mazzanti
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart.,Molecular Cardiology, ICS Maugeri, IRCCS and Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | | | | | - Anniek Corveleyn
- Department of Human Genetics, Catholic University Leuven, Leuven, Belgium
| | - Birgit Stallmeyer
- University Hospital Münster, Institute for Genetics of Heart Diseases (IfGH), Münster, Germany
| | - Sven Dittmann
- University Hospital Münster, Institute for Genetics of Heart Diseases (IfGH), Münster, Germany
| | - Johan Saenen
- Cardiology, Electrophysiology - Cardiogenetics, University of Antwerp/Antwerp University Hospital, Edegem, Belgium
| | - Antoine Noël
- Department of Cardiology, University Hospital of Brest, Brest, France
| | | | - Boris Rudic
- Department 1st of Medicine, Cardiology, University Medical Center Mannheim, Mannheim, Germany.,German Center for Cardiovascular Research (DZHK), Mannheim, Germany
| | - Halim Marzak
- Department of Cardiology, University Hospital of Strasbourg, Strasbourg, France
| | - Matthew K Rowe
- Medicine, Cardiology, Western University, London, Ontario, Canada
| | - Claire Federspiel
- Department of Cardiovascular Medicine, Vendée Hospital, Service de Cardiologie, La Roche sur Yon, France
| | | | - Leslie Placide
- Department of Cardiology, CHU Montpellier, Montpellier, France
| | - Antoine Milhem
- Department of Cardiology, CH La Rochelle, La Rochelle, France
| | | | - Britt-Maria Beckmann
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany.,University Hospital of the Johann Wolfgang Goethe University Frankfurt, Institute of Legal Medicine, Frankfurt, Germany
| | - Ingrid P Krapels
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Johannes Steinfurt
- Department of Cardiology and Angiology I, Heart Center, University Freiburg, Freiburg, Germany
| | - Bo Gregers Winkel
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart.,The Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Reza Jabbari
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart.,The Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Moore B Shoemaker
- Medicine, Cardiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bas J Boukens
- Department of Medical Biology, University of Amsterdam, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Doris Škorić-Milosavljević
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Hennie Bikker
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart.,Genome Diagnostics Laboratory, Clinical Genetics, Amsterdam UMC, Amsterdam, The Netherlands
| | - Federico C Manevy
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Peter Lichtner
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Marta Ribasés
- Psychiatric Genetics Unit, Institute Vall d'Hebron Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,IBE, LMU Munich, Munich, Germany.,Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany.,Department of Internal Medicine I (Cardiology), Hospital of the Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | | | - Jan H Veldink
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Leonard H van den Berg
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Philip Van Damme
- Neurology Department University Hospital Leuven, Neuroscience Department KU Leuven, Center for Brain & Disease Research VIB, Leuven, Belgium
| | - Daniele Cusi
- Scientific Unit, Bio4Dreams - Business Nursery for Life Sciences, Milan, Italy
| | - Chiara Lanzani
- Nephrology, Genomics of Renal Diseases and Hypertension Unit, Università Vita Salute San Raffaele, Milan, Italy
| | - Sidwell Rigade
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Eric Charpentier
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France.,Université de Nantes, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, Nantes, France
| | - Estelle Baron
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Stéphanie Bonnaud
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France.,Université de Nantes, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, Nantes, France
| | - Simon Lecointe
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Audrey Donnart
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France.,Université de Nantes, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, Nantes, France
| | - Hervé Le Marec
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Stéphanie Chatel
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Matilde Karakachoff
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Stéphane Bézieau
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Barry London
- Department of Internal Medicine, Division of Cardiovascular Medicine, Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Jacob Tfelt-Hansen
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart.,The Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Department of Forensic Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Dan Roden
- Medicine, Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.,Medicine, Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.,Medicine, Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Katja E Odening
- Department of Cardiology and Angiology I, Heart Center, University Freiburg, Freiburg, Germany.,Department of Cardiology, Translational Cardiology, University Hospital Bern, Bern, Switzerland
| | - Marina Cerrone
- Medicine, Leon H. Charney Division of Cardiology, Heart Rhythm Center and Cardiovascular Genetics Program, New York University School of Medicine, New York, NY, USA
| | - Larry A Chinitz
- Medicine, Leon H. Charney Division of Cardiology, Heart Rhythm Center and Cardiovascular Genetics Program, New York University School of Medicine, New York, NY, USA
| | - Paul G Volders
- Department of Cardiology, CARIM, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Maarten P van de Berg
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gabriel Laurent
- Cardiology Department, ImVia lab team IFTIM, University Hospital Dijon, Dijon, France
| | | | | | - Stefan Kääb
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart.,Department of Medicine I, University Hospital, LMU Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partnersite Munich, Munich, Germany
| | | | | | - Jean-Luc Pasquie
- Department of Cardiology, CNRS UMR9214 - Inserm U1046 - PHYMEDEXP, Université de Montpellier et CHU Montpellier, Montpellier, France
| | - Olivier Billon
- Department of Cardiovascular Medicine, Vendée Hospital, Service de Cardiologie, La Roche sur Yon, France
| | - Jason D Roberts
- Medicine, Cardiology, Western University, London, Ontario, Canada
| | - Laurence Jesel
- Department of Cardiology, University Hospital of Strasbourg, Strasbourg, France.,INSERM 1260 - Regenerative Nanomedecine, University of Strasbourg, Strasbourg, France
| | - Martin Borggrefe
- Department 1st of Medicine, Cardiology, University Medical Center Mannheim, Mannheim, Germany.,German Center for Cardiovascular Research (DZHK), Mannheim, Germany
| | - Pier D Lambiase
- Cardiology, Medicine, Barts Heart Centre, London, UK.,Institute of Cardiovasculr Science, UCL, Population Health, UCL, London, UK
| | | | - Bart Loeys
- Center for Medical Genetics, Cardiogenetics, University of Antwerp/Antwerp University Hospital, Edegem, Belgium
| | - Antoine Leenhardt
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart.,Department of Cardiology, Hopital Bichat, Paris, France
| | - Pascale Guicheney
- Sorbonne Université, Paris, France.,UMR_S1166, Faculté de médecine, Sorbonne Université, INSERM, Paris, France
| | - Philippe Maury
- Service de cardiologie, Hôpital Rangueil, CHU de Toulouse, Toulouse, France
| | - Eric Schulze-Bahr
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart.,University Hospital Münster, Institute for Genetics of Heart Diseases (IfGH), Münster, Germany
| | - Tomas Robyns
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart.,Cardiovascular Diseases, University Hospitals Leuven, Leuven, Belgium.,Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Jeroen Breckpot
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart.,Department of Human Genetics, Catholic University Leuven, Leuven, Belgium
| | | | - Silvia G Priori
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart.,Molecular Cardiology, ICS Maugeri, IRCCS and Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Carlo Napolitano
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart.,Molecular Cardiology, ICS Maugeri, IRCCS and Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | | | - Carlo de Asmundis
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Heart Rhythm Management Center, Postgraduate Program in Cardiac Electrophysiology and Pacing Universitair Ziekenhuis, Brussel-Vrije Universiteit Brussel, ERN Heart Guard Center, Brussels, Belgium.,IDIBAPS, Institut d'Investigació August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Pedro Brugada
- Heart Rhythm Management Center, UZ Brussel-VUB, Brussels, Belgium
| | - Ramon Brugada
- Hospital Trueta, CiberCV, University of Girona, IDIBGI, Girona, Spain, Barcelona, Spain
| | - Elena Arbelo
- Arrhythmia Section, Cardiology Department, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Josep Brugada
- Cardiovascular Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Philippe Mabo
- Cardiologie et Maladies vasculaires, Université Rennes1 - CHU Rennes, Rennes, France
| | - Nathalie Behar
- Cardiologie et Maladies vasculaires, Université Rennes1 - CHU Rennes, Rennes, France
| | - Carla Giustetto
- Medical Sciences, Cardiology, University of Torino, Torino, Italy
| | - Maria Sabater Molina
- Cardiogenetic, Unidad de Cardiopatías Familiares, Instituto Murciano de Investigación Biosanitaria, Universidad de Murcia, Murcia, Spain
| | - Juan R Gimeno
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart.,Cardiology, Unidad de Cardiopatías Familiares, Hospital Universitario Virgen de la Arrixaca, Universidad de Murcia, Murcia, Spain
| | - Can Hasdemir
- Department of Cardiology, Ege University School of Medicine, Bornova, Turkey
| | - Peter J Schwartz
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart.,Center for Cardiac Arrhythmias of Genetic Origin, Istituto Auxologico Italiano IRCCS, Milan, Italy.,Laboratory of Cardiovascular Genetics, Istituto Auxologico Italiano IRCCS, Cusano Milanino, Italy
| | - Lia Crotti
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart.,Center for Cardiac Arrhythmias of Genetic Origin, Istituto Auxologico Italiano IRCCS, Milan, Italy.,Laboratory of Cardiovascular Genetics, Istituto Auxologico Italiano IRCCS, Cusano Milanino, Italy.,Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, Istituto Auxologico Italiano IRCCS, Milan, Italy.,Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Pascal P McKeown
- Cardiology, Belfast Health and Social Care Trust and Queen's University Belfast, Belfast, UK
| | - Sanjay Sharma
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, London, UK.,Cardiology Clinical Academic Group, St. George's University Hospitals' NHS Foundation Trust, London, UK
| | - Elijah R Behr
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart.,Molecular and Clinical Sciences Research Institute, St. George's, University of London, London, UK.,Cardiology Clinical Academic Group, St. George's University Hospitals' NHS Foundation Trust, London, UK
| | - Michel Haissaguerre
- IHU Liryc, Electrophysiology and Heart Modeling Institute, fondation Bordeaux Université, Pessac-Bordeaux, France.,Université Bordeaux, Centre de recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,INSERM, Centre de recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,Electrophysiology and Ablation Unit, Bordeaux University Hospital (CHU), Pessac, France
| | - Frédéric Sacher
- IHU Liryc, Electrophysiology and Heart Modeling Institute, fondation Bordeaux Université, Pessac-Bordeaux, France.,Université Bordeaux, Centre de recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,INSERM, Centre de recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,Electrophysiology and Ablation Unit, Bordeaux University Hospital (CHU), Pessac, France
| | - Caroline Rooryck
- CHU Bordeaux, Service de Génétique Médicale, Bordeaux, France.,Université de Bordeaux, Maladies Rares: Génétique et Métabolisme (MRGM), INSERM U1211, Bordeaux, France
| | - Hanno L Tan
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Netherlands Heart Institute, Utrecht, The Netherlands
| | - Carol A Remme
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Pieter G Postema
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart.,Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Mario Delmar
- Medicine, Cardiology, New York University School of Medicine, New York, NY, USA
| | - Patrick T Ellinor
- Cardiac Arrhythmia Service and Cardiovascular Research Center, Massachusetts General Hospital and Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Steven A Lubitz
- Cardiac Arrhythmia Service and Cardiovascular Research Center, Massachusetts General Hospital and Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Jean-Baptiste Gourraud
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France.,European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart
| | - Michael W Tanck
- Clinical Epidemiology, Biostatistics and Bioinformatics, Clinical Methods and Public Health, Amsterdam Public Health, Amsterdam, The Netherlands
| | - Alfred L George
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Calum A MacRae
- Medicine, Cardiovascular Medicine, Genetics and Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Paul W Burridge
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Christian Dina
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Vincent Probst
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France.,European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart
| | - Arthur A Wilde
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart.,Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jean-Jacques Schott
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France.,European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart
| | - Richard Redon
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France.,European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart
| | - Connie R Bezzina
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart, . .,Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
36
|
Veldhuizen J, Chavan R, Moghadas B, Park JG, Kodibagkar VD, Migrino RQ, Nikkhah M. Cardiac ischemia on-a-chip to investigate cellular and molecular response of myocardial tissue under hypoxia. Biomaterials 2022; 281:121336. [PMID: 35026670 PMCID: PMC10440189 DOI: 10.1016/j.biomaterials.2021.121336] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 12/18/2021] [Accepted: 12/24/2021] [Indexed: 12/31/2022]
Abstract
Tissue engineering has enabled the development of advanced and physiologically relevant models of cardiovascular diseases, with advantages over conventional 2D in vitro assays. We have previously demonstrated development of a heart on-a-chip microfluidic model with mature 3D anisotropic tissue formation that incorporates both stem cell-derived cardiomyocytes and cardiac fibroblasts within a collagen-based hydrogel. Using this platform, we herein present a model of myocardial ischemia on-a-chip, that recapitulates ischemic insult through exposure of mature 3D cardiac tissues to hypoxic environments. We report extensive validation and molecular-level analyses of the model in its ability to recapitulate myocardial ischemia in response to hypoxia, demonstrating the 1) induction of tissue fibrosis through upregulation of contractile fibers, 2) dysregulation in tissue contraction through functional assessment, 3) upregulation of hypoxia-response genes and downregulation of contractile-specific genes through targeted qPCR, and 4) transcriptomic pathway regulation of hypoxic tissues. Further, we investigated the complex response of ischemic myocardial tissues to reperfusion, identifying 5) cell toxicity, 6) sustained contractile irregularities, as well as 7) re-establishment of lactate levels and 8) gene expression, in hypoxic tissues in response to ischemia reperfusion injury.
Collapse
Affiliation(s)
- Jaimeson Veldhuizen
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85287, USA
| | - Ramani Chavan
- Center for Personalized Diagnostics (CPD), Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA
| | - Babak Moghadas
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85287, USA
| | - Jin G Park
- Center for Personalized Diagnostics (CPD), Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA
| | - Vikram D Kodibagkar
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85287, USA
| | - Raymond Q Migrino
- Phoenix Veterans Affairs Health Care System, Phoenix, AZ, 85012, USA; University of Arizona College of Medicine, Phoenix, AZ, 85004, USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85287, USA; Center for Personalized Diagnostics (CPD), Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA.
| |
Collapse
|
37
|
van den Brink L, Brandão KO, Yiangou L, Blanch-Asensio A, Mol MPH, Mummery CL, Verkerk AO, Davis RP. The Linkage Phase of the Polymorphism KCNH2-K897T Influences the Electrophysiological Phenotype in hiPSC Models of LQT2. Front Physiol 2022; 12:755642. [PMID: 34992545 PMCID: PMC8726482 DOI: 10.3389/fphys.2021.755642] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/10/2021] [Indexed: 12/29/2022] Open
Abstract
While rare mutations in ion channel genes are primarily responsible for inherited cardiac arrhythmias, common genetic variants are also an important contributor to the clinical heterogeneity observed among mutation carriers. The common single nucleotide polymorphism (SNP) KCNH2-K897T is associated with QT interval duration, but its influence on the disease phenotype in patients with long QT syndrome type 2 (LQT2) remains unclear. Human induced pluripotent stem cells (hiPSCs), coupled with advances in gene editing technologies, are proving an invaluable tool for modeling cardiac genetic diseases and identifying variants responsible for variability in disease expressivity. In this study, we have used isogenic hiPSC-derived cardiomyocytes (hiPSC-CMs) to establish the functional consequences of having the KCNH2-K897T SNP in cis- or trans-orientation with LQT2-causing missense variants either within the pore-loop domain (KCNH2A561T/WT) or tail region (KCNH2N996I/WT) of the potassium ion channel, human ether-a-go-go-related gene (hERG). When KCNH2-K897T was on the same allele (cis) as the primary mutation, the hERG channel in hiPSC-CMs exhibited faster activation and deactivation kinetics compared to their trans-oriented counterparts. Consistent with this, hiPSC-CMs with KCNH2-K897T in cis orientation had longer action and field potential durations. Furthermore, there was an increased occurrence of arrhythmic events upon pharmacological blocking of hERG. Collectively, these results indicate that the common polymorphism KCNH2-K897T differs in its influence on LQT2-causing KCNH2 mutations depending on whether it is present in cis or trans. This study corroborates hiPSC-CMs as a powerful platform to investigate the modifying effects of common genetic variants on inherited cardiac arrhythmias and aids in unraveling their contribution to the variable expressivity of these diseases.
Collapse
Affiliation(s)
- Lettine van den Brink
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Karina O Brandão
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Loukia Yiangou
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Albert Blanch-Asensio
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Mervyn P H Mol
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands.,Department of Applied Stem Cell Technologies, University of Twente, Enschede, Netherlands
| | - Arie O Verkerk
- Department of Medical Biology, Amsterdam UMC, Amsterdam, Netherlands
| | - Richard P Davis
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
38
|
Alam P, Maliken BD, Jones SM, Ivey MJ, Wu Z, Wang Y, Kanisicak O. Cardiac Remodeling and Repair: Recent Approaches, Advancements, and Future Perspective. Int J Mol Sci 2021; 22:ijms222313104. [PMID: 34884909 PMCID: PMC8658114 DOI: 10.3390/ijms222313104] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
The limited ability of mammalian adult cardiomyocytes to proliferate following an injury to the heart, such as myocardial infarction, is a major factor that results in adverse fibrotic and myocardial remodeling that ultimately leads to heart failure. The continued high degree of heart failure-associated morbidity and lethality requires the special attention of researchers worldwide to develop efficient therapeutics for cardiac repair. Recently, various strategies and approaches have been developed and tested to extrinsically induce regeneration and restoration of the myocardium after cardiac injury have yielded encouraging results. Nevertheless, these interventions still lack adequate success to be used for clinical interventions. This review highlights and discusses both cell-based and cell-free therapeutic approaches as well as current advancements, major limitations, and future perspectives towards developing an efficient therapeutic method for cardiac repair.
Collapse
Affiliation(s)
- Perwez Alam
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (P.A.); (S.M.J.); (M.J.I.); (Z.W.); (Y.W.)
| | - Bryan D. Maliken
- Harrington Physician-Scientist Pathway, Department of Internal Medicine, University Hospitals Case Medical Center, Cleveland, OH 44106, USA;
| | - Shannon M. Jones
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (P.A.); (S.M.J.); (M.J.I.); (Z.W.); (Y.W.)
| | - Malina J. Ivey
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (P.A.); (S.M.J.); (M.J.I.); (Z.W.); (Y.W.)
| | - Zhichao Wu
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (P.A.); (S.M.J.); (M.J.I.); (Z.W.); (Y.W.)
| | - Yigang Wang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (P.A.); (S.M.J.); (M.J.I.); (Z.W.); (Y.W.)
| | - Onur Kanisicak
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (P.A.); (S.M.J.); (M.J.I.); (Z.W.); (Y.W.)
- Correspondence: ; Tel.: +1-513-558-2029; Fax: +1-513-584-3892
| |
Collapse
|
39
|
Murphy SA, Chen EZ, Tung L, Boheler KR, Kwon C. Maturing heart muscle cells: Mechanisms and transcriptomic insights. Semin Cell Dev Biol 2021; 119:49-60. [PMID: 33952430 PMCID: PMC8653577 DOI: 10.1016/j.semcdb.2021.04.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 03/23/2021] [Accepted: 04/19/2021] [Indexed: 12/17/2022]
Abstract
Cardiomyocyte (CM) maturation is the transformation of differentiated fetal CMs into adult CMs that involves changes in morphology, cell function and metabolism, and the transcriptome. This process is, however, incomplete and ultimately arrested in pluripotent stem cell-derived CMs (PSC-CMs) in culture, which hinders their broad biomedical application. For this reason, enormous efforts are currently being made with the goal of generating mature PSC-CMs. In this review, we summarize key aspects of maturation observed in native CMs and discuss recent findings on the factors and mechanisms that regulate the process. Particular emphasis is put on transcriptional regulation and single-cell RNA-sequencing analysis that has emerged as a key tool to study time-series gene regulation and to determine the maturation state. We then discuss different biomimetic strategies to enhance PSC-CM maturation and discuss their effects at the single cell transcriptomic and functional levels.
Collapse
Affiliation(s)
- Sean A Murphy
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute of Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Elaine Zhelan Chen
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Leslie Tung
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Kenneth R Boheler
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Chulan Kwon
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute of Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
40
|
Research Highlights. Transplantation 2021. [DOI: 10.1097/tp.0000000000003956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
41
|
Roshanzadeh A, Oyunbaatar NE, Ganjbakhsh SE, Park S, Kim DS, Kanade PP, Lee S, Lee DW, Kim ES. Exposure to nanoplastics impairs collective contractility of neonatal cardiomyocytes under electrical synchronization. Biomaterials 2021; 278:121175. [PMID: 34628193 DOI: 10.1016/j.biomaterials.2021.121175] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/16/2021] [Accepted: 09/30/2021] [Indexed: 10/20/2022]
Abstract
Nanoplastics are global pollutants that have been increasingly released into the environment following the degradation process of industrial and consumer products. These tiny particles have been reported to adversely affect various organs in the body, including the heart. Since it is probable that the less-developed hearts of newborn offspring are more vulnerable to nanoplastic insult during the infant feeding compared with mature hearts of adults, the acute effects of nanoplastics on the collective contractility of neonatal cardiomyocytes are to be elucidated. Here, we traced the aggregation of nanoplastics on the cell membrane and their internalization into the cytosol of neonatal rat ventricular myocytes (NRVMs) for 60 min in the presence of electrical pulses to synchronize the cardiac contraction in vitro. The time-coursed linkage of collective contraction forces, intracellular Ca2+ concentrations, mitochondrial membrane potentials, extracellular field potentials, and reactive oxygen species levels enabled us to build up the sequence of the cellular events associated with the detrimental effects of nanoplastics with positive surface charges on the immature cardiomyocytes. A significant decrease in intracellular Ca2+ levels and electrophysiological activities of NRVMs resulted in the reduction of contraction forces in the early phase (0-15 min). The further reduction of contraction force in the late phase (30-60 min) was attributed to remarkable decreases in mitochondrial membrane potentials and cellular metabolism. Our multifaceted assessments on the effect of positively surface charged nanoplastics on NRVM may offer better understanding of substantial risks of ever-increasing nanoplastic pollution in the hearts of human infants or adults.
Collapse
Affiliation(s)
- Amir Roshanzadeh
- School of Biological Sciences and Biotechnology, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Nomin-Erdene Oyunbaatar
- School of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | | | - Sangwoo Park
- Gwangju Center, Korea Basic Science Institute (KBSI), 49 Dosicheomdansaneopro, Nam-gu, Gwangju, 61751, Republic of Korea
| | - Dong-Su Kim
- School of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Pooja P Kanade
- School of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Seongsoo Lee
- Gwangju Center, Korea Basic Science Institute (KBSI), 49 Dosicheomdansaneopro, Nam-gu, Gwangju, 61751, Republic of Korea
| | - Dong-Weon Lee
- School of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Center for Next Generation Sensor Research and Development, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Eung-Sam Kim
- School of Biological Sciences and Biotechnology, Chonnam National University, Gwangju, 61186, Republic of Korea; Center for Next Generation Sensor Research and Development, Chonnam National University, Gwangju, 61186, Republic of Korea; Department of Biological Sciences and Research Center of Ecomimetics, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
42
|
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
|
43
|
QuinoMit Q10-Fluid attenuates hydrogen peroxide-induced irregular beating in mouse pluripotent stem cell-derived cardiomyocytes. Biomed Pharmacother 2021; 142:112089. [PMID: 34449318 DOI: 10.1016/j.biopha.2021.112089] [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: 03/20/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Coenzyme Q10 (CoQ10) is a crucial component of the mitochondrial structure which is involved in producing more than 90% of cellular ATP. This study aimed to investigate the protective effects and underlying mechanisms of QuinoMit Q10-Fluid against hydrogen peroxide (H2O2)-induced arrhythmias on cardiomyocytes (CMs). METHODS Undifferentiated stem cell-derived CMs were cultured in the presence of different concentrations of QuinoMit Q10-Fluid. To investigate if CoQ10 has anti-apoptotic activity, CMs were exposed to H2O2 for up to 100 h with or without CoQ10. The expression levels of cardiac reference genes were determined by RT-PCR. The structural and functional properties of CMs were examined by immunofluorescence and the xCELLigence system. Caspase 3/7 assay was also performed for cell apoptosis study. RESULTS The study showed that QuinoMit Q10-Fluid inhibits the proliferation of pluripotent stem cells at high concentrations and had less effect on cardiomyogenesis. However, the beating rate of clusters containing CMs generated under QuinoMit Q10-Fluid (1:100) was significantly increased. This increase was accompanied by the up-regulated expression level of some important cardiac markers during differentiation. Treatment of CMs with H2O2 notably induced irregular beating and decreased the amplitude of the beating signal of CMs, concomitantly with increased caspase-3/7 activity. However, CMs pretreated with QuinoMit exhibited a protective effect against H2O2-induced arrhythmia. CONCLUSION Our results reveal that QuinoMit Q10-Fluid attenuates H2O2-induced irregular beating in mouse pluripotent stem cell-derived CMs, at least partly by reducing the generation of ROS, suggesting a protective effect against CM dysfunctions.
Collapse
|
44
|
Tan JJ, Guyette JP, Miki K, Xiao L, Kaur G, Wu T, Zhu L, Hansen KJ, Ling KH, Milan DJ, Ott HC. Human iPS-derived pre-epicardial cells direct cardiomyocyte aggregation expansion and organization in vitro. Nat Commun 2021; 12:4997. [PMID: 34404774 PMCID: PMC8370973 DOI: 10.1038/s41467-021-24921-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 07/10/2021] [Indexed: 12/13/2022] Open
Abstract
Epicardial formation is necessary for normal myocardial morphogenesis. Here, we show that differentiating hiPSC-derived lateral plate mesoderm with BMP4, RA and VEGF (BVR) can generate a premature form of epicardial cells (termed pre-epicardial cells, PECs) expressing WT1, TBX18, SEMA3D, and SCX within 7 days. BVR stimulation after Wnt inhibition of LPM demonstrates co-differentiation and spatial organization of PECs and cardiomyocytes (CMs) in a single 2D culture. Co-culture consolidates CMs into dense aggregates, which then form a connected beating syncytium with enhanced contractility and calcium handling; while PECs become more mature with significant upregulation of UPK1B, ITGA4, and ALDH1A2 expressions. Our study also demonstrates that PECs secrete IGF2 and stimulate CM proliferation in co-culture. Three-dimensional PEC-CM spheroid co-cultures form outer smooth muscle cell layers on cardiac micro-tissues with organized internal luminal structures. These characteristics suggest PECs could play a key role in enhancing tissue organization within engineered cardiac constructs in vitro. The authors form pre-epicardial cells (PECs) from hiPSC-derived lateral plate mesoderm on treating with BMP4, RA and VEGF, and co-culture these PECs with cardiomyocytes, inducing cardiomyocyte aggregation, proliferation and network formation with more mature structures and improved beating/contractility.
Collapse
Affiliation(s)
- Jun Jie Tan
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA. .,Harvard Medical School, Boston, MA, USA. .,Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia.
| | - Jacques P Guyette
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Kenji Miki
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Center for iPS Cell Research and Applications, Kyoto University, Kyoto, Japan
| | - Ling Xiao
- Harvard Medical School, Boston, MA, USA.,Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Gurbani Kaur
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Tong Wu
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Liye Zhu
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Katrina J Hansen
- Worcester Polytechnic Institute, Dept. of Biomedical Engineering, Worcester, MA, USA
| | - King-Hwa Ling
- Department of Genetics, Harvard Medical School, Boston, MA, USA.,Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - David J Milan
- Harvard Medical School, Boston, MA, USA.,Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA.,Leducq Foundation, Boston, MA, USA
| | - Harald C Ott
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA. .,Harvard Medical School, Boston, MA, USA. .,Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA. .,Harvard Stem Cell Institute, Boston, MA, USA.
| |
Collapse
|
45
|
Chu X, Wang M, Qiu X, Huang Y, Li T, Otieno E, Li N, Luo L, Xiao X. Strategies for constructing pluripotent stem cell- and progenitor cell-derived three-dimensional cardiac micro-tissues. J Biomed Mater Res A 2021; 110:488-503. [PMID: 34397148 DOI: 10.1002/jbm.a.37298] [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: 02/20/2021] [Revised: 07/31/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022]
Abstract
Three-dimensional (3D) cardiac micro-tissue is a promising model for simulating the structural and functional features of heart in vitro. This scientific achievement provides a platform for exploration about the mechanisms on the development, damage, and regeneration of tissue, hence, paving a way toward development of novel therapies for heart diseases. However, 3D micro-tissue technology is still in its infant stages faced with many challenges such as incompleteness of the tissue microarchitecture, loss of the resident immune cells, poor reproducibility, and deficiencies in continuously feeding the nutrients and removing wastes during micro-tissue culturing. There is an urgent need to optimize the construction of 3D cardiac micro-tissue and improve functions of the involved cells. Therefore, scaffolds and cell resources for building 3D cardiac micro-tissues, strategies for inducing the maturation and functionalization of pluripotent stem cell- or cardiac progenitor cell-derived cardiomyocytes, and the major challenges were reviewed in this writing to enable future fabrication of 3D cardiac micro-tissues or organoids for drug screening, disease modeling, regeneration treatment, and so on.
Collapse
Affiliation(s)
- Xinyue Chu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Mingyu Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China.,Institute of Laboratory Animals Science, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Xiaoyan Qiu
- Department of Animal Husbandry Engineering, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Yun Huang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Tong Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Edward Otieno
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Na Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Li Luo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Xiong Xiao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| |
Collapse
|
46
|
Giacomelli E, Sala L, Oostwaard DWV, Bellin M. Cardiac microtissues from human pluripotent stem cells recapitulate the phenotype of long-QT syndrome. Biochem Biophys Res Commun 2021; 572:118-124. [PMID: 34364290 DOI: 10.1016/j.bbrc.2021.07.068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 07/19/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Human induced pluripotent stem cells (hiPSCs) and their derivative cardiomyocytes (hiPSC-CMs) have been successfully used to study the electrical phenotype of cardiac ion channel diseases. However, strategies to mature CMs and more comprehensive systems recapitulating the heart complexity are required to advance our ability to capture adult phenotypes. METHODS We differentiated wild-type (WT) and long QT syndrome type 1 (LQT1) hiPSCs into CMs, endothelial cells and cardiac fibroblasts. The three cell types were combined to form three-dimensional (3D) spheroids, termed "cardiac microtissues" (cMTs) and the electrophysiological properties were measured using 96-well multi-electrode arrays. RESULTS LQT1 cMTs displayed prolonged field potential duration compared to WT controls, thus recapitulating the typical feature of LQTS. Isoprenaline caused a positive chronotropic effect on both LQT1 and WT cMTs. The 96-well multi-electrode array format proved suitable to detect electrical changes directly in the 3D tissues. CONCLUSIONS 3D hiPSC cMTs are a scalable tool that can be used to identify LQT electrical hallmarks and drug responses. We anticipate this tool can be adopted by pharmaceutical companies to screen cardio-active compounds.
Collapse
Affiliation(s)
- Elisa Giacomelli
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), 2333, ZA, Leiden, the Netherlands; The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY, USA
| | - Luca Sala
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), 2333, ZA, Leiden, the Netherlands; Istituto Auxologico Italiano IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milan, Italy
| | - Dorien Ward-van Oostwaard
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), 2333, ZA, Leiden, the Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), 2333, ZA, Leiden, the Netherlands; Department of Biology, University of Padua, 35121, Padua, Italy; Veneto Institute of Molecular Medicine, 35129, Padua, Italy.
| |
Collapse
|
47
|
Grancharova T, Gerbin KA, Rosenberg AB, Roco CM, Arakaki JE, DeLizo CM, Dinh SQ, Donovan-Maiye RM, Hirano M, Nelson AM, Tang J, Theriot JA, Yan C, Menon V, Palecek SP, Seelig G, Gunawardane RN. A comprehensive analysis of gene expression changes in a high replicate and open-source dataset of differentiating hiPSC-derived cardiomyocytes. Sci Rep 2021; 11:15845. [PMID: 34349150 PMCID: PMC8338992 DOI: 10.1038/s41598-021-94732-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/12/2021] [Indexed: 12/13/2022] Open
Abstract
We performed a comprehensive analysis of the transcriptional changes occurring during human induced pluripotent stem cell (hiPSC) differentiation to cardiomyocytes. Using single cell RNA-seq, we sequenced > 20,000 single cells from 55 independent samples representing two differentiation protocols and multiple hiPSC lines. Samples included experimental replicates ranging from undifferentiated hiPSCs to mixed populations of cells at D90 post-differentiation. Differentiated cell populations clustered by time point, with differential expression analysis revealing markers of cardiomyocyte differentiation and maturation changing from D12 to D90. We next performed a complementary cluster-independent sparse regression analysis to identify and rank genes that best assigned cells to differentiation time points. The two highest ranked genes between D12 and D24 (MYH7 and MYH6) resulted in an accuracy of 0.84, and the three highest ranked genes between D24 and D90 (A2M, H19, IGF2) resulted in an accuracy of 0.94, revealing that low dimensional gene features can identify differentiation or maturation stages in differentiating cardiomyocytes. Expression levels of select genes were validated using RNA FISH. Finally, we interrogated differences in cardiac gene expression resulting from two differentiation protocols, experimental replicates, and three hiPSC lines in the WTC-11 background to identify sources of variation across these experimental variables.
Collapse
Affiliation(s)
| | | | - Alexander B Rosenberg
- Department of Electrical & Computer Engineering, University of Washington, Seattle, WA, USA.,, Parse Biosciences, Seattle, WA, USA
| | - Charles M Roco
- , Parse Biosciences, Seattle, WA, USA.,Department of Bioengineering, University of Washington, Seattle, WA, USA
| | | | | | | | | | - Matthew Hirano
- Department of Electrical & Computer Engineering, University of Washington, Seattle, WA, USA
| | | | - Joyce Tang
- Allen Institute for Cell Science, Seattle, WA, USA
| | - Julie A Theriot
- Allen Institute for Cell Science, Seattle, WA, USA.,Department of Biology, Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Calysta Yan
- Allen Institute for Cell Science, Seattle, WA, USA
| | - Vilas Menon
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin - Madison, Madison, WI, USA
| | - Georg Seelig
- Department of Electrical & Computer Engineering, University of Washington, Seattle, WA, USA.,Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
48
|
Marchal GA, Jouni M, Chiang DY, Pérez-Hernández M, Podliesna S, Yu N, Casini S, Potet F, Veerman CC, Klerk M, Lodder EM, Mengarelli I, Guan K, Vanoye CG, Rothenberg E, Charpentier F, Redon R, George AL, Verkerk AO, Bezzina CR, MacRae CA, Burridge PW, Delmar M, Galjart N, Portero V, Remme CA. Targeting the Microtubule EB1-CLASP2 Complex Modulates Na V1.5 at Intercalated Discs. Circ Res 2021; 129:349-365. [PMID: 34092082 PMCID: PMC8298292 DOI: 10.1161/circresaha.120.318643] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Gerard A Marchal
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Mariam Jouni
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - David Y Chiang
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA (D.Y.C., C.A.M.)
| | | | - Svitlana Podliesna
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Nuo Yu
- Department of Cell Biology, Erasmus Medical Centre Rotterdam, The Netherlands (N.Y., N.G.)
| | - Simona Casini
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Franck Potet
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - Christiaan C Veerman
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Mischa Klerk
- Department of Medical Biology, Amsterdam UMC - location AMC, The Netherlands (M.K., A.O.V.)
| | - Elisabeth M Lodder
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Isabella Mengarelli
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Germany (K.G.)
| | - Carlos G Vanoye
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - Eli Rothenberg
- Department of Biochemistry and Pharmacology (E.R.), NYU School of Medicine
| | - Flavien Charpentier
- Université de Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France (F.C., R.R., V.P.)
| | - Richard Redon
- Université de Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France (F.C., R.R., V.P.)
| | - Alfred L George
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - Arie O Verkerk
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
- Department of Medical Biology, Amsterdam UMC - location AMC, The Netherlands (M.K., A.O.V.)
| | - Connie R Bezzina
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Calum A MacRae
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA (D.Y.C., C.A.M.)
| | - Paul W Burridge
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - Mario Delmar
- Division of Cardiology (M.P.-H., M.D.), NYU School of Medicine
| | - Niels Galjart
- Department of Cell Biology, Erasmus Medical Centre Rotterdam, The Netherlands (N.Y., N.G.)
| | - Vincent Portero
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
- Université de Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France (F.C., R.R., V.P.)
| | - Carol Ann Remme
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| |
Collapse
|
49
|
Aghasafari P, Yang PC, Kernik DC, Sakamoto K, Kanda Y, Kurokawa J, Vorobyov I, Clancy CE. A deep learning algorithm to translate and classify cardiac electrophysiology. eLife 2021; 10:68335. [PMID: 34212860 PMCID: PMC8282335 DOI: 10.7554/elife.68335] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/29/2021] [Indexed: 01/15/2023] Open
Abstract
The development of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) has been a critical in vitro advance in the study of patient-specific physiology, pathophysiology, and pharmacology. We designed a new deep learning multitask network approach intended to address the low throughput, high variability, and immature phenotype of the iPSC-CM platform. The rationale for combining translation and classification tasks is because the most likely application of the deep learning technology we describe here is to translate iPSC-CMs following application of a perturbation. The deep learning network was trained using simulated action potential (AP) data and applied to classify cells into the drug-free and drugged categories and to predict the impact of electrophysiological perturbation across the continuum of aging from the immature iPSC-CMs to the adult ventricular myocytes. The phase of the AP extremely sensitive to perturbation due to a steep rise of the membrane resistance was found to contain the key information required for successful network multitasking. We also demonstrated successful translation of both experimental and simulated iPSC-CM AP data validating our network by prediction of experimental drug-induced effects on adult cardiomyocyte APs by the latter.
Collapse
Affiliation(s)
- Parya Aghasafari
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, United States
| | - Pei-Chi Yang
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, United States
| | - Divya C Kernik
- Washington University in St. Louis, St. Louis, United States
| | - Kazuho Sakamoto
- Department of Bio-Informational Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, Kanagawa, Japan
| | - Junko Kurokawa
- Department of Bio-Informational Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Igor Vorobyov
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, United States.,Department of Pharmacology, University of California, Davis, Davis, United States
| | - Colleen E Clancy
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, United States
| |
Collapse
|
50
|
Dwyer KD, Coulombe KL. Cardiac mechanostructure: Using mechanics and anisotropy as inspiration for developing epicardial therapies in treating myocardial infarction. Bioact Mater 2021; 6:2198-2220. [PMID: 33553810 PMCID: PMC7822956 DOI: 10.1016/j.bioactmat.2020.12.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 12/14/2022] Open
Abstract
The mechanical environment and anisotropic structure of the heart modulate cardiac function at the cellular, tissue and organ levels. During myocardial infarction (MI) and subsequent healing, however, this landscape changes significantly. In order to engineer cardiac biomaterials with the appropriate properties to enhance function after MI, the changes in the myocardium induced by MI must be clearly identified. In this review, we focus on the mechanical and structural properties of the healthy and infarcted myocardium in order to gain insight about the environment in which biomaterial-based cardiac therapies are expected to perform and the functional deficiencies caused by MI that the therapy must address. From this understanding, we discuss epicardial therapies for MI inspired by the mechanics and anisotropy of the heart focusing on passive devices, which feature a biomaterials approach, and active devices, which feature robotic and cellular components. Through this review, a detailed analysis is provided in order to inspire further development and translation of epicardial therapies for MI.
Collapse
Affiliation(s)
- Kiera D. Dwyer
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, USA
| | - Kareen L.K. Coulombe
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, USA
| |
Collapse
|