1
|
Li W, Luo X, Strano A, Arun S, Gamm O, Poetsch MS, Hasse M, Steiner RP, Fischer K, Pöche J, Ulbricht Y, Lesche M, Trimaglio G, El-Armouche A, Dahl A, Mirtschink P, Guan K, Schubert M. Comprehensive promotion of iPSC-CM maturation by integrating metabolic medium with nanopatterning and electrostimulation. Nat Commun 2025; 16:2785. [PMID: 40118846 PMCID: PMC11928738 DOI: 10.1038/s41467-025-58044-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 03/03/2025] [Indexed: 03/24/2025] Open
Abstract
The immaturity of human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) is a major limitation for their use in drug screening to identify pro-arrhythmogenic or cardiotoxic molecules. Here, we demonstrate an approach that combines lipid-enriched maturation medium with a high concentration of calcium, nanopatterning of culture surfaces and electrostimulation to generate iPSC-CMs with advanced electrophysiological, structural and metabolic phenotypes. Systematic testing reveals that electrostimulation is the key driver of enhanced mitochondrial development and metabolic maturation and improved electrophysiological properties of iPSC-CMs. Increased calcium concentration strongly promotes electrophysiological maturation, while nanopatterning primarily facilitates sarcomere organisation with minor effect on electrophysiological properties. Transcriptome analysis reveals that activation of HMCES and TFAM targets contributes to mitochondrial development, whereas downregulation of MAPK/PI3K and SRF targets is associated with iPSC-CM polyploidy. These findings provide mechanistic insights into iPSC-CM maturation, paving the way for pharmacological responses that more closely resemble those of adult CMs.
Collapse
Affiliation(s)
- Wener Li
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Xiaojing Luo
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Anna Strano
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Shakthi Arun
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Oliver Gamm
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Mareike S Poetsch
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Marcel Hasse
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Robert-Patrick Steiner
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Konstanze Fischer
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Jessie Pöche
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Ying Ulbricht
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Mathias Lesche
- DRESDEN-concept Genome Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Giulia Trimaglio
- Institute of Clinical Chemistry and Laboratory Medicine, Department of Clinical Pathobiochemistry, University Hospital Dresden, Dresden, Germany
- National Center for Tumor Diseases, Partner Site Dresden, 01307 Dresden, and German Cancer Research Center, Heidelberg, Germany
| | - Ali El-Armouche
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Andreas Dahl
- DRESDEN-concept Genome Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Peter Mirtschink
- Institute of Clinical Chemistry and Laboratory Medicine, Department of Clinical Pathobiochemistry, University Hospital Dresden, Dresden, Germany
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany.
| | - Mario Schubert
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
2
|
Sun Z, Sen P, Hamers J, Seidel T, Dendorfer A, Kameritsch P. Optimized Conditions for Electrical Tissue Stimulation with Biphasic, Charge-Balanced Impulses. Bioengineering (Basel) 2025; 12:234. [PMID: 40150698 PMCID: PMC11939772 DOI: 10.3390/bioengineering12030234] [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: 01/10/2025] [Revised: 02/15/2025] [Accepted: 02/22/2025] [Indexed: 03/29/2025] Open
Abstract
The cultivation of excitable cells typically profits from continuous electrical stimulation, but electrochemical consequences are mostly harmful and must be minimized. The properties of the electrode materials and stimulation impulses are key. Here, we developed an easy method to analyze the electrochemical impact of biphasic, current-controlled impulses, applied via graphite electrodes, using phenol red as the redox indicator. We also tested the stimulation conditions for the long-term cultivation of myocardial tissue. The colorimetric assay was able to detect ±0.2% deviations in typical positive and negative pulse charges. Phenol red was best preserved (20% degradation over 24 h) by impulses of equivalent positive and negative charges (full charge balance), generated with either manual calibration, capacitive electrode coupling, or feedback regulation of electrode polarization. Feedback regulation established full charge balance at pre-pulse voltages of about 300 mV, but also provided the option to selectively compensate irreversible electrode reactions. Modifications to shape and timing did not affect the electrochemical effects of symmetric impulses. Charge-balanced stimulation maintained more than 80% of the contractility of porcine left ventricular myocardium after 10 days of culture, whereas disbalances of 2-4% provoked weakening and discoloration of the tissues. Active polarization regulation, in contrast to capacitive electrode coupling, reproduced the biological advantages of full charge balance.
Collapse
Affiliation(s)
- Zhengwu Sun
- Walter-Brendel-Centre of Experimental Medicine, LMU Klinikum, Ludwig-Maximilians-University, 81377 München, Germany; (Z.S.); (P.S.); (J.H.); (A.D.)
| | - Payel Sen
- Walter-Brendel-Centre of Experimental Medicine, LMU Klinikum, Ludwig-Maximilians-University, 81377 München, Germany; (Z.S.); (P.S.); (J.H.); (A.D.)
- DZHK (German Center for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), 80336 Munich, Germany
| | - Jules Hamers
- Walter-Brendel-Centre of Experimental Medicine, LMU Klinikum, Ludwig-Maximilians-University, 81377 München, Germany; (Z.S.); (P.S.); (J.H.); (A.D.)
- DZHK (German Center for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), 80336 Munich, Germany
| | - Thomas Seidel
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Andreas Dendorfer
- Walter-Brendel-Centre of Experimental Medicine, LMU Klinikum, Ludwig-Maximilians-University, 81377 München, Germany; (Z.S.); (P.S.); (J.H.); (A.D.)
- DZHK (German Center for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), 80336 Munich, Germany
| | - Petra Kameritsch
- Walter-Brendel-Centre of Experimental Medicine, LMU Klinikum, Ludwig-Maximilians-University, 81377 München, Germany; (Z.S.); (P.S.); (J.H.); (A.D.)
| |
Collapse
|
3
|
Lodrini AM, Groeneveld EJ, Palmen M, Hjortnaes J, Smits AM, Goumans M. Cryopreservation of Human Adult Ventricular Tissue for the Preparation of Viable Myocardial Slices. Curr Protoc 2024; 4:e70068. [PMID: 39625241 PMCID: PMC11613814 DOI: 10.1002/cpz1.70068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2024]
Abstract
Living myocardial slices (LMS) are ultrathin sections of adult myocardium that can be maintained in culture. These slices provide a unique platform for studying interactions between cardiomyocytes (CMs), other cardiac cell types, and the extracellular matrix while maintaining the cytoarchitecture and electrical phenotype of CMs over extended periods. Despite their advantages over other cardiac models, LMS have limitations, particularly their reliance on slice quality. The primary factor influencing the quality of the slices is the method used to process the cardiac tissue block. Current methods typically require immediate slice preparation following the excision of the tissue block, which restricts the timing of experiments. To address this limitation, we developed a simple procedure for cryopreserving human adult myocardium, allowing the preparation of LMS at a later stage. The protocol provides a list of required equipment and reagents, as well as a detailed description of the methodology for processing the myocardium and slice preparation. We present typical results demonstrating that cryopreserved human cardiac tissue retains biomass and structural integrity comparable to freshly obtained myocardium. Furthermore, we assessed the LMS derived from both fresh and cryopreserved samples. Histological analysis confirmed the preservation of viability, normal cytomorphology, and gap junctions between CMs in all LMS after 24 h and up to 5 days of culture in the absence of electrical stimulation. Cryopreservation extends the interval between tissue collection and LMS preparation, facilitating longer-term and more complex experiments. Further research into the impact of cryopreservation on various cardiac cell types will promote better donor organ management and efficient banking of cardiac samples from a multitude of donors and disease states. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Preparation and preservation of human adult myocardium Basic Protocol 2: Preparation of adult living myocardial slices from cryopreserved blocks.
Collapse
Affiliation(s)
- Alessandra M. Lodrini
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenNetherlands
| | - Esmee J. Groeneveld
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenNetherlands
| | - Meindert Palmen
- Department of Cardiothoracic SurgeryLeiden University Medical CenterLeidenNetherlands
| | - Jesper Hjortnaes
- Department of Cardiothoracic SurgeryLeiden University Medical CenterLeidenNetherlands
| | - Anke M. Smits
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenNetherlands
| | - Marie‐José Goumans
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenNetherlands
| |
Collapse
|
4
|
Tan S, Yang J, Hu S, Lei W. Cell-cell interactions in the heart: advanced cardiac models and omics technologies. Stem Cell Res Ther 2024; 15:362. [PMID: 39396018 PMCID: PMC11470663 DOI: 10.1186/s13287-024-03982-z] [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: 08/14/2024] [Accepted: 10/06/2024] [Indexed: 10/14/2024] Open
Abstract
A healthy heart comprises various cell types, including cardiomyocytes, endothelial cells, fibroblasts, immune cells, and among others, which work together to maintain optimal cardiac function. These cells engage in complex communication networks, known as cell-cell interactions (CCIs), which are essential for homeostasis, cardiac structure, and efficient function. However, in the context of cardiac diseases, the heart undergoes damage, leading to alterations in the cellular composition. Such pathological conditions trigger significant changes in CCIs, causing cell rearrangement and the transition between cell types. Studying these interactions can provide valuable insights into cardiac biology and disease mechanisms, enabling the development of new therapeutic strategies. While the development of cardiac organoids and advanced 3D co-culture technologies has revolutionized in vitro studies of CCIs, recent advancements in single-cell and spatial multi-omics technologies provide researchers with powerful and convenient tools to investigate CCIs at unprecedented resolution. This article provides a concise overview of CCIs observed in both normal and injured heart, with an emphasis on the cutting-edge methods used to study these interactions. It highlights recent advancements such as 3D co-culture systems, single-cell and spatial omics technologies, that have enhanced the understanding of CCIs. Additionally, it summarizes the practical applications of CCI research in advancing cardiovascular therapies, offering potential solutions for treating heart disease by targeting intercellular communication.
Collapse
Affiliation(s)
- Shuai Tan
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Jingsi Yang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China.
| | - Wei Lei
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China.
| |
Collapse
|
5
|
Hwang DG, Kang W, Park SM, Jang J. Biohybrid printing approaches for cardiac pathophysiological studies. Biosens Bioelectron 2024; 260:116420. [PMID: 38805890 DOI: 10.1016/j.bios.2024.116420] [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: 02/24/2024] [Revised: 04/30/2024] [Accepted: 05/20/2024] [Indexed: 05/30/2024]
Abstract
Bioengineered hearts, which include single cardiomyocytes, engineered heart tissue, and chamber-like models, generate various biosignals, such as contractility, electrophysiological, and volume-pressure dynamic signals. Monitoring changes in these signals is crucial for understanding the mechanisms of disease progression and developing potential treatments. However, current methodologies face challenges in the continuous monitoring of bioengineered hearts over extended periods and typically require sacrificing the sample post-experiment, thereby limiting in-depth analysis. Thus, a biohybrid system consisting of living and nonliving components was developed. This system primarily features heart tissue alongside nonliving elements designed to support or comprehend its functionality. Biohybrid printing technology has simplified the creation of such systems and facilitated the development of various functional biohybrid systems capable of measuring or even regulating multiple functions, such as pacemakers, which demonstrates its versatility and potential applications. The future of biohybrid printing appears promising, with the ongoing exploration of its capabilities and potential directions for advancement.
Collapse
Affiliation(s)
- Dong Gyu Hwang
- Center for 3D Organ Printing and Stem Cells, Pohang University of Science and Technology (POSTECH), Pohang, 37563, Republic of Korea
| | - Wonok Kang
- Department of Convergence IT Engineering (POSTECH), Pohang, 37666, Republic of Korea
| | - Sung-Min Park
- Department of Convergence IT Engineering (POSTECH), Pohang, 37666, Republic of Korea; Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37666, Republic of Korea; School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, 37666, Republic of Korea; Department of Electrical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea; Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, 03722, Republic of Korea.
| | - Jinah Jang
- Center for 3D Organ Printing and Stem Cells, Pohang University of Science and Technology (POSTECH), Pohang, 37563, Republic of Korea; Department of Convergence IT Engineering (POSTECH), Pohang, 37666, Republic of Korea; Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37666, Republic of Korea; School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, 37666, Republic of Korea; Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
6
|
Baron V, Sommer ST, Fiegle DJ, Pfeuffer AKM, Peyronnet R, Volk T, Seidel T. Effects of electro-mechanical uncouplers, hormonal stimulation and pacing rate on the stability and function of cultured rabbit myocardial slices. Front Bioeng Biotechnol 2024; 12:1363538. [PMID: 38646013 PMCID: PMC11026719 DOI: 10.3389/fbioe.2024.1363538] [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: 12/30/2023] [Accepted: 03/18/2024] [Indexed: 04/23/2024] Open
Abstract
Introduction: Recent advances have enabled organotypic culture of beating human myocardial slices that are stable for weeks. However, human myocardial samples are rare, exhibit high variability and frequently originate from diseased hearts. Thus, there is a need to adapt long-term slice culture for animal myocardium. When applied to animal cardiac slices, studies in healthy or genetically modified myocardium will be possible. We present the culture of slices from rabbit hearts, which resemble the human heart in microstructure, electrophysiology and excitation-contraction coupling. Methods: Left ventricular myocardium from New Zealand White rabbits was cut using a vibratome and cultured in biomimetic chambers for up to 7 days (d). Electro-mechanical uncoupling agents 2,3-butanedione monoxime (BDM) and cytochalasin D (CytoD) were added during initiation of culture and effects on myocyte survival were quantified. We investigated pacing rates (0.5 Hz, 1 Hz, and 2 Hz) and hormonal supplements (cortisol, T3, catecholamines) at physiological plasma concentrations. T3 was buffered using BSA. Contractile force was recorded continuously. Glucose consumption and lactate production were measured. Whole-slice Ca2+ transients and action potentials were recorded. Effects of culture on microstructure were investigated with confocal microscopy and image analysis. Results: Protocols for human myocardial culture resulted in sustained contracture and myocyte death in rabbit slices within 24 h, which could be prevented by transient application of a combination of BDM and CytoD. Cortisol stabilized contraction amplitude and kinetics in culture. T3 and catecholaminergic stimulation did not further improve stability. T3 and higher pacing rates increased metabolic rate and lactate production. T3 stabilized the response to β-adrenergic stimulation over 7 d. Pacing rates above 1 Hz resulted in progredient decline in contraction force. Image analysis revealed no changes in volume fractions of cardiomyocytes or measures of fibrosis over 7 d. Ca2+ transient amplitudes and responsiveness to isoprenaline were comparable after 1 d and 7 d, while Ca2+ transient duration was prolonged after 7 d in culture. Conclusions: A workflow for rabbit myocardial culture has been established, preserving function for up to 7 d. This research underscores the importance of glucocorticoid signaling in maintaining tissue function and extending culture duration. Furthermore, BDM and CytoD appear to protect from tissue damage during the initiation phase of tissue culture.
Collapse
Affiliation(s)
- V. Baron
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - S. T. Sommer
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - D. J. Fiegle
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - A.-K. M. Pfeuffer
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - R. Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - T. Volk
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - T. Seidel
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
7
|
Pitoulis FG, Smith JJ, Pamias‐Lopez B, de Tombe PP, Hayman D, Terracciano CM. MyoLoop: Design, development and validation of a standalone bioreactor for pathophysiological electromechanical in vitro cardiac studies. Exp Physiol 2024; 109:405-415. [PMID: 37847495 PMCID: PMC10988681 DOI: 10.1113/ep091247] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 10/06/2023] [Indexed: 10/18/2023]
Abstract
Mechanical load is one of the main determinants of cardiac structure and function. Mechanical load is studied in vitro using cardiac preparations together with loading protocols (e.g., auxotonic, isometric). However, such studies are often limited by reductionist models and poorly simulated mechanical load profiles. This hinders the physiological relevance of findings. Living myocardial slices have been used to study load in vitro. Living myocardial slices (LMS) are 300-μm-thick intact organotypic preparations obtained from explanted animal or human hearts. They have preserved cellular populations and the functional, structural, metabolic and molecular profile of the tissue from which they are prepared. Using a three-element Windkessel (3EWK) model we previously showed that LMSs can be cultured while performing cardiac work loops with different preload and afterload. Under such conditions, LMSs remodel as a function of the mechanical load applied to them (physiological load, pressure or volume overload). These studies were conducted in commercially available length actuators that had to be extensively modified for culture experiments. In this paper, we demonstrate the design, development and validation of a novel device, MyoLoop. MyoLoop is a bioreactor that can pace, thermoregulate, acquire and process data, and chronically load LMSs and other cardiac tissues in vitro. In MyoLoop, load is parametrised using a 3EWK model, which can be used to recreate physiological and pathological work loops and the remodelling response to these. We believe MyoLoop is the next frontier in basic cardiovascular research enabling reductionist but physiologically relevant in vitro mechanical studies.
Collapse
Affiliation(s)
| | - Jacob J. Smith
- National Heart & Lung InstituteImperial College LondonLondonUK
| | | | - Pieter P. de Tombe
- Department of Physiology and BiophysicsUniversity of Illinois at ChicagoChicagoIllinoisUSA
- Laboratoire “Physiologie Et Médecine Expérimentale du Coeur Et Des Muscles,” PhymedexpINSERM, CNRSMontpellier University, CHU Arnaud de Villeneuve
MontpellierFrance
| | - Danika Hayman
- National Heart & Lung InstituteImperial College LondonLondonUK
| | | |
Collapse
|
8
|
Amesz JH, Langmuur SJJ, Zhang L, Manintveld OC, Schinkel AFL, de Jong PL, de Groot NMS, Taverne YJHJ. Biomechanical response of ultrathin slices of hypertrophic cardiomyopathy tissue to myosin modulator mavacamten. Biomed Pharmacother 2024; 170:116036. [PMID: 38134635 DOI: 10.1016/j.biopha.2023.116036] [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: 09/20/2023] [Revised: 12/07/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common inherited myocardial disorder of the heart, but effective treatment options remain limited. Mavacamten, a direct myosin modulator, has been presented as novel pharmacological therapy for HCM. The aim of this study was to analyze the biomechanical response of HCM tissue to Mavacamten using living myocardial slices (LMS). LMS (n = 58) from patients with HCM (n = 10) were cultured under electromechanical stimulation, and Verapamil and Mavacamten were administered on consecutive days to evaluate their effects on cardiac biomechanics. Mavacamten and Verapamil reduced contractile force and dF/dt and increased time-to-relaxation in a similar manner. Yet, the time-to-peak of the cardiac contraction was prolonged after administration of Mavacamten (221.0 ms (208.8 - 236.3) vs. 237.7 (221.0 - 254.7), p = 0.004). In addition, Mavacamten prolonged the functional refractory period (FRP) (330 ms (304 - 351) vs. 355 ms (313 - 370), p = 0.023) and better preserved twitch force with increasing stimulation frequencies, compared to Verapamil. As such, Mavacamten reduced (hyper-)contractility and prolonged contraction duration of HCM LMS, suggesting a reduction in cardiac wall stress. Also, Mavacamten might protect against the development of ventricular tachyarrhythmias due to prolongation of the FRP, and improve toleration of tachycardia due to better preservation of twitch force at tachycardiac stimulation frequencies.
Collapse
Affiliation(s)
- Jorik H Amesz
- Translational Cardiothoracic Surgery Research Lab, Department of Cardiothoracic Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands; Translational Electrophysiology Lab, Department of Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Cardiothoracic Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Sanne J J Langmuur
- Translational Cardiothoracic Surgery Research Lab, Department of Cardiothoracic Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Cardiothoracic Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Lu Zhang
- Translational Electrophysiology Lab, Department of Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Olivier C Manintveld
- Department of Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Arend F L Schinkel
- Department of Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Peter L de Jong
- Department of Cardiothoracic Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Natasja M S de Groot
- Translational Electrophysiology Lab, Department of Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Yannick J H J Taverne
- Translational Cardiothoracic Surgery Research Lab, Department of Cardiothoracic Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Cardiothoracic Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
9
|
Shi R, Reichardt M, Fiegle DJ, Küpfer LK, Czajka T, Sun Z, Salditt T, Dendorfer A, Seidel T, Bruegmann T. Contractility measurements for cardiotoxicity screening with ventricular myocardial slices of pigs. Cardiovasc Res 2023; 119:2469-2481. [PMID: 37934066 PMCID: PMC10651213 DOI: 10.1093/cvr/cvad141] [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: 11/08/2022] [Revised: 05/22/2023] [Accepted: 07/10/2023] [Indexed: 11/08/2023] Open
Abstract
AIMS Cardiotoxicity is one major reason why drugs do not enter or are withdrawn from the market. Thus, approaches are required to predict cardiotoxicity with high specificity and sensitivity. Ideally, such methods should be performed within intact cardiac tissue with high relevance for humans and detect acute and chronic side effects on electrophysiological behaviour, contractility, and tissue structure in an unbiased manner. Herein, we evaluate healthy pig myocardial slices and biomimetic cultivation setups (BMCS) as a new cardiotoxicity screening approach. METHODS AND RESULTS Pig left ventricular samples were cut into slices and spanned into BMCS with continuous electrical pacing and online force recording. Automated stimulation protocols were established to determine the force-frequency relationship (FFR), frequency dependence of contraction duration, effective refractory period (ERP), and pacing threshold. Slices generated 1.3 ± 0.14 mN/mm2 force at 0.5 Hz electrical pacing and showed a positive FFR and a shortening of contraction duration with increasing pacing rates. Approximately 62% of slices were able to contract for at least 6 days while showing stable ERP, contraction duration-frequency relationship, and preserved cardiac structure confirmed by confocal imaging and X-ray diffraction analysis. We used specific blockers of the most important cardiac ion channels to determine which analysis parameters are influenced. To validate our approach, we tested five drug candidates selected from the Comprehensive in vitro Proarrhythmia Assay list as well as acetylsalicylic acid and DMSO as controls in a blinded manner in three independent laboratories. We were able to detect all arrhythmic drugs and their respective mode of action on cardiac tissue including inhibition of Na+, Ca2+, and hERG channels as well as Na+/Ca2+ exchanger. CONCLUSION We systematically evaluate this approach for cardiotoxicity screening, which is of high relevance for humans and can be upscaled to medium-throughput screening. Thus, our approach will improve the predictive value and efficiency of preclinical cardiotoxicity screening.
Collapse
Affiliation(s)
- Runzhu Shi
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Humboldtallee 23, 37073 Göttingen, Göttingen, Germany
- International Research Training Group 1816, University Medical Center Göttingen, Göttingen, Germany
| | - Marius Reichardt
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Humboldtallee 23, 37073 Göttingen, Göttingen, Germany
- Institute for X-ray Physics, University of Göttingen, Göttingen, Germany
| | - Dominik J Fiegle
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Linda K Küpfer
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Titus Czajka
- Institute for X-ray Physics, University of Göttingen, Göttingen, Germany
| | - Zhengwu Sun
- Walter-Brendel-Centre of Experimental Medicine, Hospital of the University Munich, Munich, Germany
| | - Tim Salditt
- Institute for X-ray Physics, University of Göttingen, Göttingen, Germany
- Cluster of Excellence ‘Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells’ (MBExC), University of Göttingen, Göttingen, Germany
| | - Andreas Dendorfer
- Walter-Brendel-Centre of Experimental Medicine, Hospital of the University Munich, Munich, Germany
- German Centre of Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Thomas Seidel
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Tobias Bruegmann
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Humboldtallee 23, 37073 Göttingen, Göttingen, Germany
- Cluster of Excellence ‘Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells’ (MBExC), University of Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Göttingen, Göttingen, Germany
| |
Collapse
|
10
|
Ross AJ, Krumova I, Tunc B, Wu Q, Wu C, Camelliti P. A novel method to extend viability and functionality of living heart slices. Front Cardiovasc Med 2023; 10:1244630. [PMID: 37881724 PMCID: PMC10597746 DOI: 10.3389/fcvm.2023.1244630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 08/09/2023] [Indexed: 10/27/2023] Open
Abstract
Living heart slices have recently emerged as a powerful experimental model for fundamental cardiac research. By retaining the structure and function of the native myocardium while maintaining the simplicity of cell culture models, heart slices can be easily employed in electrophysiological, pharmacological, biochemical, and structural investigations. One single heart yields many slices (>20 slices for rodents, >100 slices for porcine or human hearts), however due to the low throughput of most assays and rapid slice degeneration within 24 h of preparation, many slices remain unused and are discarded at the end of the preparation day. Here we present a novel method to extend viability and functionality of living heart slices, enabling their use in experiments over several consecutive days following preparation. By combining hypothermic conditions with inhibition of myosin II ATPase using 2,3-butanedione monoxime (BDM), slices prepared from the left ventricle of porcine hearts remain viable and exhibit preserved contractile function and morphology for up to 6 days. Electrophysiological function was also confirmed over the 6 days by extracellular field potentials recordings. This simple method not only maximizes the use of slices prepared from one single heart, thus reducing the number of animals required, but also increases data reproducibility by allowing multiple electrophysiological, pharmacological, biochemical, and structural studies to be performed from the same heart.
Collapse
Affiliation(s)
- Abigail J. Ross
- School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| | - Iva Krumova
- School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| | - Berfin Tunc
- School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| | - Qin Wu
- School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
- School of Medicine, Jiangsu Vocational College of Medicine, Yancheng, China
| | - Changhao Wu
- School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| | - Patrizia Camelliti
- School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
11
|
Chabanovska O, Lemcke H, Lang H, Vollmar B, Dohmen PM, David R, Etz C, Neßelmann C. Sarcomeric network analysis of ex vivo cultivated human atrial appendage tissue using super-resolution microscopy. Sci Rep 2023; 13:13041. [PMID: 37563225 PMCID: PMC10415305 DOI: 10.1038/s41598-023-39962-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 08/02/2023] [Indexed: 08/12/2023] Open
Abstract
Investigating native human cardiac tissue with preserved 3D macro- and microarchitecture is fundamental for clinical and basic research. Unfortunately, the low accessibility of the human myocardium continues to limit scientific progress. To overcome this issue, utilizing atrial appendages of the human heart may become highly beneficial. Atrial appendages are often removed during open-heart surgery and can be preserved ex vivo as living tissue with varying durability depending on the culture method. In this study, we prepared living thin myocardial slices from left atrial appendages that were cultured using an air-liquid interface system for overall 10 days. Metabolic activity of the cultured slices was assessed using a conventional methyl thiazolyl tetrazolium (MTT) assay. To monitor the structural integrity of cardiomyocytes within the tissue, we implemented our recently described super-resolution microscopy approach that allows both qualitative and quantitative in-depth evaluation of sarcomere network based on parameters such as overall sarcomere content, filament size and orientation. Additionally, expression of mRNAs coding for key structural and functional proteins was analyzed by real-time reverse transcription polymerase chain reaction (qRT-PCR). Our findings demonstrate highly significant disassembly of contractile apparatus represented by degradation of [Formula: see text]-actinin filaments detected after three days in culture, while metabolic activity was constantly rising and remained high for up to seven days. However, gene expression of crucial cardiac markers strongly decreased after the first day in culture indicating an early destructive response to ex vivo conditions. Therefore, we suggest static cultivation of living myocardial slices derived from left atrial appendage and prepared according to our protocol only for short-termed experiments (e.g. medicinal drug testing), while introduction of electro-mechanical stimulation protocols may offer the possibility for long-term integrity of such constructs.
Collapse
Affiliation(s)
- Oleksandra Chabanovska
- Reference and Translation Center for Cardiac Stem Cell therapy (RTC), Department of Cardiac Surgery, Rostock University Medical Center, 18057, Rostock, Germany
- Department of Life, Light, and Matter of the Interdisciplinary Faculty, Rostock University, 18059, Rostock, Germany
- Department of Operative Dentistry and Periodontology, Rostock University Medical Center, 18059, Rostock, Germany
| | - Heiko Lemcke
- Reference and Translation Center for Cardiac Stem Cell therapy (RTC), Department of Cardiac Surgery, Rostock University Medical Center, 18057, Rostock, Germany
- Department of Life, Light, and Matter of the Interdisciplinary Faculty, Rostock University, 18059, Rostock, Germany
| | - Hermann Lang
- Department of Operative Dentistry and Periodontology, Rostock University Medical Center, 18059, Rostock, Germany
| | - Brigitte Vollmar
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18059, Rostock, Germany
| | - Pascal M Dohmen
- Department of Cardiac Surgery, Rostock University Medical Center, 18059, Rostock, Germany
- Department of Cardiothoracic Surgery, Faculty of Health Science, University of the Free State, Bloemfontein, 9301, South Africa
| | - Robert David
- Reference and Translation Center for Cardiac Stem Cell therapy (RTC), Department of Cardiac Surgery, Rostock University Medical Center, 18057, Rostock, Germany.
- Department of Life, Light, and Matter of the Interdisciplinary Faculty, Rostock University, 18059, Rostock, Germany.
| | - Christian Etz
- Department of Cardiac Surgery, Rostock University Medical Center, 18059, Rostock, Germany
| | - Catharina Neßelmann
- Department of Cardiac Surgery, Rostock University Medical Center, 18059, Rostock, Germany
| |
Collapse
|
12
|
Poch CM, Dendorfer A, Laugwitz KL, Moretti A. Ex vivo Culture and Contractile Force Measurements of Non-human Primate Heart Slices. Bio Protoc 2023; 13:e4750. [PMID: 37456341 PMCID: PMC10338344 DOI: 10.21769/bioprotoc.4750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/15/2023] [Accepted: 05/22/2023] [Indexed: 07/18/2023] Open
Abstract
Cardiovascular diseases are the leading cause of death and morbidity worldwide. Patient mortality has been successfully reduced by nearly half in the last four decades, mainly due to advances in minimally invasive surgery techniques and interventional cardiology methods. However, a major hurdle is still the translational gap between preclinical findings and the conversion into effective therapies, which is partly due to the use of model systems that fail to recapitulate key aspects of human physiology and disease. Large animal models such as pigs and non-human primates are highly valuable because they closely resemble humans but are costly and time intensive. Here, we provide a method for long-term ex vivo culture of non-human primate (NHP) myocardial tissue that offers a powerful alternative for a wide range of applications including electrophysiology studies, drug screening, and gene function analyses.
Collapse
Affiliation(s)
- Christine M. Poch
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Andreas Dendorfer
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Germany
- DZHK (German Centre of Cardiovascular Research) – Munich Heart Alliance, Munich, Germany
| | - Karl-Ludwig Laugwitz
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- DZHK (German Centre of Cardiovascular Research) – Munich Heart Alliance, Munich, Germany
| | - Alessandra Moretti
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- DZHK (German Centre of Cardiovascular Research) – Munich Heart Alliance, Munich, Germany
| |
Collapse
|
13
|
Dokuchaev A, Kursanov A, Balakina-Vikulova NA, Katsnelson LB, Solovyova O. The importance of mechanical conditions in the testing of excitation abnormalities in a population of electro-mechanical models of human ventricular cardiomyocytes. Front Physiol 2023; 14:1187956. [PMID: 37362439 PMCID: PMC10285544 DOI: 10.3389/fphys.2023.1187956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/25/2023] [Indexed: 06/28/2023] Open
Abstract
Background: Populations of in silico electrophysiological models of human cardiomyocytes represent natural variability in cell activity and are thoroughly calibrated and validated using experimental data from the human heart. The models have been shown to predict the effects of drugs and their pro-arrhythmic risks. However, excitation and contraction are known to be tightly coupled in the myocardium, with mechanical loads and stretching affecting both mechanics and excitation through mechanisms of mechano-calcium-electrical feedback. However, these couplings are not currently a focus of populations of cell models. Aim: We investigated the role of cardiomyocyte mechanical activity under different mechanical conditions in the generation, calibration, and validation of a population of electro-mechanical models of human cardiomyocytes. Methods: To generate a population, we assumed 11 input parameters of ionic currents and calcium dynamics in our recently developed TP + M model as varying within a wide range. A History matching algorithm was used to generate a non-implausible parameter space by calibrating the action potential and calcium transient biomarkers against experimental data and rejecting models with excitation abnormalities. The population was further calibrated using experimental data on human myocardial force characteristics and mechanical tests involving variations in preload and afterload. Models that passed the mechanical tests were validated with additional experimental data, including the effects of drugs with high or low pro-arrhythmic risk. Results: More than 10% of the models calibrated on electrophysiological data failed mechanical tests and were rejected from the population due to excitation abnormalities at reduced preload or afterload for cell contraction. The final population of accepted models yielded action potential, calcium transient, and force/shortening outputs consistent with experimental data. In agreement with experimental and clinical data, the models demonstrated a high frequency of excitation abnormalities in simulations of Dofetilide action on the ionic currents, in contrast to Verapamil. However, Verapamil showed a high frequency of failed contractions at high concentrations. Conclusion: Our results highlight the importance of considering mechanoelectric coupling in silico cardiomyocyte models. Mechanical tests allow a more thorough assessment of the effects of interventions on cardiac function, including drug testing.
Collapse
Affiliation(s)
- Arsenii Dokuchaev
- Laboratory of Mathematical Physiology, Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, Ekaterinburg, Russia
| | - Alexander Kursanov
- Laboratory of Mathematical Physiology, Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, Ekaterinburg, Russia
- Laboratory of Mathematical Modeling in Physiology and Medicine Based on Supercomputers, Ural Federal University, Ekaterinburg, Russia
| | - Nathalie A. Balakina-Vikulova
- Laboratory of Mathematical Physiology, Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, Ekaterinburg, Russia
- Laboratory of Mathematical Modeling in Physiology and Medicine Based on Supercomputers, Ural Federal University, Ekaterinburg, Russia
| | - Leonid B. Katsnelson
- Laboratory of Mathematical Physiology, Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, Ekaterinburg, Russia
- Laboratory of Mathematical Modeling in Physiology and Medicine Based on Supercomputers, Ural Federal University, Ekaterinburg, Russia
| | - Olga Solovyova
- Laboratory of Mathematical Physiology, Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, Ekaterinburg, Russia
- Laboratory of Mathematical Modeling in Physiology and Medicine Based on Supercomputers, Ural Federal University, Ekaterinburg, Russia
| |
Collapse
|
14
|
Biomimetic cultivation of atrial tissue slices as novel platform for in-vitro atrial arrhythmia studies. Sci Rep 2023; 13:3648. [PMID: 36871094 PMCID: PMC9985600 DOI: 10.1038/s41598-023-30688-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Living myocardial slices (LMS) are beating sections of intact human myocardium that maintain 3D microarchitecture and multicellularity, thereby overcoming most limitations of conventional myocardial cell cultures. We introduce a novel method to produce LMS from human atria and apply pacing modalities to bridge the gap between in-vitro and in-vivo atrial arrhythmia studies. Human atrial biopsies from 15 patients undergoing cardiac surgery were dissected to tissue blocks of ~ 1 cm2 and cut to 300 µm thin LMS with a precision-cutting vibratome. LMS were placed in a biomimetic cultivation chamber, filled with standard cell culture medium, under diastolic preload (1 mN) and continuous electrical stimulation (1000 ms cycle length (CL)), resulting in 68 beating LMS. Atrial LMS refractory period was determined at 192 ± 26 ms. Fixed rate pacing with a CL of 333 ms was applied as atrial tachyarrhythmia (AT) model. This novel state-of-the-art platform for AT research can be used to investigate arrhythmia mechanisms and test novel therapies.
Collapse
|
15
|
ActionPytential: An open source tool for analyzing and visualizing cardiac action potential data. Heliyon 2023; 9:e14440. [PMID: 36967904 PMCID: PMC10031321 DOI: 10.1016/j.heliyon.2023.e14440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/14/2023] Open
Abstract
The action potential forms the basis of cardiac pacemaking, conduction, and contraction. Action potentials can be recorded from numerous preparation types, including ventricular or atrial trabecules, Purkinje fibers, isolated cardiac myocytes. Numerous techniques are also available as well, such as the conventional microelectrode and the single-cell current clamp techniques, optical mapping, or in silico modeling. With such a vast array of electrophysiological methods comes an array of available hardware and software solutions. In this work, we present a software with an intuitive graphical user interface, ActionPytential, that enables the analysis of any type of cardiac action potential, regardless of acquisition method or tissue type. In most available software tools, the analysis of continuous (gap-free) recordings often requires manual user interaction to segment the individual action potentials. We provide an automated solution for this, both for slow-response and for externally paced action potentials. As of now, ActionPytential calculates 34 parameters from each action potential. The most often utilized ones, including amplitude, maximal rate of depolarization, and action potential duration values, were validated on 1200 action potentials from human, dog, rabbit, guinea pig, and rat cardiac preparations. We also provide new parameters that were previously only measurable manually, including the position and the depth of the notch in potentials showing a spike-and-dome morphology. Further notable features include a Butterworth-type low-pass filter, the averaging of multiple potentials, automated corrections for baseline drifting, aided manual analysis, high-quality plots, and batch processing for any number of potentials. ActionPytential is available for all major platforms (Windows, MacOS, GNU + Linux, BSD).
Collapse
|
16
|
Optimized Conditions for the Long-Term Maintenance of Precision-Cut Murine Myocardium in Biomimetic Tissue Culture. Bioengineering (Basel) 2023; 10:bioengineering10020171. [PMID: 36829664 PMCID: PMC9952453 DOI: 10.3390/bioengineering10020171] [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: 11/14/2022] [Revised: 01/11/2023] [Accepted: 01/23/2023] [Indexed: 02/03/2023] Open
Abstract
Organotypic heart slices from mice might provide a promising in vitro model for cardiac research because of the vast availability of genetically modified specimens, combined with the unrestricted feasibility of experimental interventions. However, murine heart slices undergo rapid degeneration in culture. Therefore, we developed optimal conditions to preserve their structure and function in culture. Mouse ventricular heart samples were transversely cut into 300 µm thick slices. Slices were then cultured under various conditions of diastolic preload, systolic compliance and medium agitation. Continuous stimulation was performed either by optical stimulation or by electrical field stimulation. Contractility was continuously measured, and cellular survival, structure and gene expression were analyzed. Significant improvements in viability and function were achieved by elastic fixation with the appropriate diastolic preload and the rapid shaking of a ß-mercaptoethanol-supplemented medium. At 1 Hz pacing, mouse heart slices maintained stable contractility for up to 48 h under optogenetic pacing and for one week under electrical pacing. In cultured slices, the native myofibril structure was well preserved, and the mRNAs of myosin light chain, titin and connexin 43 were constantly expressed. Conclusions: Adult murine heart slices can be preserved for one week and provide a new opportunity to study cardiac functions.
Collapse
|
17
|
Amesz JH, Zhang L, Everts BR, De Groot NMS, Taverne YJHJ. Living myocardial slices: Advancing arrhythmia research. Front Physiol 2023; 14:1076261. [PMID: 36711023 PMCID: PMC9880234 DOI: 10.3389/fphys.2023.1076261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 01/04/2023] [Indexed: 01/15/2023] Open
Abstract
Living myocardial slices (LMS) are ultrathin (150-400 µm) sections of intact myocardium that can be used as a comprehensive model for cardiac arrhythmia research. The recent introduction of biomimetic electromechanical cultivation chambers enables long-term cultivation and easy control of living myocardial slices culture conditions. The aim of this review is to present the potential of this biomimetic interface using living myocardial slices in electrophysiological studies outlining advantages, disadvantages and future perspectives of the model. Furthermore, different electrophysiological techniques and their application on living myocardial slices will be discussed. The developments of living myocardial slices in electrophysiology research will hopefully lead to future breakthroughs in the understanding of cardiac arrhythmia mechanisms and the development of novel therapeutic options.
Collapse
Affiliation(s)
- Jorik H. Amesz
- Translational Cardiothoracic Surgery Research Lab, Lowlands Institute for Bioelectric Medicine, Department of Cardiothoracic Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
- Translational Electrophysiology, Lowlands Institute for Bioelectric Medicine, Department of Cardiology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Lu Zhang
- Translational Electrophysiology, Lowlands Institute for Bioelectric Medicine, Department of Cardiology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Bian R. Everts
- Translational Cardiothoracic Surgery Research Lab, Lowlands Institute for Bioelectric Medicine, Department of Cardiothoracic Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Natasja M. S. De Groot
- Translational Electrophysiology, Lowlands Institute for Bioelectric Medicine, Department of Cardiology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Yannick J. H. J. Taverne
- Translational Cardiothoracic Surgery Research Lab, Lowlands Institute for Bioelectric Medicine, Department of Cardiothoracic Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
18
|
Hofmann J, Pühringer M, Steinkellner S, Holl AS, Meszaros AT, Schneeberger S, Troppmair J, Hautz T. Novel, Innovative Models to Study Ischemia/Reperfusion-Related Redox Damage in Organ Transplantation. Antioxidants (Basel) 2022; 12:antiox12010031. [PMID: 36670893 PMCID: PMC9855021 DOI: 10.3390/antiox12010031] [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: 12/03/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
The implementation of ex vivo organ machine perfusion (MP) into clinical routine undoubtedly helped to increase the donor pool. It enables not just organ assessment, but potentially regeneration and treatment of marginal organs in the future. During organ procurement, redox-stress triggered ischemia-reperfusion injury (IRI) is inevitable, which in addition to pre-existing damage negatively affects such organs. Ex vivo MP enables to study IRI-associated tissue damage and its underlying mechanisms in a near to physiological setting. However, research using whole organs is limited and associated with high costs. Here, in vitro models well suited for early stage research or for studying particular disease mechanisms come into play. While cell lines convince with simplicity, they do not exert all organ-specific functions. Tissue slice cultures retain the three-dimensional anatomical architecture and cells remain within their naïve tissue-matrix configuration. Organoids may provide an even closer modelling of physiologic organ function and spatial orientation. In this review, we discuss the role of oxidative stress during ex vivo MP and the suitability of currently available in vitro models to further study the underlying mechanisms and to pretest potential treatment strategies.
Collapse
|
19
|
Zabielska-Kaczorowska MA, Bogucka AE, Macur K, Czaplewska P, Watson SA, Perbellini F, Terracciano CM, Smolenski RT. Label-free quantitative SWATH-MS proteomic analysis of adult myocardial slices in vitro after biomimetic electromechanical stimulation. Sci Rep 2022; 12:16533. [PMID: 36192624 PMCID: PMC9529937 DOI: 10.1038/s41598-022-20494-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/14/2022] [Indexed: 11/24/2022] Open
Abstract
A special in vitro model maintained with ultrathin cardiac slices with a preserved architecture, multi-cellularity, and physiology of the heart tissue was used. In our experiments, we performed label-free quantitative SWATH-MS proteomic analysis of the adult myocardial slices in vitro after biomimetic electromechanical stimulation. Rat myocardial slices were stretched to sarcomere lengths (SL) within the physiological range of 1.8–2.2 μm. Electromechanically stimulated slices were compared with slices cultured without electromechanical stimulation (unloaded and nonstimulated-TW) on a liquid–air interface and with fresh myocardial slices (0 h-C). Quantitative (relative) proteomic analyses were performed using a label-free SWATH-MS technique on a high-resolution microLC-MS/MS TripleTOF 5600+ system (SCIEX). The acquired MS/MS spectra from the DDA LC–MS/MS analyses of the rat heart samples were searched against the UniProt Rattus norvegicus database (version of 15.05.2018) using the Paragon algorithm incorporated into ProteinPilot 4.5 (SCIEX) software. The highest number of differential proteins was observed in the TW group—121 when compared to the C group. In the 1.8 and 2.2 groups, 79 and 52 proteins present at a significantly different concentration from the control samples were found, respectively. A substantial fraction of these proteins were common for two or more comparisons, resulting in a list of 169 significant proteins for at least one of the comparisons. This study found the most prominent changes in the proteomic pattern related to mitochondrial respiration, energy metabolism, and muscle contraction in the slices that were stretched and fresh myocardial slices cultured without electromechanical stimulation.
Collapse
Affiliation(s)
- M A Zabielska-Kaczorowska
- Department of Biochemistry, Medical University of Gdansk, Gdansk, Poland. .,Department of Physiology, Medical University of Gdansk, Gdansk, Poland.
| | - A E Bogucka
- Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland.,Institute of Biochemistry, Medical Faculty, Justus Liebig University of Giessen, Giessen, Germany
| | - K Macur
- Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - P Czaplewska
- Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - S A Watson
- National Heart & Lung Institute, Imperial College London, London, UK
| | - F Perbellini
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Hannover, Germany
| | - C M Terracciano
- National Heart & Lung Institute, Imperial College London, London, UK
| | - R T Smolenski
- Department of Biochemistry, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
20
|
Kałużna E, Nadel A, Zimna A, Rozwadowska N, Kolanowski T. Modeling the human heart ex vivo-current possibilities and strive for future applications. J Tissue Eng Regen Med 2022; 16:853-874. [PMID: 35748158 PMCID: PMC9796015 DOI: 10.1002/term.3335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/20/2022] [Accepted: 06/03/2022] [Indexed: 12/30/2022]
Abstract
The high organ specification of the human heart is inversely proportional to its functional recovery after damage. The discovery of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) has accelerated research in human heart regeneration and physiology. Nevertheless, due to the immaturity of iPSC-CMs, they are far from being an representative model of the adult heart physiology. Therefore, number of laboratories strive to obtain a heart tissues by engineering methods by structuring iPSC-CMs into complex and advanced platforms. By using the iPSC-CMs and arranging them in 3D cultures it is possible to obtain a human heart muscle with physiological capabilities potentially similar to the adult heart, while remaining in vitro. Here, we attempt to describe existing examples of heart muscle either in vitro or ex vivo models and discuss potential options for the further development of such structures. This will be a crucial step for ultimate derivation of complete heart tissue-mimicking organs and their future use in drug development, therapeutic approaches testing, pre-clinical studies, and clinical applications. This review particularly aims to compile available models of advanced human heart tissue for scientists considering which model would best fit their research needs.
Collapse
Affiliation(s)
- Ewelina Kałużna
- Institute of Human GeneticsPolish Academy of SciencesPoznanPoland
| | - Agnieszka Nadel
- Institute of Human GeneticsPolish Academy of SciencesPoznanPoland
| | - Agnieszka Zimna
- Institute of Human GeneticsPolish Academy of SciencesPoznanPoland
| | | | | |
Collapse
|
21
|
Maselli D, Matos RS, Johnson RD, Martella D, Caprettini V, Chiappini C, Camelliti P, Campagnolo P. Porcine Organotypic Epicardial Slice Protocol: A Tool for the Study of Epicardium in Cardiovascular Research. Front Cardiovasc Med 2022; 9:920013. [PMID: 35924218 PMCID: PMC9339655 DOI: 10.3389/fcvm.2022.920013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
The epicardium has recently gained interest in the cardiovascular field due to its capacity to support heart regeneration after ischemic injury. Models to study the epicardium of large animals in vitro are limited and mainly based on epicardial cell isolation/differentiation from stem cells, followed by 2D cells culture. In this method paper, we describe the procedure to obtain and culture 3D organotypic heart slices presenting an intact epicardium, as a novel model to study the epicardial physiology and activation. Epicardial slices are obtained from porcine hearts using a high-precision vibratome and retain a healthy epicardial layer embedded in its native extracellular environment and connected with other cardiac cells (cardiomyocytes, fibroblasts, vascular cells etc.). Epicardial slices can be cultured for 72 h, providing an ideal model for studying the epicardium physiology or perform pharmacological interventions/gene therapy approaches. We also report on methods to assesses the viability and composition of the epicardial slices, and evaluate their architecture in 3D through tissue decoloration. Finally, we present a potential application for a nanomaterial-based gene transfer method for tracking of epicardial cells within the slice. Crucially, given the similarity in morphology and physiology of porcine heart with its human counterpart, our system provides a platform for translational research while providing a clinically relevant and ethical alternative to the use of small animals in this type of research.
Collapse
Affiliation(s)
- Davide Maselli
- Cardiovascular Section, Department of Biochemical Sciences, University of Surrey, Guildford, United Kingdom
| | - Rolando S. Matos
- Cardiovascular Section, Department of Biochemical Sciences, University of Surrey, Guildford, United Kingdom
| | - Robert D. Johnson
- Cardiovascular Section, Department of Biochemical Sciences, University of Surrey, Guildford, United Kingdom
| | - Davide Martella
- London Centre for Nanotechnology, King's College London, London, United Kingdom
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Valeria Caprettini
- London Centre for Nanotechnology, King's College London, London, United Kingdom
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Ciro Chiappini
- London Centre for Nanotechnology, King's College London, London, United Kingdom
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Patrizia Camelliti
- Cardiovascular Section, Department of Biochemical Sciences, University of Surrey, Guildford, United Kingdom
| | - Paola Campagnolo
- Cardiovascular Section, Department of Biochemical Sciences, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
22
|
Visualization of regenerating and repairing hearts. Clin Sci (Lond) 2022; 136:787-798. [PMID: 35621122 PMCID: PMC9886236 DOI: 10.1042/cs20211116] [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: 03/04/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 02/01/2023]
Abstract
With heart failure continuing to become more prevalent, investigating the mechanisms of heart injury and repair holds much incentive. In contrast with adult mammals, other organisms such as teleost fish, urodele amphibians, and even neonatal mammals are capable of robust cardiac regeneration to replenish lost or damaged myocardial tissue. Long-term high-resolution intravital imaging of the behaviors and interactions of different cardiac cell types in their native environment could yield unprecedented insights into heart regeneration and repair. However, this task remains challenging for the heart due to its rhythmic contraction and anatomical location. Here, we summarize recent advances in live imaging of heart regeneration and repair, discuss the advantages and limitations of current systems, and suggest future directions for novel imaging technology development.
Collapse
|
23
|
Huebsch N, Charrez B, Neiman G, Siemons B, Boggess SC, Wall S, Charwat V, Jæger KH, Cleres D, Telle Å, Lee-Montiel FT, Jeffreys NC, Deveshwar N, Edwards AG, Serrano J, Snuderl M, Stahl A, Tveito A, Miller EW, Healy KE. Metabolically driven maturation of human-induced-pluripotent-stem-cell-derived cardiac microtissues on microfluidic chips. Nat Biomed Eng 2022; 6:372-388. [PMID: 35478228 PMCID: PMC10344596 DOI: 10.1038/s41551-022-00884-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 03/14/2022] [Indexed: 12/29/2022]
Abstract
The immature physiology of cardiomyocytes derived from human induced pluripotent stem cells (hiPSCs) limits their utility for drug screening and disease modelling. Here we show that suitable combinations of mechanical stimuli and metabolic cues can enhance the maturation of hiPSC-derived cardiomyocytes, and that the maturation-inducing cues have phenotype-dependent effects on the cells' action-potential morphology and calcium handling. By using microfluidic chips that enhanced the alignment and extracellular-matrix production of cardiac microtissues derived from genetically distinct sources of hiPSC-derived cardiomyocytes, we identified fatty-acid-enriched maturation media that improved the cells' mitochondrial structure and calcium handling, and observed divergent cell-source-dependent effects on action-potential duration (APD). Specifically, in the presence of maturation media, tissues with abnormally prolonged APDs exhibited shorter APDs, and tissues with aberrantly short APDs displayed prolonged APDs. Regardless of cell source, tissue maturation reduced variabilities in spontaneous beat rate and in APD, and led to converging cell phenotypes (with APDs within the 300-450 ms range characteristic of human left ventricular cardiomyocytes) that improved the modelling of the effects of pro-arrhythmic drugs on cardiac tissue.
Collapse
Affiliation(s)
- Nathaniel Huebsch
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA
- Department of Biomedical Engineering, The Washington University in Saint Louis, Saint Louis, MO, USA
| | - Berenice Charrez
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA
| | - Gabriel Neiman
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA
| | - Brian Siemons
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA
| | - Steven C Boggess
- Department of Chemistry, University of California at Berkeley, Berkeley, CA, USA
| | | | - Verena Charwat
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA
| | | | - David Cleres
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA
| | | | - Felipe T Lee-Montiel
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA
| | - Nicholas C Jeffreys
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA
| | - Nikhil Deveshwar
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA
| | - Andrew G Edwards
- Department of Pharmacology, School of Medicine, University of California, Davis, CA, USA
| | - Jonathan Serrano
- Department of Pathology, New York University Langone Health and Medical School, New York, NY, USA
| | - Matija Snuderl
- Department of Pathology, New York University Langone Health and Medical School, New York, NY, USA
| | - Andreas Stahl
- Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA, USA
| | | | - Evan W Miller
- Department of Chemistry, University of California at Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Kevin E Healy
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA.
- Department of Materials Science and Engineering, University of California at Berkeley, Berkeley, CA, USA.
| |
Collapse
|
24
|
Klumm MJ, Heim C, Fiegle DJ, Weyand M, Volk T, Seidel T. Long-Term Cultivation of Human Atrial Myocardium. Front Physiol 2022; 13:839139. [PMID: 35283779 PMCID: PMC8905341 DOI: 10.3389/fphys.2022.839139] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 01/25/2022] [Indexed: 11/16/2022] Open
Abstract
Organotypic culture of human ventricular myocardium is emerging in basic and translational cardiac research. However, few institutions have access to human ventricular tissue, whereas atrial tissue is more commonly available and important for studying atrial physiology. This study presents a method for long-term cultivation of beating human atrial myocardium. After written informed consent, tissues from the right-atrial appendage were obtained from patients with sinus rhythm undergoing open heart surgery with cardiopulmonary bypass. Trabeculae (pectinate muscles) prepared from the samples were installed into cultivation chambers at 37°C with a diastolic preload of 500 μN. After 2 days with 0.5 Hz pacing, stimulation frequency was set to 1 Hz. Contractile force was monitored continuously. Beta-adrenergic response, refractory period (RP) and maximum captured frequency (fmax) were assessed periodically. After cultivation, viability and electromechanical function were investigated, as well as the expression of several genes important for intracellular Ca2+ cycling and electrophysiology. Tissue microstructure was analyzed by confocal microscopy. We cultivated 19 constantly beating trabeculae from 8 patient samples for 12 days and 4 trabeculae from 3 specimen for 21 days. Functional parameters were compared directly after installation (0 d) with those after 12 d in culture. Contraction force was 384 ± 69 μN at 0 d and 255 ± 90 μN at 12 d (p = 0.8, n = 22), RP 480 ± 97 ms and 408 ± 78 ms (p = 0.3, n = 9), fmax 3.0 ± 0.5 Hz and 3.8 ± 0.5 Hz (p = 0.18, n = 9), respectively. Application of 100 nM isoprenaline to 11 trabeculae at 7 d increased contraction force from 168 ± 35 μN to 361 ± 60 μN (p < 0.01), fmax from 6.4 ± 0.6 Hz to 8.5 ± 0.4 Hz (p < 0.01) and lowered RP from 319 ± 22 ms to 223 ± 15 ms. CACNA1c (L-type Ca2+ channel subunit) and GJA1 (connexin-43) mRNA expressions were not significantly altered at 12 d vs 0 d, while ATP2A (SERCA) and KCNJ4 (Kir2.3) were downregulated, and KCNJ2 (Kir2.1) was upregulated. Simultaneous Ca2+ imaging and force recording showed preserved excitation-contraction coupling in cultivated trabeculae. Confocal microscopy indicated preserved cardiomyocyte structure, unaltered amounts of extracellular matrix and gap junctions. MTT assays confirmed viability at 12 d. We established a workflow that allows for stable cultivation and functional analysis of beating human atrial myocardium for up to 3 weeks. This method may lead to novel insights into the physiology and pathophysiology of human atrial myocardium.
Collapse
Affiliation(s)
- Maximilian J Klumm
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Cardiac Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Heim
- Department of Cardiac Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Dominik J Fiegle
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Weyand
- Department of Cardiac Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Tilmann Volk
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Seidel
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
25
|
Maselli D, Matos RS, Johnson RD, Chiappini C, Camelliti P, Campagnolo P. Epicardial slices: an innovative 3D organotypic model to study epicardial cell physiology and activation. NPJ Regen Med 2022; 7:7. [PMID: 35039552 PMCID: PMC8764051 DOI: 10.1038/s41536-021-00202-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 11/30/2021] [Indexed: 11/26/2022] Open
Abstract
The epicardium constitutes an untapped reservoir for cardiac regeneration. Upon heart injury, the adult epicardium re-activates, leading to epithelial-to-mesenchymal transition (EMT), migration, and differentiation. While interesting mechanistic and therapeutic findings arose from lower vertebrates and rodent models, the introduction of an experimental system representative of large mammals would undoubtedly facilitate translational advancements. Here, we apply innovative protocols to obtain living 3D organotypic epicardial slices from porcine hearts, encompassing the epicardial/myocardial interface. In culture, our slices preserve the in vivo architecture and functionality, presenting a continuous epicardium overlaying a healthy and connected myocardium. Upon thymosin β4 treatment of the slices, the epicardial cells become activated, upregulating epicardial and EMT genes, resulting in epicardial cell mobilization and differentiation into epicardial-derived mesenchymal cells. Our 3D organotypic model enables to investigate the reparative potential of the adult epicardium, offering an advanced tool to explore ex vivo the complex 3D interactions occurring within the native heart environment.
Collapse
Affiliation(s)
- D Maselli
- Faculty of Health & Medical Sciences, School of Biosciences & Medicine, Section of Cardiovascular Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - R S Matos
- Faculty of Health & Medical Sciences, School of Biosciences & Medicine, Section of Cardiovascular Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - R D Johnson
- Faculty of Health & Medical Sciences, School of Biosciences & Medicine, Section of Cardiovascular Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - C Chiappini
- Centre for Craniofacial and Regenerative Biology, King's College London, SE1 9RT, London, United Kingdom
| | - P Camelliti
- Faculty of Health & Medical Sciences, School of Biosciences & Medicine, Section of Cardiovascular Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - P Campagnolo
- Faculty of Health & Medical Sciences, School of Biosciences & Medicine, Section of Cardiovascular Sciences, University of Surrey, Guildford, GU2 7XH, UK.
| |
Collapse
|
26
|
Borysova L, Ng YYH, Wragg ES, Wallis LE, Fay E, Ascione R, Dora KA. High spatial and temporal resolution Ca 2+ imaging of myocardial strips from human, pig and rat. Nat Protoc 2021; 16:4650-4675. [PMID: 34400840 DOI: 10.1038/s41596-021-00590-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 06/14/2021] [Indexed: 11/09/2022]
Abstract
Ca2+ handling within cardiac myocytes underpins coordinated contractile function within the beating heart. This protocol enables high spatial and temporal Ca2+ imaging of ex vivo multicellular myocardial strips. The endocardial surface is retained, and strips of 150-300-µm thickness are dissected, loaded with Ca2+ indicators and mounted within 1.5 h. A list of the equipment and reagents used and the key methodological aspects allowing the use of this technique on strips from any chamber of the mammalian heart are described. We have successfully used this protocol on human, pig and rat biopsy samples. On use of this protocol with intact endocardial endothelium, we demonstrated that the myocytes develop asynchronous spontaneous Ca2+ events, which can be ablated by electrically evoked Ca2+ transients, and subsequently redevelop spontaneously after cessation of stimulation. This protocol thus offers a rapid and reliable method for studying the Ca2+ signaling underpinning cardiomyocyte contraction, in both healthy and diseased tissue.
Collapse
Affiliation(s)
- Lyudmyla Borysova
- Vascular Pharmacology Group, Department of Pharmacology, University of Oxford, Oxford, UK
| | - Y Y Hanson Ng
- Vascular Pharmacology Group, Department of Pharmacology, University of Oxford, Oxford, UK
| | - Edward S Wragg
- Vascular Pharmacology Group, Department of Pharmacology, University of Oxford, Oxford, UK
| | - Lillian E Wallis
- Vascular Pharmacology Group, Department of Pharmacology, University of Oxford, Oxford, UK
| | - Emily Fay
- Vascular Pharmacology Group, Department of Pharmacology, University of Oxford, Oxford, UK
| | - Raimondo Ascione
- Bristol Heart Institute, Bristol Royal Infirmary, University of Bristol, Bristol, UK
| | - Kim A Dora
- Vascular Pharmacology Group, Department of Pharmacology, University of Oxford, Oxford, UK.
| |
Collapse
|
27
|
Suckert T, Nexhipi S, Dietrich A, Koch R, Kunz-Schughart LA, Bahn E, Beyreuther E. Models for Translational Proton Radiobiology-From Bench to Bedside and Back. Cancers (Basel) 2021; 13:4216. [PMID: 34439370 PMCID: PMC8395028 DOI: 10.3390/cancers13164216] [Citation(s) in RCA: 12] [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: 07/14/2021] [Revised: 08/09/2021] [Accepted: 08/17/2021] [Indexed: 12/25/2022] Open
Abstract
The number of proton therapy centers worldwide are increasing steadily, with more than two million cancer patients treated so far. Despite this development, pending questions on proton radiobiology still call for basic and translational preclinical research. Open issues are the on-going discussion on an energy-dependent varying proton RBE (relative biological effectiveness), a better characterization of normal tissue side effects and combination treatments with drugs originally developed for photon therapy. At the same time, novel possibilities arise, such as radioimmunotherapy, and new proton therapy schemata, such as FLASH irradiation and proton mini-beams. The study of those aspects demands for radiobiological models at different stages along the translational chain, allowing the investigation of mechanisms from the molecular level to whole organisms. Focusing on the challenges and specifics of proton research, this review summarizes the different available models, ranging from in vitro systems to animal studies of increasing complexity as well as complementing in silico approaches.
Collapse
Affiliation(s)
- Theresa Suckert
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (T.S.); (S.N.); (A.D.); (L.A.K.-S.)
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sindi Nexhipi
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (T.S.); (S.N.); (A.D.); (L.A.K.-S.)
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01309 Dresden, Germany
| | - Antje Dietrich
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (T.S.); (S.N.); (A.D.); (L.A.K.-S.)
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Robin Koch
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany; (R.K.); (E.B.)
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Leoni A. Kunz-Schughart
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (T.S.); (S.N.); (A.D.); (L.A.K.-S.)
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
| | - Emanuel Bahn
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany; (R.K.); (E.B.)
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
- German Cancer Research Center (DKFZ), Clinical Cooperation Unit Radiation Oncology, 69120 Heidelberg, Germany
| | - Elke Beyreuther
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (T.S.); (S.N.); (A.D.); (L.A.K.-S.)
- Helmholtz-Zentrum Dresden—Rossendorf, Institute of Radiation Physics, 01328 Dresden, Germany
| |
Collapse
|
28
|
Fusco-Allison G, Li DK, Hunter B, Jackson D, Bannon PG, Lal S, O'Sullivan JF. Optimizing the discovery and assessment of therapeutic targets in heart failure with preserved ejection fraction. ESC Heart Fail 2021; 8:3643-3655. [PMID: 34342166 PMCID: PMC8497375 DOI: 10.1002/ehf2.13504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 06/02/2021] [Accepted: 06/21/2021] [Indexed: 01/09/2023] Open
Abstract
There is an urgent need for models that faithfully replicate heart failure with preserved ejection fraction (HFpEF), now recognized as the most common form of heart failure in the world. In vitro approaches have several shortcomings, most notably the immature nature of stem cell‐derived human cardiomyocytes [induced pluripotent stem cells (iPSC)] and the relatively short lifespan of primary cardiomyocytes. Three‐dimensional ‘organoids’ incorporating mature iPSCs with other cell types such as endothelial cells and fibroblasts are a significant advance, but lack the complexity of true myocardium. Animal models can replicate many features of human HFpEF, and rodent models are the most common, and recent attempts to incorporate haemodynamic, metabolic, and ageing contributions are encouraging. Differences relating to species, physiology, heart rate, and heart size are major limitations for rodent models. Porcine models mitigate many of these shortcomings and approximate human physiology more closely, but cost and time considerations limit their potential for widespread use. Ex vivo analysis of failing hearts from animal models offer intriguing possibilities regarding cardiac substrate utilisation, but are ultimately subject to the same constrains as the animal models from which the hearts are obtained. Ex vivo approaches using human myocardial biopsies can uncover new insights into pathobiology leveraging myocardial energetics, substrate turnover, molecular changes, and systolic/diastolic function. In collaboration with a skilled cardiothoracic surgeon, left ventricular endomyocardial biopsies can be obtained at the time of valvular surgery in HFpEF patients. Critically, these tissues maintain their disease phenotype, preserving inter‐relationship of myocardial cells and extracellular matrix. This review highlights a novel approach, where ultra‐thin myocardial tissue slices from human HFpEF hearts can be used to assess changes in myocardial structure and function. We discuss current approaches to modelling HFpEF, describe in detail the novel tissue slice model, expand on exciting opportunities this model provides, and outline ways to improve this model further.
Collapse
Affiliation(s)
- Gabrielle Fusco-Allison
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Heart Research Institute, Newtown, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Desmond K Li
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Heart Research Institute, Newtown, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Benjamin Hunter
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Dan Jackson
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Discipline of Surgery, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Paul G Bannon
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Discipline of Surgery, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Sean Lal
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - John F O'Sullivan
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Heart Research Institute, Newtown, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia.,Faculty of Medicine, TU Dresden, Dresden, Germany
| |
Collapse
|
29
|
Arai K, Kitsuka T, Nakayama K. Scaffold-based and scaffold-free cardiac constructs for drug testing. Biofabrication 2021; 13. [PMID: 34233316 DOI: 10.1088/1758-5090/ac1257] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 07/07/2021] [Indexed: 12/24/2022]
Abstract
The safety and therapeutic efficacy of new drugs are tested in experimental animals. However, besides being a laborious, costly process, differences in drug responses between humans and other animals and potential cardiac adverse effects lead to the discontinued development of new drugs. Thus, alternative approaches to animal tests are needed. Cardiotoxicity and responses of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) to drugs are conventionally evaluated by cell seeding and two-dimensional (2D) culture, which allows measurements of field potential duration and the action potentials of CMs using multielectrode arrays. However, 2D-cultured hiPSC-CMs lack 3D spatial adhesion, and have fewer intercellular and extracellular matrix interactions, as well as different contractile behavior from CMsin vivo. This issue has been addressed using tissue engineering to fabricate three-dimensional (3D) cardiac constructs from hiPSC-CMs culturedin vitro. Tissue engineering can be categorized as scaffold-based and scaffold-free. In scaffold-based tissue engineering, collagen and fibrin gel scaffolds comprise a 3D culture environment in which seeded cells exhibit cardiac-specific functions and drug responses, whereas 3D cardiac constructs fabricated by tissue engineering without a scaffold have high cell density and form intercellular interactions. This review summarizes the characteristics of scaffold-based and scaffold-free cardiac tissue engineering and discusses the applications of fabricated cardiac constructs to drug screening.
Collapse
Affiliation(s)
- Kenichi Arai
- Center for Regenerative Medicine Research, Faculty of Medicine, Saga University, Saga, Japan.,Department of Clinical Biomaterial Applied Science, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Takahiro Kitsuka
- Department of Cardiovascular Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Koichi Nakayama
- Center for Regenerative Medicine Research, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
30
|
Lu K, Seidel T, Cao-Ehlker X, Dorn T, Batcha AMN, Schneider CM, Semmler M, Volk T, Moretti A, Dendorfer A, Tomasi R. Progressive stretch enhances growth and maturation of 3D stem-cell-derived myocardium. Am J Cancer Res 2021; 11:6138-6153. [PMID: 33995650 PMCID: PMC8120210 DOI: 10.7150/thno.54999] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 03/23/2021] [Indexed: 12/22/2022] Open
Abstract
Bio-engineered myocardium has great potential to substitute damaged myocardium and for studies of myocardial physiology and disease, but structural and functional immaturity still implies limitations. Current protocols of engineered heart tissue (EHT) generation fall short of simulating the conditions of postnatal myocardial growth, which are characterized by tissue expansion and increased mechanical load. To investigate whether these two parameters can improve EHT maturation, we developed a new approach for the generation of cardiac tissues based on biomimetic stimulation under application of continuously increasing stretch. Methods: EHTs were generated by assembling cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CM) at high cell density in a low collagen hydrogel. Maturation and growth of the EHTs were induced in a custom-made biomimetic tissue culture system that provided continuous electrical stimulation and medium agitation along with progressive stretch at four different increments. Tissues were characterized after a three week conditioning period. Results: The highest rate of stretch (S3 = 0.32 mm/day) increased force development by 5.1-fold compared to tissue with a fixed length, reaching contractility of 11.28 mN/mm². Importantly, intensely stretched EHTs developed physiological length-dependencies of active and passive forces (systolic/diastolic ratio = 9.47 ± 0.84), and a positive force-frequency relationship (1.25-fold contractility at 180 min-1). Functional markers of stretch-dependent maturation included enhanced and more rapid Ca2+ transients, higher amplitude and upstroke velocity of action potentials, and pronounced adrenergic responses. Stretch conditioned hiPSC-CMs displayed structural improvements in cellular volume, linear alignment, and sarcomere length (2.19 ± 0.1 µm), and an overall upregulation of genes that are specifically expressed in adult cardiomyocytes. Conclusions: With the intention to simulate postnatal heart development, we have established techniques of tissue assembly and biomimetic culture that avoid tissue shrinkage and yield muscle fibers with contractility and compliance approaching the properties of adult myocardium. This study demonstrates that cultivation under progressive stretch is a feasible way to induce growth and maturation of stem cell-derived myocardium. The novel tissue-engineering approach fulfills important requirements of disease modelling and therapeutic tissue replacement.
Collapse
|
31
|
Kreutzer FP, Meinecke A, Schmidt K, Fiedler J, Thum T. Alternative strategies in cardiac preclinical research and new clinical trial formats. Cardiovasc Res 2021; 118:746-762. [PMID: 33693475 PMCID: PMC7989574 DOI: 10.1093/cvr/cvab075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
An efficient and safe drug development process is crucial for the establishment of new drugs on the market aiming to increase quality of life and life-span of our patients. Despite technological advances in the past decade, successful launches of drug candidates per year remain low. We here give an overview about some of these advances and suggest improvements for implementation to boost preclinical and clinical drug development with a focus on the cardiovascular field. We highlight advantages and disadvantages of animal experimentation and thoroughly review alternatives in the field of three-dimensional cell culture as well as preclinical use of spheroids and organoids. Microfluidic devices and their potential as organ-on-a-chip systems, as well as the use of living animal and human cardiac tissues are additionally introduced. In the second part, we examine recent gold standard randomized clinical trials and present possible modifications to increase lead candidate throughput: adaptive designs, master protocols, and drug repurposing. In silico and N-of-1 trials have the potential to redefine clinical drug candidate evaluation. Finally, we briefly discuss clinical trial designs during pandemic times.
Collapse
Affiliation(s)
- Fabian Philipp Kreutzer
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Anna Meinecke
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Kevin Schmidt
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Jan Fiedler
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.,Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.,Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| |
Collapse
|
32
|
Meki MH, Miller JM, Mohamed TMA. Heart Slices to Model Cardiac Physiology. Front Pharmacol 2021; 12:617922. [PMID: 33613292 PMCID: PMC7890402 DOI: 10.3389/fphar.2021.617922] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 01/05/2021] [Indexed: 12/02/2022] Open
Abstract
Translational research in the cardiovascular field is hampered by the unavailability of cardiac models that can recapitulate organ-level physiology of the myocardium. Outside the body, cardiac tissue undergoes rapid dedifferentiation and maladaptation in culture. There is an ever-growing demand for preclinical platforms that allow for accurate, standardized, long-term, and rapid drug testing. Heart slices is an emerging technology that solves many of the problems with conventional myocardial culture systems. Heart slices are thin (<400 µm) slices of heart tissue from the adult ventricle. Several recent studies using heart slices have shown their ability to maintain the adult phenotype for prolonged periods in a multi cell-type environment. Here, we review the current status of cardiac culture systems and highlight the unique advantages offered by heart slices in the light of recent efforts in developing physiologically relevant heart slice culture systems.
Collapse
Affiliation(s)
- Moustafa H. Meki
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY, United States
- Department of Bioengineering, University of Louisville, Louisville, KY, United States
| | - Jessica M. Miller
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY, United States
- Department of Bioengineering, University of Louisville, Louisville, KY, United States
| | - Tamer M. A. Mohamed
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY, United States
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, United States
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
33
|
Li J, Hua Y, Miyagawa S, Zhang J, Li L, Liu L, Sawa Y. hiPSC-Derived Cardiac Tissue for Disease Modeling and Drug Discovery. Int J Mol Sci 2020; 21:E8893. [PMID: 33255277 PMCID: PMC7727666 DOI: 10.3390/ijms21238893] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 12/20/2022] Open
Abstract
Relevant, predictive normal, or disease model systems are of vital importance for drug development. The difference between nonhuman models and humans could contribute to clinical trial failures despite ideal nonhuman results. As a potential substitute for animal models, human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs) provide a powerful tool for drug toxicity screening, modeling cardiovascular diseases, and drug discovery. Here, we review recent hiPSC-CM disease models and discuss the features of hiPSC-CMs, including subtype and maturation and the tissue engineering technologies for drug assessment. Updates from the international multisite collaborators/administrations for development of novel drug discovery paradigms are also summarized.
Collapse
Affiliation(s)
- Junjun Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
- Department of Cell Design for Tissue Construction, Faculty of Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ying Hua
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Jingbo Zhang
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Lingjun Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Li Liu
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
- Department of Design for Tissue Regeneration, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| |
Collapse
|
34
|
Liu Z, Klose K, Neuber S, Jiang M, Gossen M, Stamm C. Comparative analysis of adeno-associated virus serotypes for gene transfer in organotypic heart slices. J Transl Med 2020; 18:437. [PMID: 33208161 PMCID: PMC7673099 DOI: 10.1186/s12967-020-02605-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 11/05/2020] [Indexed: 12/26/2022] Open
Abstract
Background Vectors derived from adeno-associated viruses (AAVs) are widely used for gene transfer both in vitro and in vivo and have gained increasing interest as shuttle systems to deliver therapeutic genes to the heart. However, there is little information on their tissue penetration and cytotoxicity, as well as the optimal AAV serotype for transferring genes to diseased hearts. Therefore, we aimed to establish an organotypic heart slice culture system for mouse left ventricular (LV) myocardium and use this platform to analyze gene transfer efficiency, cell tropism, and toxicity of different AAV serotypes. Methods LV tissue slices, 300 µm thick, were prepared from 15- to 17-day-old transgenic alpha-myosin heavy-chain-mCherry mice using a vibrating microtome. Tissue slice viability in air-liquid culture was evaluated by calcein-acetoxymethyl ester staining, mCherry fluorescence intensity, and the tetrazolium assay. Four recombinant AAV serotypes (1, 2, 6, 8) expressing green fluorescent protein (GFP) under the CAG promoter were added to the slice surface. Gene transfer efficiency was quantified as the number of GFP-positive cells per slice. AAV cell tropism was examined by comparing the number of GFP-positive cardiomyocytes (CMs) and fibroblasts within heart slices. Results Slices retained viability in in vitro culture for at least 5 days. After adding AAV particles, AAV6-infected slices showed the highest number of GFP-expressing cells, almost exclusively CMs. Slice incubation with AAV1, 2, and 8 resulted in fewer GFP-positive cells, with AAV2 having the lowest gene transfer efficiency. None of the AAV serotypes tested caused significant cytotoxicity when compared to non-infected control slices. Conclusions We have established a readily available mouse organotypic heart slice culture model and provided evidence that AAV6 may be a promising gene therapy vector for heart failure and other cardiac diseases.
Collapse
Affiliation(s)
- Zihou Liu
- Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Kristin Klose
- Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Neuber
- Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
| | - Meng Jiang
- Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Manfred Gossen
- Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany.,Helmholtz-Zentrum Geesthacht, Institute of Biomaterial Science, Teltow, Germany
| | - Christof Stamm
- Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany. .,Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany. .,German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany. .,Helmholtz-Zentrum Geesthacht, Institute of Biomaterial Science, Teltow, Germany.
| |
Collapse
|
35
|
Oliván-Viguera A, Pérez-Zabalza M, García-Mendívil L, Mountris KA, Orós-Rodrigo S, Ramos-Marquès E, Vallejo-Gil JM, Fresneda-Roldán PC, Fañanás-Mastral J, Vázquez-Sancho M, Matamala-Adell M, Sorribas-Berjón F, Bellido-Morales JA, Mancebón-Sierra FJ, Vaca-Núñez AS, Ballester-Cuenca C, Marigil MÁ, Pastor C, Ordovás L, Köhler R, Diez E, Pueyo E. Minimally invasive system to reliably characterize ventricular electrophysiology from living donors. Sci Rep 2020; 10:19941. [PMID: 33203905 PMCID: PMC7673124 DOI: 10.1038/s41598-020-77076-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 11/03/2020] [Indexed: 01/03/2023] Open
Abstract
Cardiac tissue slices preserve the heterogeneous structure and multicellularity of the myocardium and allow its functional characterization. However, access to human ventricular samples is scarce. We aim to demonstrate that slices from small transmural core biopsies collected from living donors during routine cardiac surgery preserve structural and functional properties of larger myocardial specimens, allowing accurate electrophysiological characterization. In pigs, we compared left ventricular transmural core biopsies with transmural tissue blocks from the same ventricular region. In humans, we analyzed transmural biopsies and papillary muscles from living donors. All tissues were vibratome-sliced. By histological analysis of the transmural biopsies, we showed that tissue architecture and cellular organization were preserved. Enzymatic and vital staining methods verified viability. Optically mapped transmembrane potentials confirmed that action potential duration and morphology were similar in pig biopsies and tissue blocks. Action potential morphology and duration in human biopsies and papillary muscles agreed with published ranges. In both pigs and humans, responses to increasing pacing frequencies and β-adrenergic stimulation were similar in transmural biopsies and larger tissues. We show that it is possible to successfully collect and characterize tissue slices from human myocardial biopsies routinely extracted from living donors, whose behavior mimics that of larger myocardial preparations both structurally and electrophysiologically.
Collapse
Affiliation(s)
- Aida Oliván-Viguera
- Biomedical Signal Interpretation and Computational Simulation (BSICoS) Group, Aragón, Institute of Engineering Research (I3A) and Instituto de Investigación Sanitaria (IIS) Aragón, University of Zaragoza, Edificio I+D+i, C/Mariano Esquillor s/n, 50018, Zaragoza, Spain.
| | - María Pérez-Zabalza
- Biomedical Signal Interpretation and Computational Simulation (BSICoS) Group, Aragón, Institute of Engineering Research (I3A) and Instituto de Investigación Sanitaria (IIS) Aragón, University of Zaragoza, Edificio I+D+i, C/Mariano Esquillor s/n, 50018, Zaragoza, Spain
| | - Laura García-Mendívil
- Biomedical Signal Interpretation and Computational Simulation (BSICoS) Group, Aragón, Institute of Engineering Research (I3A) and Instituto de Investigación Sanitaria (IIS) Aragón, University of Zaragoza, Edificio I+D+i, C/Mariano Esquillor s/n, 50018, Zaragoza, Spain
| | - Konstantinos A Mountris
- Biomedical Signal Interpretation and Computational Simulation (BSICoS) Group, Aragón, Institute of Engineering Research (I3A) and Instituto de Investigación Sanitaria (IIS) Aragón, University of Zaragoza, Edificio I+D+i, C/Mariano Esquillor s/n, 50018, Zaragoza, Spain
| | - Sofía Orós-Rodrigo
- Biomedical Signal Interpretation and Computational Simulation (BSICoS) Group, Aragón, Institute of Engineering Research (I3A) and Instituto de Investigación Sanitaria (IIS) Aragón, University of Zaragoza, Edificio I+D+i, C/Mariano Esquillor s/n, 50018, Zaragoza, Spain
| | - Estel Ramos-Marquès
- Biomedical Signal Interpretation and Computational Simulation (BSICoS) Group, Aragón, Institute of Engineering Research (I3A) and Instituto de Investigación Sanitaria (IIS) Aragón, University of Zaragoza, Edificio I+D+i, C/Mariano Esquillor s/n, 50018, Zaragoza, Spain
| | - José María Vallejo-Gil
- Department of Cardiovascular Surgery, University Hospital Miguel Servet, Zaragoza, Spain
| | | | - Javier Fañanás-Mastral
- Department of Cardiovascular Surgery, University Hospital Miguel Servet, Zaragoza, Spain
| | - Manuel Vázquez-Sancho
- Department of Cardiovascular Surgery, University Hospital Miguel Servet, Zaragoza, Spain
| | - Marta Matamala-Adell
- Department of Cardiovascular Surgery, University Hospital Miguel Servet, Zaragoza, Spain
| | | | | | | | | | | | | | | | - Laura Ordovás
- Biomedical Signal Interpretation and Computational Simulation (BSICoS) Group, Aragón, Institute of Engineering Research (I3A) and Instituto de Investigación Sanitaria (IIS) Aragón, University of Zaragoza, Edificio I+D+i, C/Mariano Esquillor s/n, 50018, Zaragoza, Spain.,Aragón Agency for Research and Development (ARAID), Zaragoza, Spain
| | - Ralf Köhler
- Aragón Institute of Health Sciences (IACS), Zaragoza, Spain.,Aragón Agency for Research and Development (ARAID), Zaragoza, Spain
| | - Emiliano Diez
- Institute of Experimental Medicine and Biology of Cuyo (IMBECU), CONICET, Mendoza, Argentina
| | - Esther Pueyo
- Biomedical Signal Interpretation and Computational Simulation (BSICoS) Group, Aragón, Institute of Engineering Research (I3A) and Instituto de Investigación Sanitaria (IIS) Aragón, University of Zaragoza, Edificio I+D+i, C/Mariano Esquillor s/n, 50018, Zaragoza, Spain.,Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, Spain
| |
Collapse
|
36
|
Watson SA, Dendorfer A, Thum T, Perbellini F. A practical guide for investigating cardiac physiology using living myocardial slices. Basic Res Cardiol 2020; 115:61. [PMID: 32914360 PMCID: PMC7496048 DOI: 10.1007/s00395-020-00822-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 08/10/2020] [Indexed: 12/22/2022]
Abstract
Ex vivo multicellular preparations are essential tools to study tissue physiology. Among them, the recent methodological and technological developments in living myocardial slices (LMS) are attracting increasing interest by the cardiac research field. Despite this, this research model remains poorly perceived and utilized by most research laboratories. Here, we provide a practical guide on how to use LMS to interrogate multiple aspects of cardiac function, structure and biochemistry. We discuss issues that should be considered to conduct successful experiments, including experimental design, sample preparation, data collection and analysis. We describe how laboratory setups can be adapted to accommodate and interrogate this multicellular research model. These adaptations can often be achieved at a reasonable cost with off-the-shelf components and operated reliably using well-established protocols and freely available software, which is essential to broaden the utilization of this method. We will also highlight how current measurements can be improved to further enhance data quality and reliability to ensure inter-laboratory reproducibility. Finally, we summarize the most promising biomedical applications and envision how living myocardial slices can lead to further breakthroughs.
Collapse
Affiliation(s)
| | - A Dendorfer
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - T Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hanover, Germany. .,National Heart and Lung Institute, Imperial College London, London, UK.
| | - F Perbellini
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hanover, Germany.
| |
Collapse
|
37
|
Pitoulis FG, Watson SA, Perbellini F, Terracciano CM. Myocardial slices come to age: an intermediate complexity in vitro cardiac model for translational research. Cardiovasc Res 2020; 116:1275-1287. [PMID: 31868875 PMCID: PMC7243278 DOI: 10.1093/cvr/cvz341] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/31/2019] [Accepted: 12/19/2019] [Indexed: 12/17/2022] Open
Abstract
Although past decades have witnessed significant reductions in mortality of heart failure together with advances in our understanding of its cellular, molecular, and whole-heart features, a lot of basic cardiac research still fails to translate into clinical practice. In this review we examine myocardial slices, a novel model in the translational arena. Myocardial slices are living ultra-thin sections of heart tissue. Slices maintain the myocardium's native function (contractility, electrophysiology) and structure (multicellularity, extracellular matrix) and can be prepared from animal and human tissue. The discussion begins with the history and current advances in the model, the different interlaboratory methods of preparation and their potential impact on results. We then contextualize slices' advantages and limitations by comparing it with other cardiac models. Recently, sophisticated methods have enabled slices to be cultured chronically in vitro while preserving the functional and structural phenotype. This is more timely now than ever where chronic physiologically relevant in vitro platforms for assessment of therapeutic strategies are urgently needed. We interrogate the technological developments that have permitted this, their limitations, and future directions. Finally, we look into the general obstacles faced by the translational field, and how implementation of research systems utilizing slices could help in resolving these.
Collapse
Affiliation(s)
- Fotios G Pitoulis
- Laboratory of Cell Electrophysiology, Department of Myocardial Function, Imperial College London, National Heart and Lung Institute, 4th Floor ICTEM Building Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - Samuel A Watson
- Laboratory of Cell Electrophysiology, Department of Myocardial Function, Imperial College London, National Heart and Lung Institute, 4th Floor ICTEM Building Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - Filippo Perbellini
- Laboratory of Cell Electrophysiology, Department of Myocardial Function, Imperial College London, National Heart and Lung Institute, 4th Floor ICTEM Building Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Hannover, Germany
| | - Cesare M Terracciano
- Laboratory of Cell Electrophysiology, Department of Myocardial Function, Imperial College London, National Heart and Lung Institute, 4th Floor ICTEM Building Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
38
|
Guo J, Huebsch N. Modeling the Response of Heart Muscle to Mechanical Stimulation In Vitro. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s43152-020-00007-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
39
|
Finetti F, Capitani N, Manganaro N, Tatangelo V, Libonati F, Panattoni G, Calaresu I, Ballerini L, Baldari CT, Patrussi L. Optimization of Organotypic Cultures of Mouse Spleen for Staining and Functional Assays. Front Immunol 2020; 11:471. [PMID: 32265925 PMCID: PMC7105700 DOI: 10.3389/fimmu.2020.00471] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/28/2020] [Indexed: 12/12/2022] Open
Abstract
By preserving cell viability and three-dimensional localization, organotypic culture stands out among the newest frontiers of cell culture. It has been successfully employed for the study of diseases among which neoplasias, where tumoral cells take advantage of the surrounding stroma to promote their own proliferation and survival. Organotypic culture acquires major importance in the context of the immune system, whose cells cross-talk in a complex and dynamic fashion to elicit productive responses. However, organotypic culture has been as yet poorly developed for and applied to primary and secondary lymphoid organs. Here we describe in detail the development of a protocol suitable for the efficient cutting of mouse spleen, which overcomes technical difficulties related to the peculiar organ texture, and for optimized organotypic culture of spleen slices. Moreover, we used microscopy, immunofluorescence, flow cytometry, and qRT-PCR to demonstrate that the majority of cells residing in spleen slices remain alive and maintain their original location in the organ architecture for several days after cutting. The development of this protocol represents a significant technical improvement in the study of the lymphoid microenvironment in both physiological and pathological conditions involving the immune system.
Collapse
Affiliation(s)
| | - Nagaja Capitani
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Noemi Manganaro
- Department of Life Sciences, University of Siena, Siena, Italy
| | | | | | - Giulia Panattoni
- International School for Advanced Studies (SISSA/ISAS), Trieste, Italy
| | - Ivo Calaresu
- International School for Advanced Studies (SISSA/ISAS), Trieste, Italy
| | - Laura Ballerini
- International School for Advanced Studies (SISSA/ISAS), Trieste, Italy
| | | | - Laura Patrussi
- Department of Life Sciences, University of Siena, Siena, Italy
| |
Collapse
|
40
|
Watson SA, Terracciano CM, Perbellini F. Myocardial Slices: an Intermediate Complexity Platform for Translational Cardiovascular Research. Cardiovasc Drugs Ther 2020; 33:239-244. [PMID: 30671746 PMCID: PMC6509068 DOI: 10.1007/s10557-019-06853-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Myocardial slices, also known as “cardiac tissue slices” or “organotypic heart slices,” are ultrathin (100–400 μm) slices of living adult ventricular myocardium prepared using a high-precision vibratome. They are a model of intermediate complexity as they retain the native multicellularity, architecture, and physiology of the heart, while their thinness ensures adequate oxygen and metabolic substrate diffusion in vitro. Myocardial slices can be produced from a variety of animal models and human biopsies, thus providing a representative human in vitro platform for translational cardiovascular research. In this review, we compare myocardial slices to other in vitro models and highlight some of the unique advantages provided by this platform. Additionally, we discuss the work performed in our laboratory to optimize myocardial slice preparation methodology, which resulted in highly viable myocardial slices from both large and small mammalian hearts with only 2–3% cardiomyocyte damage and preserved structure and function. Applications of myocardial slices span both basic and translational cardiovascular science. Our laboratory has utilized myocardial slices for the investigation of cardiac multicellularity, visualizing 3D collagen distribution and micro/macrovascular networks using tissue clearing protocols and investigating the effects of novel conductive biomaterials on cardiac physiology. Myocardial slices have been widely used for pharmacological testing. Finally, the current challenges and future directions for the technology are discussed.
Collapse
Affiliation(s)
- Samuel A Watson
- National Heart & Lung Institute, Imperial College London, London, UK
| | | | | |
Collapse
|
41
|
Molecular mechanisms of Wischnewski spot development on gastric mucosa in fatal hypothermia: an experimental study in rats. Sci Rep 2020; 10:1877. [PMID: 32024924 PMCID: PMC7002760 DOI: 10.1038/s41598-020-58894-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 01/17/2020] [Indexed: 02/02/2023] Open
Abstract
Numerous dark-brown-coloured small spots called “Wischnewski spots” are often observed in the gastric mucosa in the patients dying of hypothermia, but the molecular mechanisms through which they develop remain unclear. We hypothesised that hypothermia may activate the secretion of gastric acid and pepsin, leading to the development of the spots. To investigate this, we performed experiments using organotypic rat gastric tissue slices cultured at 37 °C (control) or 32 °C (cold). Cold loading for 6 h lowered the extracellular pH in the culture medium. The mRNA expression of gastrin, which regulates gastric acid secretion, increased after cold loading for 3 h. Cold loading increased the expression of gastric H+,K+-ATPase pump protein in the apical canalicular membrane and resulted in dynamic morphological changes in parietal cells. Cold loading resulted in an increased abundance of pepsin C protein and an elevated mRNA expression of its precursor progastricsin. Collectively, our findings clarified that cold stress induces acidification by activating gastric H+,K+-ATPase pumps and promoting pepsin C release through inducing progastricsin expression on the gastric mucosa, leading to tiny haemorrhages or erosions of the gastric mucosa that manifest as Wischnewski spots in fatal hypothermia.
Collapse
|
42
|
Karbassi E, Fenix A, Marchiano S, Muraoka N, Nakamura K, Yang X, Murry CE. Cardiomyocyte maturation: advances in knowledge and implications for regenerative medicine. Nat Rev Cardiol 2020; 17:341-359. [PMID: 32015528 DOI: 10.1038/s41569-019-0331-x] [Citation(s) in RCA: 447] [Impact Index Per Article: 89.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2019] [Indexed: 12/20/2022]
Abstract
Our knowledge of pluripotent stem cell (PSC) biology has advanced to the point where we now can generate most cells of the human body in the laboratory. PSC-derived cardiomyocytes can be generated routinely with high yield and purity for disease research and drug development, and these cells are now gradually entering the clinical research phase for the testing of heart regeneration therapies. However, a major hurdle for their applications is the immature state of these cardiomyocytes. In this Review, we describe the structural and functional properties of cardiomyocytes and present the current approaches to mature PSC-derived cardiomyocytes. To date, the greatest success in maturation of PSC-derived cardiomyocytes has been with transplantation into the heart in animal models and the engineering of 3D heart tissues with electromechanical conditioning. In conventional 2D cell culture, biophysical stimuli such as mechanical loading, electrical stimulation and nanotopology cues all induce substantial maturation, particularly of the contractile cytoskeleton. Metabolism has emerged as a potent means to control maturation with unexpected effects on electrical and mechanical function. Different interventions induce distinct facets of maturation, suggesting that activating multiple signalling networks might lead to increased maturation. Despite considerable progress, we are still far from being able to generate PSC-derived cardiomyocytes with adult-like phenotypes in vitro. Future progress will come from identifying the developmental drivers of maturation and leveraging them to create more mature cardiomyocytes for research and regenerative medicine.
Collapse
Affiliation(s)
- Elaheh Karbassi
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Department of Pathology, University of Washington, Seattle, WA, USA
| | - Aidan Fenix
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Department of Pathology, University of Washington, Seattle, WA, USA
| | - Silvia Marchiano
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Department of Pathology, University of Washington, Seattle, WA, USA
| | - Naoto Muraoka
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Department of Pathology, University of Washington, Seattle, WA, USA
| | - Kenta Nakamura
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Xiulan Yang
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Department of Pathology, University of Washington, Seattle, WA, USA
| | - Charles E Murry
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA. .,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA. .,Department of Pathology, University of Washington, Seattle, WA, USA. .,Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA. .,Department of Bioengineering, University of Washington, Seattle, WA, USA.
| |
Collapse
|
43
|
Ou Q, Jacobson Z, Abouleisa RRE, Tang XL, Hindi SM, Kumar A, Ivey KN, Giridharan G, El-Baz A, Brittian K, Rood B, Lin YH, Watson SA, Perbellini F, McKinsey TA, Hill BG, Jones SP, Terracciano CM, Bolli R, Mohamed TMA. Physiological Biomimetic Culture System for Pig and Human Heart Slices. Circ Res 2019; 125:628-642. [PMID: 31310161 DOI: 10.1161/circresaha.119.314996] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
RATIONALE Preclinical testing of cardiotoxicity and efficacy of novel heart failure therapies faces a major limitation: the lack of an in situ culture system that emulates the complexity of human heart tissue and maintains viability and functionality for a prolonged time. OBJECTIVE To develop a reliable, easily reproducible, medium-throughput method to culture pig and human heart slices under physiological conditions for a prolonged period of time. METHODS AND RESULTS Here, we describe a novel, medium-throughput biomimetic culture system that maintains viability and functionality of human and pig heart slices (300 µm thickness) for 6 days in culture. We optimized the medium and culture conditions with continuous electrical stimulation at 1.2 Hz and oxygenation of the medium. Functional viability of these slices over 6 days was confirmed by assessing their calcium homeostasis, twitch force generation, and response to β-adrenergic stimulation. Temporal transcriptome analysis using RNAseq at day 2, 6, and 10 in culture confirmed overall maintenance of normal gene expression for up to 6 days, while over 500 transcripts were differentially regulated after 10 days. Electron microscopy demonstrated intact mitochondria and Z-disc ultra-structures after 6 days in culture under our optimized conditions. This biomimetic culture system was successful in keeping human heart slices completely viable and functionally and structurally intact for 6 days in culture. We also used this system to demonstrate the effects of a novel gene therapy approach in human heart slices. Furthermore, this culture system enabled the assessment of contraction and relaxation kinetics on isolated single myofibrils from heart slices after culture. CONCLUSIONS We have developed and optimized a reliable medium-throughput culture system for pig and human heart slices as a platform for testing the efficacy of novel heart failure therapeutics and reliable testing of cardiotoxicity in a 3-dimensional heart model.
Collapse
Affiliation(s)
- Qinghui Ou
- From the Department of Medicine, Institute of Molecular Cardiology (Q.O., R.R.E.A., X.-L.T., K.B., S.P.J., R.B., T.M.A.M.), University of Louisville, KY
| | - Zoë Jacobson
- Tenaya Therapeutics, South San Francisco, CA (Z.J., K.N.I.)
| | - Riham R E Abouleisa
- From the Department of Medicine, Institute of Molecular Cardiology (Q.O., R.R.E.A., X.-L.T., K.B., S.P.J., R.B., T.M.A.M.), University of Louisville, KY
| | - Xian-Liang Tang
- From the Department of Medicine, Institute of Molecular Cardiology (Q.O., R.R.E.A., X.-L.T., K.B., S.P.J., R.B., T.M.A.M.), University of Louisville, KY
| | - Sajedah M Hindi
- Departments of Anatomical Sciences and Neurobiology (S.M.H., A.K.), University of Louisville, KY
| | - Ashok Kumar
- Departments of Anatomical Sciences and Neurobiology (S.M.H., A.K.), University of Louisville, KY
| | - Kathryn N Ivey
- Tenaya Therapeutics, South San Francisco, CA (Z.J., K.N.I.)
| | | | - Ayman El-Baz
- Department of Bioengineering (G.G., A.E.-B.), University of Louisville, KY
| | - Kenneth Brittian
- From the Department of Medicine, Institute of Molecular Cardiology (Q.O., R.R.E.A., X.-L.T., K.B., S.P.J., R.B., T.M.A.M.), University of Louisville, KY
| | - Benjamin Rood
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine (B.R., B.G.H., S.P.J., T.M.A.M.), University of Louisville, KY
| | - Ying-Hsi Lin
- Division of Cardiology and Consortium for Fibrosis Research & Translation, Department of Medicine, University of Colorado, Aurora (Y.-H.L., T.A.M.)
| | - Samuel A Watson
- National Heart & Lung Institute, Imperial College London, United Kingdom (S.A.W., F.P., C.M.T.)
| | - Filippo Perbellini
- National Heart & Lung Institute, Imperial College London, United Kingdom (S.A.W., F.P., C.M.T.).,Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Germany (F.P.)
| | - Timothy A McKinsey
- Division of Cardiology and Consortium for Fibrosis Research & Translation, Department of Medicine, University of Colorado, Aurora (Y.-H.L., T.A.M.)
| | - Bradford G Hill
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine (B.R., B.G.H., S.P.J., T.M.A.M.), University of Louisville, KY
| | - Steven P Jones
- From the Department of Medicine, Institute of Molecular Cardiology (Q.O., R.R.E.A., X.-L.T., K.B., S.P.J., R.B., T.M.A.M.), University of Louisville, KY.,Envirome Institute, Diabetes and Obesity Center, Department of Medicine (B.R., B.G.H., S.P.J., T.M.A.M.), University of Louisville, KY
| | - Cesare M Terracciano
- National Heart & Lung Institute, Imperial College London, United Kingdom (S.A.W., F.P., C.M.T.)
| | - Roberto Bolli
- From the Department of Medicine, Institute of Molecular Cardiology (Q.O., R.R.E.A., X.-L.T., K.B., S.P.J., R.B., T.M.A.M.), University of Louisville, KY
| | - Tamer M A Mohamed
- From the Department of Medicine, Institute of Molecular Cardiology (Q.O., R.R.E.A., X.-L.T., K.B., S.P.J., R.B., T.M.A.M.), University of Louisville, KY.,Envirome Institute, Diabetes and Obesity Center, Department of Medicine (B.R., B.G.H., S.P.J., T.M.A.M.), University of Louisville, KY.,Department of Pharmacology and Toxicology (T.M.A.M.), University of Louisville, KY.,Institute of Cardiovascular Sciences, University of Manchester, United Kingdom (T.M.A.M.).,Faculty of Pharmacy, Zagazig University, Egypt (T.M.A.M.)
| |
Collapse
|
44
|
Multiparametric slice culture platform for the investigation of human cardiac tissue physiology. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2019; 144:139-150. [DOI: 10.1016/j.pbiomolbio.2018.06.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/14/2018] [Accepted: 06/03/2018] [Indexed: 12/23/2022]
|
45
|
Schroer A, Pardon G, Castillo E, Blair C, Pruitt B. Engineering hiPSC cardiomyocyte in vitro model systems for functional and structural assessment. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2019; 144:3-15. [PMID: 30579630 PMCID: PMC6919215 DOI: 10.1016/j.pbiomolbio.2018.12.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 09/24/2018] [Accepted: 12/04/2018] [Indexed: 02/06/2023]
Abstract
The study of human cardiomyopathies and the development and testing of new therapies has long been limited by the availability of appropriate in vitro model systems. Cardiomyocytes are highly specialized cells whose internal structure and contractile function are sensitive to the local microenvironment and the combination of mechanical and biochemical cues they receive. The complementary technologies of human induced pluripotent stem cell (hiPSC) derived cardiomyocytes (CMs) and microphysiological systems (MPS) allow for precise control of the genetics and microenvironment of human cells in in vitro contexts. These combined systems also enable quantitative measurement of mechanical function and intracellular organization. This review describes relevant factors in the myocardium microenvironment that affect CM structure and mechanical function and demonstrates the application of several engineered microphysiological systems for studying development, disease, and drug discovery.
Collapse
Affiliation(s)
- Alison Schroer
- Departments of Mechanical Engineering and Bioengineering, Stanford University, Stanford, CA, 94305, USA.
| | - Gaspard Pardon
- Departments of Mechanical Engineering and Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Erica Castillo
- Departments of Mechanical Engineering and Bioengineering, Stanford University, Stanford, CA, 94305, USA; Department of Mechanical Engineering, University of California at Santa Barbara, USA
| | - Cheavar Blair
- Departments of Mechanical Engineering and Bioengineering, Stanford University, Stanford, CA, 94305, USA; Department of Mechanical Engineering, University of California at Santa Barbara, USA
| | - Beth Pruitt
- Departments of Mechanical Engineering and Bioengineering, Stanford University, Stanford, CA, 94305, USA; Department of Mechanical Engineering, University of California at Santa Barbara, USA
| |
Collapse
|
46
|
Perbellini F, Watson SA, Scigliano M, Alayoubi S, Tkach S, Bardi I, Quaife N, Kane C, Dufton NP, Simon A, Sikkel MB, Faggian G, Randi AM, Gorelik J, Harding SE, Terracciano CM. Investigation of cardiac fibroblasts using myocardial slices. Cardiovasc Res 2019; 114:77-89. [PMID: 29016704 PMCID: PMC5852538 DOI: 10.1093/cvr/cvx152] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 08/17/2017] [Indexed: 12/15/2022] Open
Abstract
Aims Cardiac fibroblasts (CFs) are considered the principal regulators of cardiac fibrosis. Factors that influence CF activity are difficult to determine. When isolated and cultured in vitro, CFs undergo rapid phenotypic changes including increased expression of α-SMA. Here we describe a new model to study CFs and their response to pharmacological and mechanical stimuli using in vitro cultured mouse, dog and human myocardial slices. Methods and results Unloading of myocardial slices induced CF proliferation without α-SMA expression up to 7 days in culture. CFs migrating onto the culture plastic support or cultured on glass expressed αSMA within 3 days. The cells on the slice remained αSMA(−) despite transforming growth factor-β (20 ng/ml) or angiotensin II (200 µM) stimulation. When diastolic load was applied to myocardial slices using A-shaped stretchers, CF proliferation was significantly prevented at Days 3 and 7 (P < 0.001). Conclusions Myocardial slices allow the study of CFs in a multicellular environment and may be used to effectively study mechanisms of cardiac fibrosis and potential targets.
Collapse
Affiliation(s)
- Filippo Perbellini
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | - Samuel A Watson
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | | | - Samha Alayoubi
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | - Sebastian Tkach
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | - Ifigeneia Bardi
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | - Nicholas Quaife
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | - Christopher Kane
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | - Neil P Dufton
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | - André Simon
- Department of Cardiothoracic Transplantation and Mechanical Circulatory Support, Royal Brompton and Harefield NHS Foundation Trust, Harefield, UK
| | - Markus B Sikkel
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | - Giuseppe Faggian
- Department of Cardiac Surgery, University of Verona, Verona, Italy
| | - Anna M Randi
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | - Julia Gorelik
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | - Sian E Harding
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | - Cesare M Terracciano
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| |
Collapse
|
47
|
Watson SA, Duff J, Bardi I, Zabielska M, Atanur SS, Jabbour RJ, Simon A, Tomas A, Smolenski RT, Harding SE, Perbellini F, Terracciano CM. Biomimetic electromechanical stimulation to maintain adult myocardial slices in vitro. Nat Commun 2019; 10:2168. [PMID: 31092830 PMCID: PMC6520377 DOI: 10.1038/s41467-019-10175-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 04/24/2019] [Indexed: 01/01/2023] Open
Abstract
Adult cardiac tissue undergoes a rapid process of dedifferentiation when cultured outside the body. The in vivo environment, particularly constant electromechanical stimulation, is fundamental to the regulation of cardiac structure and function. We investigated the role of electromechanical stimulation in preventing culture-induced dedifferentiation of adult cardiac tissue using rat, rabbit and human heart failure myocardial slices. Here we report that the application of a preload equivalent to sarcomere length (SL) = 2.2 μm is optimal for the maintenance of rat myocardial slice structural, functional and transcriptional properties at 24 h. Gene sets associated with the preservation of structure and function are activated, while gene sets involved in dedifferentiation are suppressed. The maximum contractility of human heart failure myocardial slices at 24 h is also optimally maintained at SL = 2.2 μm. Rabbit myocardial slices cultured at SL = 2.2 μm remain stable for 5 days. This approach substantially prolongs the culture of adult cardiac tissue in vitro.
Collapse
Affiliation(s)
- Samuel A Watson
- National Heart & Lung Institute, Imperial College London, London, W12 0NN, UK
| | - James Duff
- National Heart & Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Ifigeneia Bardi
- National Heart & Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Magdalena Zabielska
- Department of Biochemistry, Medical University of Gdańsk, Gdańsk, 80-210, Poland
| | - Santosh S Atanur
- Faculty of Medicine, Department of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - Richard J Jabbour
- National Heart & Lung Institute, Imperial College London, London, W12 0NN, UK
| | - André Simon
- Department of Cardiothoracic Transplantation & Mechanical Circulatory Support, Royal Brompton & Harefield NHS Foundation Trust, Harefield, UB9 6JH, UK
| | - Alejandra Tomas
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London, W12 0NN, UK
| | - Ryszard T Smolenski
- Department of Biochemistry, Medical University of Gdańsk, Gdańsk, 80-210, Poland
| | - Sian E Harding
- National Heart & Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Filippo Perbellini
- National Heart & Lung Institute, Imperial College London, London, W12 0NN, UK.
| | | |
Collapse
|
48
|
Sokolova V, Rojas-Sánchez L, Białas N, Schulze N, Epple M. Calcium phosphate nanoparticle-mediated transfection in 2D and 3D mono- and co-culture cell models. Acta Biomater 2019; 84:391-401. [PMID: 30503560 DOI: 10.1016/j.actbio.2018.11.051] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 11/25/2018] [Accepted: 11/28/2018] [Indexed: 12/28/2022]
Abstract
The transfer of nucleic acids into living cells, i.e. transfection, is a major technique in current molecular biology and medicine. As nucleic acids alone are not able to penetrate the cell membrane, an efficient carrier is needed. Calcium phosphate nanoparticles can serve as carrier due to their biocompatibility, biodegradability and high affinity to nucleic acids like DNA or RNA. Their application was extended here from two-dimensional (2D) to three-dimensional (3D) cell culture models, including co-cultures. Compared to 2D monolayer cell cultures, a 3D culture system represents a more realistic spatial, biochemical and cellular environment. The uptake of fluorescent calcium phosphate nanoparticles (diameter 40-70 nm; cationic) was studied in 2D and 3D cell culture models by confocal laser scanning microscopy. The transfection of eGFP by calcium phosphate nanoparticles was compared in 2D and 3D cell culture, including co-cultures of green fluorescing HeLa-eGFP cells and MG-63 cells in 2D and in 3D models with the red fluorescent protein mCherry. This permitted a cell-specific assessment of the local transfection efficiency. In general, the penetration of nanoparticles into the spheroids was significantly higher than that of a model oligonucleotide carried by Lipofectamine. The transfection efficiency was comparable in 3D cell cultures with 2D cell cultures, but it occurred preferentially at the surface of the spheroids, following the uptake pathway of the nanoparticles. STATEMENT OF SIGNIFICANCE: Three-dimensional cell culture models can serve as a bridge between the in-vitro cell cultures and the in-vivo situation, especially when mass transfer effects have to be considered. This is the case for nanoparticles where the incubation effect in a two-dimensional cell culture strongly differs from a three-dimensional cell culture or a living tissue. We have compared the uptake of nanoparticles and a subsequent transfection of fluorescent proteins in two-dimensional and three-dimensional cell culture models. An elegant model to investigate the transfection in co-cultures was developed using HeLa-eGFP cells (green fluorescent) together with MG-63 cells (non-fluorescent) that were transfected with the red-fluorescing protein mCherry. Thereby, the transfection of both cell types in the co-culture was easily distinguished.
Collapse
|
49
|
Long-term functional and structural preservation of precision-cut human myocardium under continuous electromechanical stimulation in vitro. Nat Commun 2019; 10:117. [PMID: 30631059 PMCID: PMC6328583 DOI: 10.1038/s41467-018-08003-1] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 12/10/2018] [Indexed: 12/26/2022] Open
Abstract
In vitro models incorporating the complexity and function of adult human tissues are highly desired for translational research. Whilst vital slices of human myocardium approach these demands, their rapid degeneration in tissue culture precludes long-term experimentation. Here, we report preservation of structure and performance of human myocardium under conditions of physiological preload, compliance, and continuous excitation. In biomimetic culture, tissue slices prepared from explanted failing human hearts attain a stable state of contractility that can be monitored for up to 4 months or 2000000 beats in vitro. Cultured myocardium undergoes particular alterations in biomechanics, structure, and mRNA expression. The suitability of the model for drug safety evaluation is exemplified by repeated assessment of refractory period that permits sensitive analysis of repolarization impairment induced by the multimodal hERG-inhibitor pentamidine. Biomimetic tissue culture will provide new opportunities to study drug targets, gene functions, and cellular plasticity in adult human myocardium. Myocardial tissue undergoes steady functional decline when cultured in vitro. Here, the authors report a protocol for culture of human cardiac slices that allows maintenance of contractility for up to four months, and show that the model is suitable for evaluation of drug safety, as exemplified for drugs interfering with cardiomyocyte repolarization.
Collapse
|
50
|
Bajpai G, Schneider C, Wong N, Bredemeyer A, Hulsmans M, Nahrendorf M, Epelman S, Kreisel D, Liu Y, Itoh A, Shankar TS, Selzman CH, Drakos SG, Lavine KJ. The human heart contains distinct macrophage subsets with divergent origins and functions. Nat Med 2018; 24:1234-1245. [PMID: 29892064 PMCID: PMC6082687 DOI: 10.1038/s41591-018-0059-x] [Citation(s) in RCA: 477] [Impact Index Per Article: 68.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 04/13/2018] [Indexed: 12/21/2022]
Abstract
Paradigm-shifting studies in the mouse have identified tissue macrophage heterogeneity as a critical determinant of immune responses. In contrast, surprisingly little is known regarding macrophage heterogeneity in humans. Macrophages within the mouse heart are partitioned into CCR2- and CCR2+ subsets with divergent origins, repopulation mechanisms, and functions. Here, we demonstrate that the human myocardium also contains distinct subsets of CCR2- and CCR2+ macrophages. Analysis of sex-mismatched heart transplant recipients revealed that CCR2- macrophages are a tissue-resident population exclusively replenished through local proliferation, whereas CCR2+ macrophages are maintained through monocyte recruitment and proliferation. Moreover, CCR2- and CCR2+ macrophages have distinct functional properties, analogous to reparative CCR2- and inflammatory CCR2+ macrophages in the mouse heart. Clinically, CCR2+ macrophage abundance is associated with left ventricular remodeling and systolic function in heart failure patients. Collectively, these observations provide initial evidence for the functional importance of macrophage heterogeneity in the human heart.
Collapse
Affiliation(s)
- Geetika Bajpai
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Caralin Schneider
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Nicole Wong
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Andrea Bredemeyer
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Maarten Hulsmans
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Slava Epelman
- Peter Munk Cardiac Center, Ted Rogers Center for Heart Failure Research, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Daniel Kreisel
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Yongjian Liu
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Akinobu Itoh
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Thirupura S Shankar
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Craig H Selzman
- Division of Cardiothoracic Surgery & Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Stavros G Drakos
- Division of Cardiovascular Medicine & Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Kory J Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Immunology and Pathology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|