1
|
Salmenov R, Mummery C, ter Huurne M. Cell cycle visualization tools to study cardiomyocyte proliferation in real-time. Open Biol 2024; 14:240167. [PMID: 39378987 PMCID: PMC11461051 DOI: 10.1098/rsob.240167] [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: 06/19/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 10/10/2024] Open
Abstract
Cardiomyocytes in the adult human heart are quiescent and those lost following heart injury are not replaced by proliferating survivors. Considerable effort has been made to understand the mechanisms underlying cardiomyocyte cell cycle exit and re-entry, with view to discovering therapeutics that could stimulate cardiomyocyte proliferation and heart regeneration. The advent of large compound libraries and robotic liquid handling platforms has enabled the screening of thousands of conditions in a single experiment but success of these screens depends on the appropriateness and quality of the model used. Quantification of (human) cardiomyocyte proliferation in high throughput has remained problematic because conventional antibody-based staining is costly, technically challenging and does not discriminate between cardiomyocyte division and failure in karyokinesis or cytokinesis. Live cell imaging has provided alternatives that facilitate high-throughput screening but these have other limitations. Here, we (i) review the cell cycle features of cardiomyocytes, (ii) discuss various cell cycle fluorescent reporter systems, and (iii) speculate on what could improve their predictive value in the context of cardiomyocyte proliferation. Finally, we consider how these new methods can be used in combination with state-of-the-art three-dimensional human cardiac organoid platforms to identify pro-proliferative signalling pathways that could stimulate regeneration of the human heart.
Collapse
Affiliation(s)
- Rustem Salmenov
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden2300RC, The Netherlands
| | - Christine Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden2300RC, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden2300RC, The Netherlands
| | - Menno ter Huurne
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden2300RC, The Netherlands
| |
Collapse
|
2
|
Martini S, Drzeniek NM, Stark R, Kollert MR, Du W, Reinke S, Ort M, Hardt S, Kotko I, Kath J, Schlickeiser S, Geißler S, Wagner DL, Krebs AC, Volk HD. Long-term in vitromaintenance of plasma cells in a hydrogel-enclosed human bone marrow microphysiological 3D model system. Biofabrication 2024; 16:045005. [PMID: 38955197 DOI: 10.1088/1758-5090/ad5dfe] [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: 10/16/2023] [Accepted: 07/01/2024] [Indexed: 07/04/2024]
Abstract
Plasma cells (PCs) in bone marrow (BM) play an important role in both protective and pathogenic humoral immune responses, e.g. in various malignant and non-malignant diseases such as multiple myeloma, primary and secondary immunodeficiencies and autoimmune diseases. Dedicated microenvironmental niches in the BM provide PCs with biomechanical and soluble factors that support their long-term survival. There is a high need for appropriate and robust model systems to better understand PCs biology, to develop new therapeutic strategies for PCs-related diseases and perform targeted preclinical studies with high predictive value. Most preclinical data have been derived fromin vivostudies in mice, asin vitrostudies of human PCs are limited due to restricted survival and functionality in conventional 2D cultures that do not reflect the unique niche architecture of the BM. We have developed a microphysiological, dynamic 3D BM culture system (BM-MPS) based on human primary tissue (femoral biopsies), mechanically supported by a hydrogel scaffold casing. While a bioinert agarose casing did not support PCs survival, a photo-crosslinked collagen-hyaluronic acid (Col-HA) hydrogel preserved the native BM niche architecture and allowed PCs survivalin vitrofor up to 2 weeks. Further, the Col-HA hydrogel was permissive to lymphocyte migration into the microphysiological system´s circulation. Long-term PCs survival was related to the stable presence in the culture of soluble factors, as APRIL, BAFF, and IL-6. Increasing immunoglobulins concentrations in the medium confirm their functionality over culture time. To the best of our knowledge, this study is the first report of successful long-term maintenance of primary-derived non-malignant PCsin vitro. Our innovative model system is suitable for in-depthin vitrostudies of human PCs regulation and exploration of targeted therapeutic approaches such as CAR-T cell therapy or biologics.
Collapse
Affiliation(s)
- Stefania Martini
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Norman Michael Drzeniek
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Regina Stark
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Matthias Reiner Kollert
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Julius Wolff Institute, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Weijie Du
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Simon Reinke
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Melanie Ort
- Julius Wolff Institute, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Sebastian Hardt
- Center for Musculoskeletal Surgery Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Iuliia Kotko
- Institute of Medical Immunology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Jonas Kath
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Stephan Schlickeiser
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- CheckImmune GmbH, Berlin, Germany
| | - Sven Geißler
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Julius Wolff Institute, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Dimitrios Laurin Wagner
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Institute of Transfusion Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Anna-Catharina Krebs
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Hans-Dieter Volk
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
3
|
Nappi F. Non-Coding RNA-Targeted Therapy: A State-of-the-Art Review. Int J Mol Sci 2024; 25:3630. [PMID: 38612441 PMCID: PMC11011542 DOI: 10.3390/ijms25073630] [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: 01/23/2024] [Revised: 03/11/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
The use of non-coding RNAs (ncRNAs) as drug targets is being researched due to their discovery and their role in disease. Targeting ncRNAs, including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), is an attractive approach for treating various diseases, such as cardiovascular disease and cancer. This seminar discusses the current status of ncRNAs as therapeutic targets in different pathological conditions. Regarding miRNA-based drugs, this approach has made significant progress in preclinical and clinical testing for cardiovascular diseases, where the limitations of conventional pharmacotherapy are evident. The challenges of miRNA-based drugs, including specificity, delivery, and tolerability, will be discussed. New approaches to improve their success will be explored. Furthermore, it extensively discusses the potential development of targeted therapies for cardiovascular disease. Finally, this document reports on the recent advances in identifying and characterizing microRNAs, manipulating them, and translating them into clinical applications. It also addresses the challenges and perspectives towards clinical application.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France
| |
Collapse
|
4
|
Sun Z, Lu K, Kamla C, Kameritsch P, Seidel T, Dendorfer A. Synchronous force and Ca 2+ measurements for repeated characterization of excitation-contraction coupling in human myocardium. Commun Biol 2024; 7:220. [PMID: 38388802 PMCID: PMC10884022 DOI: 10.1038/s42003-024-05886-3] [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: 09/04/2023] [Accepted: 02/02/2024] [Indexed: 02/24/2024] Open
Abstract
Dysfunctional Ca2+ signaling affects the myocardial systole and diastole, may trigger arrhythmia and cause transcriptomic and proteomic modifications in heart failure. Thus, synchronous real-time measurement of Ca2+ and force is essential to investigate the relationship between contractility and Ca2+ signaling and the alteration of excitation-contraction coupling (ECC) in human failing myocardium. Here, we present a method for synchronized acquisition of intracellular Ca2+ and contraction force in long-term cultivated slices of human failing myocardium. Synchronous time series of contraction force and intracellular Ca2+ were used to calculate force-calcium loops and to analyze the dynamic alterations of ECC in response to various pacing frequencies, post-pause potentiation, high mechanical preload and pharmacological interventions in human failing myocardium. We provide an approach to simultaneously and repeatedly investigate alterations of contractility and Ca2+ signals in long-term cultured myocardium, which will allow detecting the effects of electrophysiological or pharmacological interventions on human myocardial ECC.
Collapse
Affiliation(s)
- Zhengwu Sun
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Kun Lu
- Department of Cardiac Surgery, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner site Munich Heart Alliance, Munich, Germany
| | - Christine Kamla
- Department of Cardiac Surgery, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Petra Kameritsch
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Thomas Seidel
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Dendorfer
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany.
- DZHK (German Center for Cardiovascular Research), Partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
5
|
Cohrs CM, Chen C, Atkinson MA, Drotar DM, Speier S. Bridging the Gap: Pancreas Tissue Slices From Organ and Tissue Donors for the Study of Diabetes Pathogenesis. Diabetes 2024; 73:11-22. [PMID: 38117999 PMCID: PMC10784654 DOI: 10.2337/dbi20-0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 10/14/2023] [Indexed: 12/22/2023]
Abstract
Over the last two decades, increased availability of human pancreatic tissues has allowed for major expansions in our understanding of islet biology in health and disease. Indeed, studies of fixed and frozen pancreatic tissues, as well as efforts using viable isolated islets obtained from organ donors, have provided significant insights toward our understanding of diabetes. However, the procedures associated with islet isolation result in distressed cells that have been removed from any surrounding influence. The pancreas tissue slice technology was developed as an in situ approach to overcome certain limitations associated with studies on isolated islets or fixed tissue. In this Perspective, we discuss the value of this novel platform and review how pancreas tissue slices, within a short time, have been integrated in numerous studies of rodent and human islet research. We show that pancreas tissue slices allow for investigations in a less perturbed organ tissue environment, ranging from cellular processes, over peri-islet modulations, to tissue interactions. Finally, we discuss the considerations and limitations of this technology in its future applications. We believe the pancreas tissue slices will help bridge the gap between studies on isolated islets and cells to the systemic conditions by providing new insight into physiological and pathophysiological processes at the organ level. ARTICLE HIGHLIGHTS Human pancreas tissue slices represent a novel platform to study human islet biology in close to physiological conditions. Complementary to established technologies, such as isolated islets, single cells, and histological sections, pancreas tissue slices help bridge our understanding of islet physiology and pathophysiology from single cell to intact organ. Diverse sources of viable human pancreas tissue, each with distinct characteristics to be considered, are available to use in tissue slices for the study of diabetes pathogenesis.
Collapse
Affiliation(s)
- Christian M. Cohrs
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Munich at the University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Chunguang Chen
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Munich at the University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Mark A. Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL
| | - Denise M. Drotar
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL
| | - Stephan Speier
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Munich at the University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
6
|
Miller JM, Meki MM, El-Baz AS, Giridharan GA, Mohamed TMA. Culturing Cardiac Tissue Slices Under Continuous Physiological Mechanical Stretches. Methods Mol Biol 2024; 2803:61-74. [PMID: 38676885 DOI: 10.1007/978-1-0716-3846-0_5] [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] [Indexed: 04/29/2024]
Abstract
Testing drugs in vivo and in vitro have been essential elements for the discovery of new therapeutics. Due to the recent advances in in vitro cell culture models, such as human-induced pluripotent stem cell-derived cardiomyocytes and 3D multicell type organoid culture methods, the detection of adverse cardiac events prior to human clinical trials has improved. However, there are still numerous therapeutics whose adverse cardiac effects are not detected until human trials due to the inability of these cell cultures to fully model the complex multicellular organization of an intact human myocardium. Cardiac tissue slices are a possible alternative solution. Myocardial slices are a 300-micron thin snapshot of the myocardium, capturing a section of the adult heart in a 1 × 1 cm section. Using a culture method that incorporates essential nutrients and electrical stimulation, tissue slices can be maintained in culture for 6 days with full viability and functionality. With the addition of mechanical stimulation and humoral cues, tissue slices can be cultured for 12 days. Here we provide detailed methods for how to culture cardiac tissue slices under continuous mechanical stimulation in the cardiac tissue culture model (CTCM) device. The CTCM incorporates four essential factors for maintaining tissue slices in culture for 12 days: mechanical stimulation, electrical stimulation, nutrients, and humoral cues. The CTCM can also be used to model disease conditions, such as overstretch-induced cardiac hypertrophy. The versatility of the CTCM illustrates its potential to be a medium-throughput screening platform for personalized drug testing.
Collapse
Affiliation(s)
- Jessica M Miller
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY, USA
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
| | - Mostafa M Meki
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY, USA
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
| | - Ayman S El-Baz
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
| | | | - Tamer M A Mohamed
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY, USA.
- Department of Bioengineering, University of Louisville, Louisville, KY, USA.
- Surgery Department, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
7
|
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
|
8
|
Nunez-Toldra R, Del Canizo A, Secco I, Nicastro L, Giacca M, Terracciano CM. Living myocardial slices for the study of nucleic acid-based therapies. Front Bioeng Biotechnol 2023; 11:1275945. [PMID: 37941724 PMCID: PMC10628718 DOI: 10.3389/fbioe.2023.1275945] [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: 08/10/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023] Open
Abstract
Gene therapy based on viral vectors offers great potential for the study and the treatment of cardiac diseases. Here we explore the use of Living Myocardial Slices (LMS) as a platform for nucleic acid-based therapies. Rat LMS and Adeno-Associated viruses (AAV) were used to optimise and analyse gene transfer efficiency, viability, tissue functionality, and cell tropism in cardiac tissue. Human cardiac tissue from failing (dilated cardiomyopathy) hearts was also used to validate the model in a more translational setting. LMS were cultured at physiological sarcomere length for 72-h under electrical stimulation. Two recombinant AAV serotypes (AAV6 and AAV9) at different multiplicity of infection (MOI) expressing enhanced green fluorescent protein (eGFP) were added to the surface of rat LMS. AAV6 at 20,000 MOI proved to be the most suitable serotype without affecting LMS contractility or kinetics and showing high transduction and penetrability efficiency in rat LMS. This serotype exhibited 40% of transduction efficiency in cardiomyocytes and stromal cells while 20% of the endothelial cells were transduced. With great translational relevance, this protocol introduces the use of LMS as a model for nucleic acid-based therapies, allowing the acceleration of preclinical studies for cardiac diseases.
Collapse
Affiliation(s)
- R. Nunez-Toldra
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - A. Del Canizo
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - I. Secco
- King’s College London, School of Cardiovascular Medicine and Sciences, London, United Kingdom
| | - L. Nicastro
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - M. Giacca
- King’s College London, School of Cardiovascular Medicine and Sciences, London, United Kingdom
| | - C. M. Terracciano
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
9
|
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
|
10
|
Cofiño-Fabres C, Passier R, Schwach V. Towards Improved Human In Vitro Models for Cardiac Arrhythmia: Disease Mechanisms, Treatment, and Models of Atrial Fibrillation. Biomedicines 2023; 11:2355. [PMID: 37760796 PMCID: PMC10525681 DOI: 10.3390/biomedicines11092355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/18/2023] [Accepted: 08/19/2023] [Indexed: 09/29/2023] Open
Abstract
Heart rhythm disorders, arrhythmias, place a huge economic burden on society and have a large impact on the quality of life of a vast number of people. Arrhythmias can have genetic causes but primarily arise from heart tissue remodeling during aging or heart disease. As current therapies do not address the causes of arrhythmias but only manage the symptoms, it is of paramount importance to generate innovative test models and platforms for gaining knowledge about the underlying disease mechanisms which are compatible with drug screening. In this review, we outline the most important features of atrial fibrillation (AFib), the most common cardiac arrhythmia. We will discuss the epidemiology, risk factors, underlying causes, and present therapies of AFib, as well as the shortcomings and opportunities of current models for cardiac arrhythmia, including animal models, in silico and in vitro models utilizing human pluripotent stem cell (hPSC)-derived cardiomyocytes.
Collapse
Affiliation(s)
- Carla Cofiño-Fabres
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Drienerlolaan 5, 7500 AE Enschede, The Netherlands;
| | - Robert Passier
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Drienerlolaan 5, 7500 AE Enschede, The Netherlands;
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2300 RC Leiden, The Netherlands
| | - Verena Schwach
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Drienerlolaan 5, 7500 AE Enschede, The Netherlands;
| |
Collapse
|
11
|
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
|
Zhang X, Ni H, Morotti S, Smith C, Sato D, Louch W, Edwards A, Grandi E. Mechanisms of spontaneous Ca 2+ release-mediated arrhythmia in a novel 3D human atrial myocyte model: I. Transverse-axial tubule variation. J Physiol 2023; 601:2655-2683. [PMID: 36094888 PMCID: PMC10008525 DOI: 10.1113/jp283363] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/02/2022] [Indexed: 11/08/2022] Open
Abstract
Intracellular calcium (Ca2+ ) cycling is tightly regulated in the healthy heart ensuring effective contraction. This is achieved by transverse (t)-tubule membrane invaginations that facilitate close coupling of key Ca2+ -handling proteins such as the L-type Ca2+ channel and Na+ -Ca2+ exchanger (NCX) on the cell surface with ryanodine receptors (RyRs) on the intracellular Ca2+ store. Although less abundant and regular than in the ventricle, t-tubules also exist in atrial myocytes as a network of transverse invaginations with axial extensions known as the transverse-axial tubule system (TATS). In heart failure and atrial fibrillation, there is TATS remodelling that is associated with aberrant Ca2+ -handling and Ca2+ -induced arrhythmic activity; however, the mechanism underlying this is not fully understood. To address this, we developed a novel 3D human atrial myocyte model that couples electrophysiology and Ca2+ -handling with variable TATS organization and density. We extensively parameterized and validated our model against experimental data to build a robust tool examining TATS regulation of subcellular Ca2+ release. We found that varying TATS density and thus the localization of key Ca2+ -handling proteins has profound effects on Ca2+ handling. Following TATS loss, there is reduced NCX that results in increased cleft Ca2+ concentration through decreased Ca2+ extrusion. This elevated Ca2+ increases RyR open probability causing spontaneous Ca2+ releases and the promotion of arrhythmogenic waves (especially in the cell interior) leading to voltage instabilities through delayed afterdepolarizations. In summary, the present study demonstrates a mechanistic link between TATS remodelling and Ca2+ -driven proarrhythmic behaviour that probably reflects the arrhythmogenic state observed in disease. KEY POINTS: Transverse-axial tubule systems (TATS) modulate Ca2+ handling and excitation-contraction coupling in atrial myocytes, with TATS remodelling in heart failure and atrial fibrillation being associated with altered Ca2+ cycling and subsequent arrhythmogenesis. To investigate the poorly understood mechanisms linking TATS variation and spontaneous Ca2+ release, we built, parameterized and validated a 3D human atrial myocyte model coupling electrophysiology and spatially-detailed subcellular Ca2+ handling governed by the TATS. Simulated TATS loss causes diastolic Ca2+ and voltage instabilities through reduced Na+ -Ca2+ exchanger-mediated Ca2+ removal, cleft Ca2+ accumulation and increased ryanodine receptor open probability, resulting in spontaneous Ca2+ release and promotion of arrhythmogenic waves and delayed afterdepolarizations. At fast electrical rates typical of atrial tachycardia/fibrillation, spontaneous Ca2+ releases are larger and more frequent in the cell interior than at the periphery. Our work provides mechanistic insight into how atrial TATS remodelling can lead to Ca2+ -driven instabilities that may ultimately contribute to the arrhythmogenic state in disease.
Collapse
Affiliation(s)
- X. Zhang
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - H. Ni
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - S. Morotti
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - C.E.R. Smith
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - D. Sato
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - W.E. Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo Norway
| | - A.G. Edwards
- Department of Pharmacology, University of California Davis, Davis, CA, USA
- Simula Research Laboratory, Lysaker, Norway
| | - E. Grandi
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| |
Collapse
|
13
|
Wu Q, Ross AJ, Ipek T, Thompson GH, Johnson RD, Wu C, Camelliti P. Hydroxychloroquine and azithromycin alter the contractility of living porcine heart slices. Front Pharmacol 2023; 14:1127388. [PMID: 37214466 PMCID: PMC10196358 DOI: 10.3389/fphar.2023.1127388] [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: 12/19/2022] [Accepted: 04/24/2023] [Indexed: 05/24/2023] Open
Abstract
The cardiotoxicity risk of hydroxychloroquine (HCQ) and azithromycin (AZM) has been the subject of intensive research triggered by safety concerns in COVID-19 patients. HCQ and AZM have been associated with QT interval prolongation and drug-induced arrhythmias, however other cardiotoxicity mechanisms remain largely unexplored. Our group has pioneered the living heart slice preparation, an ex-vivo platform that maintains native cardiac tissue architecture and physiological electrical and contractile properties. Here, we evaluated the cardiotoxic effect of HCQ and AZM applied alone or in combination on cardiac contractility by measuring contractile force and contraction kinetics in heart slices prepared from porcine hearts. Our results show that clinically relevant concentrations of HCQ monotherapy (1-10 µM) reduced contractile force and contraction kinetics in porcine slices in a dose-dependent manner. However, AZM monotherapy decreased contractile force and contraction kinetics only at higher concentrations (30 µM). Combination of HCQ and AZM induced a dose-dependent effect similar to HCQ alone. Furthermore, pre-treating porcine heart slices with the L-type calcium channel agonist Bay K8644 prevented the effect of both drugs, while administration of Bay K8644 after drugs interventions largely reversed the effects, suggesting a mechanism involving inhibition of L-type calcium channels. These findings indicate that HCQ and AZM alter cardiac function beyond QT prolongation with significant contractile dysfunction in intact cardiac tissue. Our porcine heart slices provide a powerful platform to investigate mechanisms of drug cardiotoxicity.
Collapse
Affiliation(s)
- Qin Wu
- School of Medicine, Jiangsu Vocational College of Medicine, Yancheng, China
- School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| | - Abigail J. Ross
- School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| | - Tugce Ipek
- School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| | - Georgina H. Thompson
- School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| | - Robert D. Johnson
- School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| | - 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
|
14
|
Boukhalfa A, Robinson SR, Meola DM, Robinson NA, Ling LA, LaMastro JN, Upshaw JN, Pulakat L, Jaffe IZ, London CA, Chen HH, Yang VK. Using cultured canine cardiac slices to model the autophagic flux with doxorubicin. PLoS One 2023; 18:e0282859. [PMID: 36928870 PMCID: PMC10019679 DOI: 10.1371/journal.pone.0282859] [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: 01/18/2023] [Accepted: 02/19/2023] [Indexed: 03/18/2023] Open
Abstract
Chemotherapy-induced impairment of autophagy is implicated in cardiac toxicity induced by anti-cancer drugs. Imperfect translation from rodent models and lack of in vitro models of toxicity has limited investigation of autophagic flux dysregulation, preventing design of novel cardioprotective strategies based on autophagy control. Development of an adult heart tissue culture technique from a translational model will improve investigation of cardiac toxicity. We aimed to optimize a canine cardiac slice culture system for exploration of cancer therapy impact on intact cardiac tissue, creating a translatable model that maintains autophagy in culture and is amenable to autophagy modulation. Canine cardiac tissue slices (350 μm) were generated from left ventricular free wall collected from euthanized client-owned dogs (n = 7) free of cardiovascular disease at the Foster Hospital for Small Animals at Tufts University. Cell viability and apoptosis were quantified with MTT assay and TUNEL staining. Cardiac slices were challenged with doxorubicin and an autophagy activator (rapamycin) or inhibitor (chloroquine). Autophagic flux components (LC3, p62) were quantified by western blot. Cardiac slices retained high cell viability for >7 days in culture and basal levels of autophagic markers remained unchanged. Doxorubicin treatment resulted in perturbation of the autophagic flux and cell death, while rapamycin co-treatment restored normal autophagic flux and maintained cell survival. We developed an adult canine cardiac slice culture system appropriate for studying the effects of autophagic flux that may be applicable to drug toxicity evaluations.
Collapse
Affiliation(s)
- Asma Boukhalfa
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Sally R Robinson
- Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - Dawn M Meola
- Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - Nicholas A Robinson
- Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - Lauren A Ling
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Joey N LaMastro
- Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - Jenica N Upshaw
- Tufts University School of Medicine, Boston, Massachusetts, United States of America
- Division of Cardiology, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Lakshmi Pulakat
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
- Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
- Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Cheryl A London
- Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - Howard H Chen
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
- Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Vicky K Yang
- Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| |
Collapse
|
15
|
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: 0] [Impact Index Per Article: 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
|
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
|
Ripplinger CM, Glukhov AV, Kay MW, Boukens BJ, Chiamvimonvat N, Delisle BP, Fabritz L, Hund TJ, Knollmann BC, Li N, Murray KT, Poelzing S, Quinn TA, Remme CA, Rentschler SL, Rose RA, Posnack NG. Guidelines for assessment of cardiac electrophysiology and arrhythmias in small animals. Am J Physiol Heart Circ Physiol 2022; 323:H1137-H1166. [PMID: 36269644 PMCID: PMC9678409 DOI: 10.1152/ajpheart.00439.2022] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/11/2022] [Accepted: 10/17/2022] [Indexed: 01/09/2023]
Abstract
Cardiac arrhythmias are a major cause of morbidity and mortality worldwide. Although recent advances in cell-based models, including human-induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM), are contributing to our understanding of electrophysiology and arrhythmia mechanisms, preclinical animal studies of cardiovascular disease remain a mainstay. Over the past several decades, animal models of cardiovascular disease have advanced our understanding of pathological remodeling, arrhythmia mechanisms, and drug effects and have led to major improvements in pacing and defibrillation therapies. There exist a variety of methodological approaches for the assessment of cardiac electrophysiology and a plethora of parameters may be assessed with each approach. This guidelines article will provide an overview of the strengths and limitations of several common techniques used to assess electrophysiology and arrhythmia mechanisms at the whole animal, whole heart, and tissue level with a focus on small animal models. We also define key electrophysiological parameters that should be assessed, along with their physiological underpinnings, and the best methods with which to assess these parameters.
Collapse
Affiliation(s)
- Crystal M Ripplinger
- Department of Pharmacology, University of California Davis School of Medicine, Davis, California
| | - Alexey V Glukhov
- Department of Medicine, Cardiovascular Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Matthew W Kay
- Department of Biomedical Engineering, The George Washington University, Washington, District of Columbia
| | - Bastiaan J Boukens
- Department Physiology, University Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Medical Biology, University of Amsterdam, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Nipavan Chiamvimonvat
- Department of Pharmacology, University of California Davis School of Medicine, Davis, California
- Department of Internal Medicine, University of California Davis School of Medicine, Davis, California
- Veterans Affairs Northern California Healthcare System, Mather, California
| | - Brian P Delisle
- Department of Physiology, University of Kentucky, Lexington, Kentucky
| | - Larissa Fabritz
- University Center of Cardiovascular Science, University Heart and Vascular Center, University Hospital Hamburg-Eppendorf with DZHK Hamburg/Kiel/Luebeck, Germany
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Thomas J Hund
- Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
- Department of Biomedical Engineering, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Bjorn C Knollmann
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Na Li
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Katherine T Murray
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Steven Poelzing
- Virginia Tech Carilon School of Medicine, Center for Heart and Reparative Medicine Research, Fralin Biomedical Research Institute at Virginia Tech, Roanoke, Virginia
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - T Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
- School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Carol Ann Remme
- Department of Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Stacey L Rentschler
- Cardiovascular Division, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri
| | - Robert A Rose
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nikki G Posnack
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, District of Columbia
- Department of Pediatrics, George Washington University School of Medicine, Washington, District of Columbia
| |
Collapse
|
19
|
Perea-Gil I, Seeger T, Bruyneel AAN, Termglinchan V, Monte E, Lim EW, Vadgama N, Furihata T, Gavidia AA, Arthur Ataam J, Bharucha N, Martinez-Amador N, Ameen M, Nair P, Serrano R, Kaur B, Feyen DAM, Diecke S, Snyder MP, Metallo CM, Mercola M, Karakikes I. Serine biosynthesis as a novel therapeutic target for dilated cardiomyopathy. Eur Heart J 2022; 43:3477-3489. [PMID: 35728000 PMCID: PMC9794189 DOI: 10.1093/eurheartj/ehac305] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 04/14/2022] [Accepted: 05/24/2022] [Indexed: 12/30/2022] Open
Abstract
AIMS Genetic dilated cardiomyopathy (DCM) is a leading cause of heart failure. Despite significant progress in understanding the genetic aetiologies of DCM, the molecular mechanisms underlying the pathogenesis of familial DCM remain unknown, translating to a lack of disease-specific therapies. The discovery of novel targets for the treatment of DCM was sought using phenotypic sceening assays in induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) that recapitulate the disease phenotypes in vitro. METHODS AND RESULTS Using patient-specific iPSCs carrying a pathogenic TNNT2 gene mutation (p.R183W) and CRISPR-based genome editing, a faithful DCM model in vitro was developed. An unbiased phenotypic screening in TNNT2 mutant iPSC-derived cardiomyocytes (iPSC-CMs) with small molecule kinase inhibitors (SMKIs) was performed to identify novel therapeutic targets. Two SMKIs, Gö 6976 and SB 203580, were discovered whose combinatorial treatment rescued contractile dysfunction in DCM iPSC-CMs carrying gene mutations of various ontologies (TNNT2, TTN, LMNA, PLN, TPM1, LAMA2). The combinatorial SMKI treatment upregulated the expression of genes that encode serine, glycine, and one-carbon metabolism enzymes and significantly increased the intracellular levels of glucose-derived serine and glycine in DCM iPSC-CMs. Furthermore, the treatment rescued the mitochondrial respiration defects and increased the levels of the tricarboxylic acid cycle metabolites and ATP in DCM iPSC-CMs. Finally, the rescue of the DCM phenotypes was mediated by the activating transcription factor 4 (ATF4) and its downstream effector genes, phosphoglycerate dehydrogenase (PHGDH), which encodes a critical enzyme of the serine biosynthesis pathway, and Tribbles 3 (TRIB3), a pseudokinase with pleiotropic cellular functions. CONCLUSIONS A phenotypic screening platform using DCM iPSC-CMs was established for therapeutic target discovery. A combination of SMKIs ameliorated contractile and metabolic dysfunction in DCM iPSC-CMs mediated via the ATF4-dependent serine biosynthesis pathway. Together, these findings suggest that modulation of serine biosynthesis signalling may represent a novel genotype-agnostic therapeutic strategy for genetic DCM.
Collapse
Affiliation(s)
- Isaac Perea-Gil
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 240 Pasteur Dr, Stanford, CA 94304, USA
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Timon Seeger
- Department of Medicine III, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Arne A N Bruyneel
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Vittavat Termglinchan
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 240 Pasteur Dr, Stanford, CA 94304, USA
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Emma Monte
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Esther W Lim
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Nirmal Vadgama
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 240 Pasteur Dr, Stanford, CA 94304, USA
| | - Takaaki Furihata
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Alexandra A Gavidia
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 240 Pasteur Dr, Stanford, CA 94304, USA
| | - Jennifer Arthur Ataam
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 240 Pasteur Dr, Stanford, CA 94304, USA
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Nike Bharucha
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 240 Pasteur Dr, Stanford, CA 94304, USA
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Noel Martinez-Amador
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 240 Pasteur Dr, Stanford, CA 94304, USA
| | - Mohamed Ameen
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Pooja Nair
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 240 Pasteur Dr, Stanford, CA 94304, USA
| | - Ricardo Serrano
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Balpreet Kaur
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 240 Pasteur Dr, Stanford, CA 94304, USA
| | - Dries A M Feyen
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Sebastian Diecke
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Christian M Metallo
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Mark Mercola
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ioannis Karakikes
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 240 Pasteur Dr, Stanford, CA 94304, USA
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
20
|
Miller JM, Meki MH, Elnakib A, Ou Q, Abouleisa RRE, Tang XL, Salama ABM, Gebreil A, Lin C, Abdeltawab H, Khalifa F, Hill BG, Abi-Gerges N, Bolli R, El-Baz AS, Giridharan GA, Mohamed TMA. Biomimetic cardiac tissue culture model (CTCM) to emulate cardiac physiology and pathophysiology ex vivo. Commun Biol 2022; 5:934. [PMID: 36085302 PMCID: PMC9463130 DOI: 10.1038/s42003-022-03919-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
There is need for a reliable in vitro system that can accurately replicate the cardiac physiological environment for drug testing. The limited availability of human heart tissue culture systems has led to inaccurate interpretations of cardiac-related drug effects. Here, we developed a cardiac tissue culture model (CTCM) that can electro-mechanically stimulate heart slices with physiological stretches in systole and diastole during the cardiac cycle. After 12 days in culture, this approach partially improved the viability of heart slices but did not completely maintain their structural integrity. Therefore, following small molecule screening, we found that the incorporation of 100 nM tri-iodothyronine (T3) and 1 μM dexamethasone (Dex) into our culture media preserved the microscopic structure of the slices for 12 days. When combined with T3/Dex treatment, the CTCM system maintained the transcriptional profile, viability, metabolic activity, and structural integrity for 12 days at the same levels as the fresh heart tissue. Furthermore, overstretching the cardiac tissue induced cardiac hypertrophic signaling in culture, which provides a proof of concept for the ability of the CTCM to emulate cardiac stretch-induced hypertrophic conditions. In conclusion, CTCM can emulate cardiac physiology and pathophysiology in culture for an extended time, thereby enabling reliable drug screening.
Collapse
Affiliation(s)
- Jessica M Miller
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, USA
- Department of Bioengineering, University of Louisville, Louisville, USA
| | - Moustafa H Meki
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, USA
- Department of Bioengineering, University of Louisville, Louisville, USA
| | - Ahmed Elnakib
- Department of Bioengineering, University of Louisville, Louisville, USA
| | - Qinghui Ou
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, USA
| | - Riham R E Abouleisa
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, USA
| | - Xian-Liang Tang
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, USA
| | - Abou Bakr M Salama
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, USA
- Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ahmad Gebreil
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, USA
| | - Cindy Lin
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, USA
| | - Hisham Abdeltawab
- Department of Bioengineering, University of Louisville, Louisville, USA
| | - Fahmi Khalifa
- Department of Bioengineering, University of Louisville, Louisville, USA
| | - Bradford G Hill
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, USA
| | | | - Roberto Bolli
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, USA
| | - Ayman S El-Baz
- Department of Bioengineering, University of Louisville, Louisville, USA
| | | | - Tamer M A Mohamed
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, USA.
- Department of Bioengineering, University of Louisville, Louisville, USA.
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, USA.
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, USA.
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
21
|
Basara G, Bahcecioglu G, Ozcebe SG, Ellis BW, Ronan G, Zorlutuna P. Myocardial infarction from a tissue engineering and regenerative medicine point of view: A comprehensive review on models and treatments. BIOPHYSICS REVIEWS 2022; 3:031305. [PMID: 36091931 PMCID: PMC9447372 DOI: 10.1063/5.0093399] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/08/2022] [Indexed: 05/12/2023]
Abstract
In the modern world, myocardial infarction is one of the most common cardiovascular diseases, which are responsible for around 18 million deaths every year or almost 32% of all deaths. Due to the detrimental effects of COVID-19 on the cardiovascular system, this rate is expected to increase in the coming years. Although there has been some progress in myocardial infarction treatment, translating pre-clinical findings to the clinic remains a major challenge. One reason for this is the lack of reliable and human representative healthy and fibrotic cardiac tissue models that can be used to understand the fundamentals of ischemic/reperfusion injury caused by myocardial infarction and to test new drugs and therapeutic strategies. In this review, we first present an overview of the anatomy of the heart and the pathophysiology of myocardial infarction, and then discuss the recent developments on pre-clinical infarct models, focusing mainly on the engineered three-dimensional cardiac ischemic/reperfusion injury and fibrosis models developed using different engineering methods such as organoids, microfluidic devices, and bioprinted constructs. We also present the benefits and limitations of emerging and promising regenerative therapy treatments for myocardial infarction such as cell therapies, extracellular vesicles, and cardiac patches. This review aims to overview recent advances in three-dimensional engineered infarct models and current regenerative therapeutic options, which can be used as a guide for developing new models and treatment strategies.
Collapse
Affiliation(s)
- Gozde Basara
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Gokhan Bahcecioglu
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - S. Gulberk Ozcebe
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Bradley W Ellis
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - George Ronan
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Pinar Zorlutuna
- Present address: 143 Multidisciplinary Research Building, University of Notre Dame, Notre Dame, IN 46556. Author to whom correspondence should be addressed:. Tel.: +1 574 631 8543. Fax: +1 574 631 8341
| |
Collapse
|
22
|
Biendarra‐Tiegs SM, Yechikov S, Shergill B, Brumback B, Takahashi K, Shirure VS, Gonzalez RE, Houshmand L, Zhong D, Weng K, Silva J, Smith TW, Rentschler SL, George SC. An iPS-derived in vitro model of human atrial conduction. Physiol Rep 2022; 10:e15407. [PMID: 36117385 PMCID: PMC9483613 DOI: 10.14814/phy2.15407] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/27/2022] [Accepted: 07/14/2022] [Indexed: 11/25/2022] Open
Abstract
Atrial fibrillation (AF) is the most common arrhythmia in the United States, affecting approximately 1 in 10 adults, and its prevalence is expected to rise as the population ages. Treatment options for AF are limited; moreover, the development of new treatments is hindered by limited (1) knowledge regarding human atrial electrophysiological endpoints (e.g., conduction velocity [CV]) and (2) accurate experimental models. Here, we measured the CV and refractory period, and subsequently calculated the conduction wavelength, in vivo (four subjects with AF and four controls), and ex vivo (atrial slices from human hearts). Then, we created an in vitro model of human atrial conduction using induced pluripotent stem (iPS) cells. This model consisted of iPS-derived human atrial cardiomyocytes plated onto a micropatterned linear 1D spiral design of Matrigel. The CV (34-41 cm/s) of the in vitro model was nearly five times faster than 2D controls (7-9 cm/s) and similar to in vivo (40-64 cm/s) and ex vivo (28-51 cm/s) measurements. Our iPS-derived in vitro model recapitulates key features of in vivo atrial conduction and may be a useful methodology to enhance our understanding of AF and model patient-specific disease.
Collapse
Affiliation(s)
| | - Sergey Yechikov
- Department of Biomedical EngineeringUniversity of California, DavisDavisCaliforniaUSA
| | - Bhupinder Shergill
- Department of Biomedical EngineeringUniversity of California, DavisDavisCaliforniaUSA
| | - Brittany Brumback
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
| | - Kentaro Takahashi
- Department of MedicineWashington University in St. LouisSt. LouisMissouriUSA
| | - Venktesh S. Shirure
- Department of Biomedical EngineeringUniversity of California, DavisDavisCaliforniaUSA
| | - Ruth Estelle Gonzalez
- Department of Biomedical EngineeringUniversity of California, DavisDavisCaliforniaUSA
| | - Laura Houshmand
- Department of Biomedical EngineeringUniversity of California, DavisDavisCaliforniaUSA
| | - Denise Zhong
- Department of Biomedical EngineeringUniversity of California, DavisDavisCaliforniaUSA
| | - Kuo‐Chan Weng
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
| | - Jon Silva
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
| | - Timothy W. Smith
- Department of MedicineWashington University in St. LouisSt. LouisMissouriUSA
| | - Stacey L. Rentschler
- Department of MedicineWashington University in St. LouisSt. LouisMissouriUSA
- Department of Developmental BiologyWashington University in St. LouisSt. LouisMissouriUSA
| | - Steven C. George
- Department of Biomedical EngineeringUniversity of California, DavisDavisCaliforniaUSA
| |
Collapse
|
23
|
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
|
24
|
Kurekova S, Tomaskova ZS, Andelova N, Macejova D, Cervienkova M, Brtko J, Ferko M, Grman M, Mackova K. The effect of all-trans retinoic acid on the mitochondrial function and survival of cardiomyoblasts exposed to local photodamage. Cell Biol Int 2022; 46:947-964. [PMID: 35191136 DOI: 10.1002/cbin.11784] [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: 04/12/2021] [Revised: 12/30/2021] [Accepted: 02/12/2022] [Indexed: 11/06/2022]
Abstract
This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Simona Kurekova
- Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, 840 05, Bratislava, Slovakia
| | - Zuzana Sevcikova Tomaskova
- Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, 840 05, Bratislava, Slovakia
| | - Natalia Andelova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104, Bratislava, Slovakia
| | - Dana Macejova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 84505, Bratislava, Slovakia
| | - Michaela Cervienkova
- Institute of Biochemistry and Microbiology, Faculty of Chemical and Food Technology, Slovak University of Technology, 81237, Bratislava, Slovakia
| | - Julius Brtko
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 84505, Bratislava, Slovakia
| | - Miroslav Ferko
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104, Bratislava, Slovakia
| | - Marian Grman
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, 84505, Bratislava, Slovakia
| | - Katarina Mackova
- Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, 840 05, Bratislava, Slovakia
| |
Collapse
|
25
|
Abdeltawab H, Khalifa F, Hammouda K, Miller JM, Meki MM, Ou Q, El-Baz A, Mohamed TMA. Artificial Intelligence Based Framework to Quantify the Cardiomyocyte Structural Integrity in Heart Slices. Cardiovasc Eng Technol 2022; 13:170-180. [PMID: 34402037 PMCID: PMC8847536 DOI: 10.1007/s13239-021-00571-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 07/26/2021] [Indexed: 02/03/2023]
Abstract
PURPOSE Drug induced cardiac toxicity is a disruption of the functionality of cardiomyocytes which is highly correlated to the organization of the subcellular structures. We can analyze cellular structures by utilizing microscopy imaging data. However, conventional image analysis methods might miss structural deteriorations that are difficult to perceive. Here, we propose an image-based deep learning pipeline for the automated quantification of drug induced structural deteriorations using a 3D heart slice culture model. METHODS In our deep learning pipeline, we quantify the induced structural deterioration from three anticancer drugs (doxorubicin, sunitinib, and herceptin) with known adverse cardiac effects. The proposed deep learning framework is composed of three convolutional neural networks that process three different image sizes. The results of the three networks are combined to produce a classification map that shows the locations of the structural deteriorations in the input cardiac image. RESULTS The result of our technique is the capability of producing classification maps that accurately detect drug induced structural deterioration on the pixel level. CONCLUSION This technology could be widely applied to perform unbiased quantification of the structural effect of the cardiotoxins on heart slices.
Collapse
Affiliation(s)
- Hisham Abdeltawab
- BioImaging Laboratory, Department of Bioengineering, University of Louisville, Louisville, KY, USA
| | - Fahmi Khalifa
- BioImaging Laboratory, Department of Bioengineering, University of Louisville, Louisville, KY, USA
| | - Kamal Hammouda
- BioImaging Laboratory, Department of Bioengineering, University of Louisville, Louisville, KY, USA
| | - Jessica M. Miller
- Division of Cardiovascular Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Moustafa M. Meki
- Division of Cardiovascular Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Qinghui Ou
- Division of Cardiovascular Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Ayman El-Baz
- BioImaging Laboratory, Department of Bioengineering, University of Louisville, Louisville, KY, USA
| | - Tamer M. A. Mohamed
- Division of Cardiovascular Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA
| |
Collapse
|
26
|
Koncz I, Verkerk AO, Nicastro M, Wilders R, Árpádffy-Lovas T, Magyar T, Tóth N, Nagy N, Madrid M, Lin Z, Efimov IR. Acetylcholine Reduces IKr and Prolongs Action Potentials in Human Ventricular Cardiomyocytes. Biomedicines 2022; 10:biomedicines10020244. [PMID: 35203454 PMCID: PMC8869322 DOI: 10.3390/biomedicines10020244] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 02/07/2023] Open
Abstract
Vagal nerve stimulation (VNS) has a meaningful basis as a potentially effective treatment for heart failure with reduced ejection fraction. There is an ongoing VNS randomized study, and four studies are completed. However, relatively little is known about the effect of acetylcholine (ACh) on repolarization in human ventricular cardiomyocytes, as well as the effect of ACh on the rapid component of the delayed rectifier K+ current (IKr). Here, we investigated the effect of ACh on the action potential parameters in human ventricular preparations and on IKr in human induced pluripotent stem-cell-derived cardiomyocytes (hiPSC-CMs). Using standard microelectrode technique, we demonstrated that ACh (5 µM) significantly increased the action potential duration in human left ventricular myocardial slices. ACh (5 µM) also prolonged repolarization in a human Purkinje fiber and a papillary muscle. Optical mapping revealed that ACh increased the action potential duration in human left ventricular myocardial slices and that the effect was dose-dependent. Perforated patch clamp experiments demonstrated action potential prolongation and a significant decrease in IKr by ACh (5 µM) in hiPSC-CMs. Computer simulations of the electrical activity of a human ventricular cardiomyocyte showed an increase in action potential duration upon implementation of the experimentally observed ACh-induced changes in the fully activated conductance and steady-state activation of IKr. Our findings support the hypothesis that ACh can influence the repolarization in human ventricular cardiomyocytes by at least changes in IKr.
Collapse
Affiliation(s)
- István Koncz
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA; (I.K.); (M.M.); (Z.L.)
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary; (T.Á.-L.); (T.M.); (N.T.); (N.N.)
| | - Arie O. Verkerk
- Heart Center, Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (A.O.V.); (M.N.)
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Michele Nicastro
- Heart Center, Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (A.O.V.); (M.N.)
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Tamás Árpádffy-Lovas
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary; (T.Á.-L.); (T.M.); (N.T.); (N.N.)
| | - Tibor Magyar
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary; (T.Á.-L.); (T.M.); (N.T.); (N.N.)
| | - Noémi Tóth
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary; (T.Á.-L.); (T.M.); (N.T.); (N.N.)
| | - Norbert Nagy
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary; (T.Á.-L.); (T.M.); (N.T.); (N.N.)
- ELKH-SZTE Research Group of Cardiovascular Pharmacology, 6721 Szeged, Hungary
| | - Micah Madrid
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA; (I.K.); (M.M.); (Z.L.)
| | - Zexu Lin
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA; (I.K.); (M.M.); (Z.L.)
| | - Igor R. Efimov
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA; (I.K.); (M.M.); (Z.L.)
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Chicago, IL 60611, USA
- Correspondence: ; Tel.: +1-202-294-8182
| |
Collapse
|
27
|
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: 1.3] [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
|
28
|
Honkoop H, Nguyen PD, van der Velden VEM, Sonnen KF, Bakkers J. Live imaging of adult zebrafish cardiomyocyte proliferation ex vivo. Development 2021; 148:271839. [PMID: 34397091 PMCID: PMC8489017 DOI: 10.1242/dev.199740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/04/2021] [Indexed: 12/24/2022]
Abstract
Zebrafish are excellent at regenerating their heart by reinitiating proliferation in pre-existing cardiomyocytes. Studying how zebrafish achieve this holds great potential in developing new strategies to boost mammalian heart regeneration. Nevertheless, the lack of appropriate live-imaging tools for the adult zebrafish heart has limited detailed studies into the dynamics underlying cardiomyocyte proliferation. Here, we address this by developing a system in which cardiac slices of the injured zebrafish heart are cultured ex vivo for several days while retaining key regenerative characteristics, including cardiomyocyte proliferation. In addition, we show that the cardiac slice culture system is compatible with live timelapse imaging and allows manipulation of regenerating cardiomyocytes with drugs that normally would have toxic effects that prevent their use. Finally, we use the cardiac slices to demonstrate that adult cardiomyocytes with fully assembled sarcomeres can partially disassemble their sarcomeres in a calpain- and proteasome-dependent manner to progress through nuclear division and cytokinesis. In conclusion, we have developed a cardiac slice culture system, which allows imaging of native cardiomyocyte dynamics in real time to discover cellular mechanisms during heart regeneration.
Collapse
Affiliation(s)
- Hessel Honkoop
- Hubrecht Institute-KNAW and Utrecht University Medical Center, Utrecht 3584CT, The Netherlands
| | - Phong D Nguyen
- Hubrecht Institute-KNAW and Utrecht University Medical Center, Utrecht 3584CT, The Netherlands
| | | | - Katharina F Sonnen
- Hubrecht Institute-KNAW and Utrecht University Medical Center, Utrecht 3584CT, The Netherlands
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW and Utrecht University Medical Center, Utrecht 3584CT, The Netherlands.,Department of Pediatric Cardiology, Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584EA, The Netherlands
| |
Collapse
|
29
|
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: 0.7] [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
|
30
|
Best Practices and Progress in Precision-Cut Liver Slice Cultures. Int J Mol Sci 2021; 22:ijms22137137. [PMID: 34281187 PMCID: PMC8267882 DOI: 10.3390/ijms22137137] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 12/31/2022] Open
Abstract
Thirty-five years ago, precision-cut liver slices (PCLS) were described as a promising tool and were expected to become the standard in vitro model to study liver disease as they tick off all characteristics of a good in vitro model. In contrast to most in vitro models, PCLS retain the complex 3D liver structures found in vivo, including cell–cell and cell–matrix interactions, and therefore should constitute the most reliable tool to model and to investigate pathways underlying chronic liver disease in vitro. Nevertheless, the biggest disadvantage of the model is the initiation of a procedure-induced fibrotic response. In this review, we describe the parameters and potential of PCLS cultures and discuss whether the initially described limitations and pitfalls have been overcome. We summarize the latest advances in PCLS research and critically evaluate PCLS use and progress since its invention in 1985.
Collapse
|
31
|
Dries E, Bardi I, Nunez-Toldra R, Meijlink B, Terracciano CM. CaMKII inhibition reduces arrhythmogenic Ca2+ events in subendocardial cryoinjured rat living myocardial slices. J Gen Physiol 2021; 153:212078. [PMID: 33956073 PMCID: PMC8105719 DOI: 10.1085/jgp.202012737] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 04/13/2021] [Indexed: 01/08/2023] Open
Abstract
Spontaneous Ca2+ release (SCR) can cause triggered activity and initiate arrhythmias. Intrinsic transmural heterogeneities in Ca2+ handling and their propensity to disease remodeling may differentially modulate SCR throughout the left ventricular (LV) wall and cause transmural differences in arrhythmia susceptibility. Here, we aimed to dissect the effect of cardiac injury on SCR in different regions in the intact LV myocardium using cryoinjury on rat living myocardial slices (LMS). We studied SCR under proarrhythmic conditions using a fluorescent Ca2+ indicator and high-resolution imaging in LMS from the subendocardium (ENDO) and subepicardium (EPI). Cryoinjury caused structural remodeling, with loss in T-tubule density and an increased time of Ca2+ transients to peak after injury. In ENDO LMS, the Ca2+ transient amplitude and decay phase were reduced, while these were not affected in EPI LMS after cryoinjury. The frequency of spontaneous whole-slice contractions increased in ENDO LMS without affecting EPI LMS after injury. Cryoinjury caused an increase in foci that generates SCR in both ENDO and EPI LMS. In ENDO LMS, SCRs were more closely distributed and had reduced latencies after cryoinjury, whereas this was not affected in EPI LMS. Inhibition of CaMKII reduced the number, distribution, and latencies of SCR, as well as whole-slice contractions in ENDO LMS, but not in EPI LMS after cryoinjury. Furthermore, CaMKII inhibition did not affect the excitation–contraction coupling in cryoinjured ENDO or EPI LMS. In conclusion, we demonstrate increased arrhythmogenic susceptibility in the injured ENDO. Our findings show involvement of CaMKII and highlight the need for region-specific targeting in cardiac therapies.
Collapse
Affiliation(s)
- Eef Dries
- National Heart and Lung Institute, Imperial College London, London, UK.,Lab of Experimental Cardiology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Ifigeneia Bardi
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Bram Meijlink
- National Heart and Lung Institute, Imperial College London, London, UK
| | | |
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: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [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
- 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
- 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
- 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
|
Microelectrode Arrays: A Valuable Tool to Analyze Stem Cell-Derived Cardiomyocytes. Stem Cells 2021. [DOI: 10.1007/978-3-030-77052-5_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
34
|
House A, Atalla I, Lee EJ, Guvendiren M. Designing Biomaterial Platforms for Cardiac Tissue and Disease Modeling. ADVANCED NANOBIOMED RESEARCH 2021; 1:2000022. [PMID: 33709087 PMCID: PMC7942203 DOI: 10.1002/anbr.202000022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Heart disease is one of the leading causes of death in the world. There is a growing demand for in vitro cardiac models that can recapitulate the complex physiology of the cardiac tissue. These cardiac models can provide a platform to better understand the underlying mechanisms of cardiac development and disease and aid in developing novel treatment alternatives and platforms towards personalized medicine. In this review, a summary of engineered cardiac platforms is presented. Basic design considerations for replicating the heart's microenvironment are discussed considering the anatomy of the heart. This is followed by a detailed summary of the currently available biomaterial platforms for modeling the heart tissue in vitro. These in vitro models include 2D surface modified structures, 3D molded structures, porous scaffolds, electrospun scaffolds, bioprinted structures, and heart-on-a-chip devices. The challenges faced by current models and the future directions of in vitro cardiac models are also discussed. Engineered in vitro tissue models utilizing patients' own cells could potentially revolutionize the way we develop treatment and diagnostic alternatives.
Collapse
Affiliation(s)
- Andrew House
- Instructive Biomaterials and Additive Manufacturing Laboratory, Otto H. York Chemical and Materials Engineering, 138 York Center, University Heights, Newark, NJ 07102, USA
| | - Iren Atalla
- Instructive Biomaterials and Additive Manufacturing Laboratory, Otto H. York Chemical and Materials Engineering, 138 York Center, University Heights, Newark, NJ 07102, USA
| | - Eun Jung Lee
- Instructive Biomaterials and Additive Manufacturing Laboratory, Otto H. York Chemical and Materials Engineering, 138 York Center, University Heights, Newark, NJ 07102, USA
| | - Murat Guvendiren
- Instructive Biomaterials and Additive Manufacturing Laboratory, Otto H. York Chemical and Materials Engineering, 138 York Center, University Heights, Newark, NJ 07102, USA
| |
Collapse
|
35
|
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: 1.0] [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
|
36
|
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: 2.0] [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
|
37
|
Mikhailov AV, Kalyanasundaram A, Li N, Scott SS, Artiga EJ, Subr MM, Zhao J, Hansen BJ, Hummel JD, Fedorov VV. Comprehensive evaluation of electrophysiological and 3D structural features of human atrial myocardium with insights on atrial fibrillation maintenance mechanisms. J Mol Cell Cardiol 2020; 151:56-71. [PMID: 33130148 DOI: 10.1016/j.yjmcc.2020.10.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/14/2022]
Abstract
Atrial fibrillation (AF) occurrence and maintenance is associated with progressive remodeling of electrophysiological (repolarization and conduction) and 3D structural (fibrosis, fiber orientations, and wall thickness) features of the human atria. Significant diversity in AF etiology leads to heterogeneous arrhythmogenic electrophysiological and structural substrates within the 3D structure of the human atria. Since current clinical methods have yet to fully resolve the patient-specific arrhythmogenic substrates, mechanism-based AF treatments remain underdeveloped. Here, we review current knowledge from in-vivo, ex-vivo, and in-vitro human heart studies, and discuss how these studies may provide new insights on the synergy of atrial electrophysiological and 3D structural features in AF maintenance. In-vitro studies on surgically acquired human atrial samples provide a great opportunity to study a wide spectrum of AF pathology, including functional changes in single-cell action potentials, ion channels, and gene/protein expression. However, limited size of the samples prevents evaluation of heterogeneous AF substrates and reentrant mechanisms. In contrast, coronary-perfused ex-vivo human hearts can be studied with state-of-the-art functional and structural technologies, such as high-resolution near-infrared optical mapping and contrast-enhanced MRI. These imaging modalities can resolve atrial arrhythmogenic substrates and their role in reentrant mechanisms maintaining AF and validate clinical approaches. Nonetheless, longitudinal studies are not feasible in explanted human hearts. As no approach is perfect, we suggest that combining the strengths of direct human atrial studies with high fidelity approaches available in the laboratory and in realistic patient-specific computer models would elucidate deeper knowledge of AF mechanisms. We propose that a comprehensive translational pipeline from ex-vivo human heart studies to longitudinal clinically relevant AF animal studies and finally to clinical trials is necessary to identify patient-specific arrhythmogenic substrates and develop novel AF treatments.
Collapse
Affiliation(s)
- Aleksei V Mikhailov
- Department of Physiology & Cell Biology, Bob and Corrine Frick Center for Heart Failure and Arrhythmia, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Arrhythmology Research Department, Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - Anuradha Kalyanasundaram
- Department of Physiology & Cell Biology, Bob and Corrine Frick Center for Heart Failure and Arrhythmia, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ning Li
- Department of Physiology & Cell Biology, Bob and Corrine Frick Center for Heart Failure and Arrhythmia, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Shane S Scott
- Department of Physiology & Cell Biology, Bob and Corrine Frick Center for Heart Failure and Arrhythmia, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Esthela J Artiga
- Department of Physiology & Cell Biology, Bob and Corrine Frick Center for Heart Failure and Arrhythmia, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Megan M Subr
- Department of Physiology & Cell Biology, Bob and Corrine Frick Center for Heart Failure and Arrhythmia, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Jichao Zhao
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Brian J Hansen
- Department of Physiology & Cell Biology, Bob and Corrine Frick Center for Heart Failure and Arrhythmia, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - John D Hummel
- Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Vadim V Fedorov
- Department of Physiology & Cell Biology, Bob and Corrine Frick Center for Heart Failure and Arrhythmia, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
38
|
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: 2.5] [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
|
39
|
Miller JM, Meki MH, Ou Q, George SA, Gams A, Abouleisa RRE, Tang XL, Ahern BM, Giridharan GA, El-Baz A, Hill BG, Satin J, Conklin DJ, Moslehi J, Bolli R, Ribeiro AJS, Efimov IR, Mohamed TMA. Heart slice culture system reliably demonstrates clinical drug-related cardiotoxicity. Toxicol Appl Pharmacol 2020; 406:115213. [PMID: 32877659 DOI: 10.1016/j.taap.2020.115213] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 02/07/2023]
Abstract
The limited availability of human heart tissue and its complex cell composition are major limiting factors for the reliable testing of drug efficacy and toxicity. Recently, we developed functional human and pig heart slice biomimetic culture systems that preserve the viability and functionality of 300 μm heart slices for up to 6 days. Here, we tested the reliability of this culture system for testing the cardiotoxicity of anti-cancer drugs. We tested three anti-cancer drugs (doxorubicin, trastuzumab, and sunitinib) with known different mechanisms of cardiotoxicity at three concentrations and assessed the effect of these drugs on heart slice viability, structure, function and gene expression. Slices incubated with any of these drugs for 48 h showed diminished in viability as well as loss of cardiomyocyte structure and function. Mechanistically, RNA sequencing of doxorubicin-treated tissues demonstrated a significant downregulation of cardiac genes and upregulation of oxidative stress responses. Trastuzumab treatment downregulated cardiac muscle contraction-related genes consistent with its clinically known effect on cardiomyocytes. Interestingly, sunitinib treatment resulted in significant downregulation of angiogenesis-related genes, in line with its mechanism of action. Similar to hiPS-derived-cardiomyocytes, heart slices recapitulated the expected toxicity of doxorubicin and trastuzumab, however, slices were superior in detecting sunitinib cardiotoxicity and mechanism in the clinically relevant concentration range of 0.1-1 μM. These results indicate that heart slice culture models have the potential to become a reliable platform for testing and elucidating mechanisms of drug cardiotoxicity.
Collapse
Affiliation(s)
- Jessica M Miller
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA; Department of Bioengineering, University of Louisville, KY, USA
| | - Moustafa H Meki
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA; Department of Bioengineering, University of Louisville, KY, USA
| | - Qinghui Ou
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA
| | - Sharon A George
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
| | - Anna Gams
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
| | - Riham R E Abouleisa
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA
| | - Xian-Liang Tang
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA
| | - Brooke M Ahern
- Department of Physiology, University of Kentucky, KY, USA
| | | | - Ayman El-Baz
- Department of Bioengineering, University of Louisville, KY, USA
| | - Bradford G Hill
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, KY, USA
| | - Jonathan Satin
- Department of Physiology, University of Kentucky, KY, USA
| | - Daniel J Conklin
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, KY, USA
| | - Javid Moslehi
- Division of Cardiology, Cardio-Oncology Program, Vanderbilt University Medical Center, 2220 Pierce Avenue, Nashville, USA
| | - Roberto Bolli
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA
| | - Alexandre J S Ribeiro
- U.S. Food and Drug Administration, Center for Drug Evaluation and Research, Office of Translational Science, Office of Clinical Pharmacology, Division of Applied Regulatory Science, Silver Spring, MD, USA.
| | - Igor R Efimov
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA.
| | - Tamer M A Mohamed
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA; Department of Bioengineering, University of Louisville, KY, USA; Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, KY, USA; Department of Pharmacology and Toxicology, University of Louisville, KY, USA; Institute of Cardiovascular Sciences, University of Manchester, UK; Faculty of Pharmacy, Zagazig University, Egypt.
| |
Collapse
|
40
|
Hassan S, Barrett CJ, Crossman DJ. Imaging tools for assessment of myocardial fibrosis in humans: the need for greater detail. Biophys Rev 2020; 12:969-987. [PMID: 32705483 PMCID: PMC7429810 DOI: 10.1007/s12551-020-00738-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/08/2020] [Indexed: 02/06/2023] Open
Abstract
Myocardial fibrosis is recognized as a key pathological process in the development of cardiac disease and a target for future therapeutics. Despite this recognition, the assessment of fibrosis is not a part of routine clinical practice. This is primarily due to the difficulties in obtaining an accurate assessment of fibrosis non-invasively. Moreover, there is a clear discrepancy between the understandings of myocardial fibrosis clinically where fibrosis is predominately studied with comparatively low-resolution medical imaging technologies like MRI compared with the basic science laboratories where fibrosis can be visualized invasively with high resolution using molecularly specific fluorescence microscopes at the microscopic and nanoscopic scales. In this article, we will first review current medical imaging technologies for assessing fibrosis including echo and MRI. We will then highlight the need for greater microscopic and nanoscopic analysis of human tissue and how this can be addressed through greater utilization of human tissue available through endomyocardial biopsies and cardiac surgeries. We will then describe the relatively new field of molecular imaging that promises to translate research findings to the clinical practice by non-invasively monitoring the molecular signature of fibrosis in patients.
Collapse
Affiliation(s)
- Summer Hassan
- Department of Physiology, University of Auckland, Auckland, New Zealand
- Auckland City Hospital, Auckland District Health Board, Auckland, New Zealand
| | - Carolyn J Barrett
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | - David J Crossman
- Department of Physiology, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
41
|
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: 7.5] [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
|
42
|
Slotvitsky MM, Tsvelaya VA, Podgurskaya AD, Agladze KI. Formation of an electrical coupling between differentiating cardiomyocytes. Sci Rep 2020; 10:7774. [PMID: 32385315 PMCID: PMC7210299 DOI: 10.1038/s41598-020-64581-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/08/2020] [Indexed: 01/02/2023] Open
Abstract
Human induced pluripotent stem cell–derived cardiomyocytes (hiPSC-CMs) serve as an indispensable platform for the study of human cardiovascular disease is human induced pluripotent stem cell–derived cardiomyocytes (hiPSC-CMs). While the possibility of reproducing rare pathologies, patient-specific selection of drugs, and other issues concerning single cardiomyocytes have been well studied, little attention has been paid to the properties of the whole syncytium of CMs, in which both the functionality of individual cells and the distribution of electrophysiological connections between them are essential. The aim of this work is to directly study the ability of hiPSC-CMs to form a functional syncytium that can stably conduct an excitation wave. For that purpose, syncytium forming hiPSC-CMs were harvested and seeded (transferred) on a new substrate on different days of differentiation. The excitation conduction in a sample was characterized by the stability of the wavefront using optical mapping data. We found that the cells transferred before the 20th day of differentiation were able to organize a functional syncytium capable of further development and stable excitation conduction at high stimulation frequencies, while the cells transferred after 20 days did not form a homogeneous syncytium, and multiple instabilities of the propagating wavefront were observed with the possibility of reentry formation.
Collapse
Affiliation(s)
- M M Slotvitsky
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141700, Russian Federation
| | - V A Tsvelaya
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141700, Russian Federation
| | - A D Podgurskaya
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141700, Russian Federation
| | - K I Agladze
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141700, Russian Federation. .,M.F. Vladimirsky Moscow Regional Clinical Research Institute, Moscow, 129110, Russian Federation.
| |
Collapse
|
43
|
Kirschner Peretz N, Segal S, Yaniv Y. May the Force Not Be With You During Culture: Eliminating Mechano-Associated Feedback During Culture Preserves Cultured Atrial and Pacemaker Cell Functions. Front Physiol 2020; 11:163. [PMID: 32265724 PMCID: PMC7100534 DOI: 10.3389/fphys.2020.00163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/12/2020] [Indexed: 01/24/2023] Open
Abstract
Cultured cardiomyocytes have been shown to possess significant potential as a model for characterization of mechano-Ca2+, mechano-electric, and mechano-metabolic feedbacks in the heart. However, the majority of cultured cardiomyocytes exhibit impaired electrical, mechanical, biochemical, and metabolic functions. More specifically, the cells do not beat spontaneously (pacemaker cells) or beat at a rate far lower than their physiological counterparts and self-oscillate (atrial and ventricular cells) in culture. Thus, efforts are being invested in ensuring that cultured cardiomyocytes maintain the shape and function of freshly isolated cells. Elimination of contraction during culture has been shown to preserve the mechano-Ca2+, mechano-electric, and mechano-metabolic feedback loops of cultured cells. This review focuses on pacemaker cells, which reside in the sinoatrial node (SAN) and generate regular heartbeat through the initiation of the heart’s electrical, metabolic, and biochemical activities. In parallel, it places emphasis on atrial cells, which are responsible for bridging the electrical conductance from the SAN to the ventricle. The review provides a summary of the main mechanisms responsible for mechano-electrical, Ca2+, and metabolic feedback in pacemaker and atrial cells and of culture methods existing for both cell types. The work concludes with an explanation of how the elimination of mechano-electrical, mechano-Ca2+, and mechano-metabolic feedbacks during culture results in sustained cultured cell function.
Collapse
Affiliation(s)
- Noa Kirschner Peretz
- Biomedical Engineering Faculty, Technion Israel Institute of Technology, Haifa, Israel
| | - Sofia Segal
- Biomedical Engineering Faculty, Technion Israel Institute of Technology, Haifa, Israel
| | - Yael Yaniv
- Biomedical Engineering Faculty, Technion Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
44
|
Gintant G, Traebert M. The roles of human induced pluripotent stem cell-derived cardiomyocytes in drug discovery: managing in vitro safety study expectations. Expert Opin Drug Discov 2020; 15:719-729. [PMID: 32129680 DOI: 10.1080/17460441.2020.1736549] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) preparations are increasingly employed in in vitro cardiac safety studies to support candidate drug selection and regulatory submissions. The value of hiPSC-CM-based approaches depends on their ability to recapitulate the cellular mechanisms responsible for cardiotoxicity as well as overall assay characteristics (thus defining model performance). Different expectations at different drug development stages define the utility of these human-derived models. AREAS COVERED Herein, the authors review the importance of understanding the functional characteristics of the evolving spectrum of simpler (2D) and more complex (co-cultures, 3D constructs, and engineered tissues) human-derived cardiac preparations, and how their performance may be evaluated based on analytical sensitivity, variability, and reproducibility in order to correctly match preparations with expectations of different safety assays. The need for consensus clinical examples of electrophysiologic, contractile, and structural cardiotoxicities essential for benchmarking human-derived models is also discussed. EXPERT OPINION It is helpful (but not essential) that hiPSC-CMs preparations fully recapitulate pharmacological responses of native adult human ventricular myocytes when evaluating cardiotoxicity in vitro. Further calibration and model standardization (aligning concordance with clinical findings) are necessary to understand the role of hiPSC-CMs in guiding cardiotoxicity assessments in early drug discovery efforts.
Collapse
Affiliation(s)
- Gary Gintant
- Department of Integrative Pharmacology (ZR13), AP-9A-LL, AbbVie Inc. , North Chicago, IL, USA
| | - Martin Traebert
- Novartis Institutes for Biomedical Research , Safety Pharmacology, Basel, Switzerland
| |
Collapse
|
45
|
Ou Q, Abouleisa RRE, Tang XL, Juhardeen HR, Meki MH, Miller JM, Giridharan G, El-Baz A, Bolli R, Mohamed TMA. Slicing and Culturing Pig Hearts under Physiological Conditions. J Vis Exp 2020. [PMID: 32250357 DOI: 10.3791/60913] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Many novel drugs fail in clinical studies due to cardiotoxic side effects as the currently available in vitro assays and in vivo animal models poorly predict human cardiac liabilities, posing a multi-billion-dollar burden on the pharmaceutical industry. Hence, there is a worldwide unmet medical need for better approaches to identify drug cardiotoxicity before undertaking costly and time consuming 'first in man' trials. Currently, only immature cardiac cells (human induced pluripotent stem cell-derived cardiomyocytes [hiPSC-CMs]) are used to test therapeutic efficiency and drug toxicity as they are the only human cardiac cells that can be cultured for prolonged periods required to test drug efficacy and toxicity. However, a single cell type cannot replicate the phenotype of the complex 3D heart tissue which is formed of multiple cell types. Importantly, the effect of drugs needs to be tested on adult cardiomyocytes, which have different characteristics and toxicity responses compared to immature hiPSC-CMs. Culturing human heart slices is a promising model of intact human myocardium. This technology provides access to a complete multicellular system that mimics the human heart tissue and reflects the physiological or pathological conditions of the human myocardium. Recently, through optimization of the culture media components and the culture conditions to include continuous electrical stimulation at 1.2 Hz and intermittent oxygenation of the culture medium, we developed a new culture system setup that preserves viability and functionality of human and pig heart slices for 6 days in culture. In the current protocol, we are detailing the method for slicing and culturing pig heart as an example. The same protocol is used to culture slices from human, dog, sheep, or cat hearts. This culture system has the potential to become a powerful predictive human in situ model for acute cardiotoxicity testing that closes the gap between preclinical and clinical testing results.
Collapse
Affiliation(s)
- Qinghui Ou
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville
| | - Riham R E Abouleisa
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville
| | - Xian-Liang Tang
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville
| | - Hamzah R Juhardeen
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville
| | | | | | | | - Ayman El-Baz
- Department of Bioengineering, University of Louisville
| | - Roberto Bolli
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville
| | - Tamer M A Mohamed
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville; Diabetes and Obesity Center, Department of Medicine, University of Louisville; Department of Pharmacology and Toxicology, University of Louisville; Institute of Cardiovascular Sciences, University of Manchester; Faculty of Pharmacy, Zagazig University;
| |
Collapse
|
46
|
Abstract
RNA modulation has become a promising therapeutic approach for the treatment of several types of disease. The emerging field of noncoding RNA-based therapies has now come to the attention of cardiovascular research, in which it could provide valuable advancements in comparison to current pharmacotherapy such as small molecule drugs or antibodies. In this review, we focus on noncoding RNA-based studies conducted mainly in large-animal models, including pigs, rabbits, dogs, and nonhuman primates. The obstacles and promises of targeting long noncoding RNAs and circRNAs as therapeutic modalities in humans are specifically discussed. We also describe novel ex vivo methods based on human cells and tissues, such as engineered heart tissues and living myocardial slices that could help bridging the gap between in vivo models and clinical applications in the future. Finally, we summarize antisense oligonucleotide drugs that have already been approved by the Food and Drug Administration for targeting mRNAs and discuss the progress of noncoding RNA-based drugs in clinical trials. Additional factors, such as drug chemistry, drug formulations, different routes of administration, and the advantages of RNA-based drugs, are also included in the present review. Recently, first therapeutic miRNA-based inhibitory strategies have been tested in heart failure patients as well as healthy volunteers to study effects on wound healing (NCT04045405; NCT03603431). In summary, a combination of novel therapeutic RNA targets, large-animal models, ex vivo studies with human cells/tissues, and new delivery techniques will likely lead to significant progress in the development of noncoding RNA-based next-generation therapeutics for cardiovascular disease.
Collapse
Affiliation(s)
- Cheng-Kai Huang
- From the Institute of Molecular and Translational Therapeutic Strategies (C.-K.H., S.K.-K., T.T.), Hannover Medical School, Germany
| | - Sabine Kafert-Kasting
- From the Institute of Molecular and Translational Therapeutic Strategies (C.-K.H., S.K.-K., T.T.), Hannover Medical School, Germany
| | - Thomas Thum
- From the Institute of Molecular and Translational Therapeutic Strategies (C.-K.H., S.K.-K., T.T.), Hannover Medical School, Germany
- REBIRTH Center of Translational Regenerative Medicine (T.T.), Hannover Medical School, Germany
| |
Collapse
|
47
|
Pitoulis FG, Terracciano CM. Heart Plasticity in Response to Pressure- and Volume-Overload: A Review of Findings in Compensated and Decompensated Phenotypes. Front Physiol 2020; 11:92. [PMID: 32116796 PMCID: PMC7031419 DOI: 10.3389/fphys.2020.00092] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/27/2020] [Indexed: 12/20/2022] Open
Abstract
The adult human heart has an exceptional ability to alter its phenotype to adapt to changes in environmental demand. This response involves metabolic, mechanical, electrical, and structural alterations, and is known as cardiac plasticity. Understanding the drivers of cardiac plasticity is essential for development of therapeutic agents. This is particularly important in contemporary cardiology, which uses treatments with peripheral effects (e.g., on kidneys, adrenal glands). This review focuses on the effects of different hemodynamic loads on myocardial phenotype. We examine mechanical scenarios of pressure- and volume overload, from the initial insult, to compensated, and ultimately decompensated stage. We discuss how different hemodynamic conditions occur and are underlined by distinct phenotypic and molecular changes. We complete the review by exploring how current basic cardiac research should leverage available cardiac models to study mechanical load in its different presentations.
Collapse
|
48
|
Roelants C, Pillet C, Franquet Q, Sarrazin C, Peilleron N, Giacosa S, Guyon L, Fontanell A, Fiard G, Long JA, Descotes JL, Cochet C, Filhol O. Ex-Vivo Treatment of Tumor Tissue Slices as a Predictive Preclinical Method to Evaluate Targeted Therapies for Patients with Renal Carcinoma. Cancers (Basel) 2020; 12:cancers12010232. [PMID: 31963500 PMCID: PMC7016787 DOI: 10.3390/cancers12010232] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/08/2020] [Accepted: 01/15/2020] [Indexed: 12/14/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the third type of urologic cancer. At time of diagnosis, 30% of cases are metastatic with no effect of chemotherapy or radiotherapy. Current targeted therapies lead to a high rate of relapse and resistance after a short-term response. Thus, a major hurdle in the development and use of new treatments for ccRCC is the lack of good pre-clinical models that can accurately predict the efficacy of new drugs and allow the stratification of patients into the correct treatment regime. Here, we describe different 3D cultures models of ccRCC, emphasizing the feasibility and the advantage of ex-vivo treatment of fresh, surgically resected human tumor slice cultures of ccRCC as a robust preclinical model for identifying patient response to specific therapeutics. Moreover, this model based on precision-cut tissue slices enables histopathology measurements as tumor architecture is retained, including the spatial relationship between the tumor and tumor-infiltrating lymphocytes and the stromal components. Our data suggest that acute treatment of tumor tissue slices could represent a benchmark of further exploration as a companion diagnostic tool in ccRCC treatment and a model to develop new therapeutic drugs.
Collapse
Affiliation(s)
- Caroline Roelants
- Université Grenoble Alpes, Inserm, CEA, IRIG-Biology of Cancer and Infection, UMR_S 1036, F-38000 Grenoble, France; (C.R.); (Q.F.); (C.S.); (N.P.); (S.G.); (L.G.); (C.C.)
- Inovarion, 75005 Paris, France
| | - Catherine Pillet
- Université Grenoble Alpes, Inserm, CEA, IRIG-Biologie à Grande Echelle, UMR 1038, F-38000 Grenoble, France;
| | - Quentin Franquet
- Université Grenoble Alpes, Inserm, CEA, IRIG-Biology of Cancer and Infection, UMR_S 1036, F-38000 Grenoble, France; (C.R.); (Q.F.); (C.S.); (N.P.); (S.G.); (L.G.); (C.C.)
- Centre hospitalier universitaire Grenoble Alpes, CS 10217, 38043 Grenoble CEDEX 9, France; (A.F.); (G.F.); (J.-A.L.); (J.-L.D.)
| | - Clément Sarrazin
- Université Grenoble Alpes, Inserm, CEA, IRIG-Biology of Cancer and Infection, UMR_S 1036, F-38000 Grenoble, France; (C.R.); (Q.F.); (C.S.); (N.P.); (S.G.); (L.G.); (C.C.)
- Centre hospitalier universitaire Grenoble Alpes, CS 10217, 38043 Grenoble CEDEX 9, France; (A.F.); (G.F.); (J.-A.L.); (J.-L.D.)
| | - Nicolas Peilleron
- Université Grenoble Alpes, Inserm, CEA, IRIG-Biology of Cancer and Infection, UMR_S 1036, F-38000 Grenoble, France; (C.R.); (Q.F.); (C.S.); (N.P.); (S.G.); (L.G.); (C.C.)
- Centre hospitalier universitaire Grenoble Alpes, CS 10217, 38043 Grenoble CEDEX 9, France; (A.F.); (G.F.); (J.-A.L.); (J.-L.D.)
| | - Sofia Giacosa
- Université Grenoble Alpes, Inserm, CEA, IRIG-Biology of Cancer and Infection, UMR_S 1036, F-38000 Grenoble, France; (C.R.); (Q.F.); (C.S.); (N.P.); (S.G.); (L.G.); (C.C.)
| | - Laurent Guyon
- Université Grenoble Alpes, Inserm, CEA, IRIG-Biology of Cancer and Infection, UMR_S 1036, F-38000 Grenoble, France; (C.R.); (Q.F.); (C.S.); (N.P.); (S.G.); (L.G.); (C.C.)
| | - Amina Fontanell
- Centre hospitalier universitaire Grenoble Alpes, CS 10217, 38043 Grenoble CEDEX 9, France; (A.F.); (G.F.); (J.-A.L.); (J.-L.D.)
| | - Gaëlle Fiard
- Centre hospitalier universitaire Grenoble Alpes, CS 10217, 38043 Grenoble CEDEX 9, France; (A.F.); (G.F.); (J.-A.L.); (J.-L.D.)
| | - Jean-Alexandre Long
- Centre hospitalier universitaire Grenoble Alpes, CS 10217, 38043 Grenoble CEDEX 9, France; (A.F.); (G.F.); (J.-A.L.); (J.-L.D.)
| | - Jean-Luc Descotes
- Centre hospitalier universitaire Grenoble Alpes, CS 10217, 38043 Grenoble CEDEX 9, France; (A.F.); (G.F.); (J.-A.L.); (J.-L.D.)
| | - Claude Cochet
- Université Grenoble Alpes, Inserm, CEA, IRIG-Biology of Cancer and Infection, UMR_S 1036, F-38000 Grenoble, France; (C.R.); (Q.F.); (C.S.); (N.P.); (S.G.); (L.G.); (C.C.)
| | - Odile Filhol
- Université Grenoble Alpes, Inserm, CEA, IRIG-Biology of Cancer and Infection, UMR_S 1036, F-38000 Grenoble, France; (C.R.); (Q.F.); (C.S.); (N.P.); (S.G.); (L.G.); (C.C.)
- Correspondence: ; Tel.: +33-(0)4-38785645; Fax: +33-(0)4-38785058
| |
Collapse
|
49
|
Abi-Gerges N, Miller PE, Ghetti A. Human Heart Cardiomyocytes in Drug Discovery and Research: New Opportunities in Translational Sciences. Curr Pharm Biotechnol 2019; 21:787-806. [PMID: 31820682 DOI: 10.2174/1389201021666191210142023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/14/2019] [Accepted: 10/28/2019] [Indexed: 12/28/2022]
Abstract
In preclinical drug development, accurate prediction of drug effects on the human heart is critically important, whether in the context of cardiovascular safety or for the purpose of modulating cardiac function to treat heart disease. Current strategies have significant limitations, whereby, cardiotoxic drugs can escape detection or potential life-saving therapies are abandoned due to false positive toxicity signals. Thus, new and more reliable translational approaches are urgently needed to help accelerate the rate of new therapy development. Renewed efforts in the recovery of human donor hearts for research and in cardiomyocyte isolation methods, are providing new opportunities for preclinical studies in adult primary cardiomyocytes. These cells exhibit the native physiological and pharmacological properties, overcoming the limitations presented by artificial cellular models, animal models and have great potential for providing an excellent tool for preclinical drug testing. Adult human primary cardiomyocytes have already shown utility in assessing drug-induced cardiotoxicity risk and helping in the identification of new treatments for cardiac diseases, such as heart failure and atrial fibrillation. Finally, strategies with actionable decision-making trees that rely on data derived from adult human primary cardiomyocytes will provide the holistic insights necessary to accurately predict human heart effects of drugs.
Collapse
Affiliation(s)
- Najah Abi-Gerges
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA 92109, United States
| | - Paul E Miller
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA 92109, United States
| | - Andre Ghetti
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA 92109, United States
| |
Collapse
|
50
|
Kussauer S, David R, Lemcke H. hiPSCs Derived Cardiac Cells for Drug and Toxicity Screening and Disease Modeling: What Micro- Electrode-Array Analyses Can Tell Us. Cells 2019; 8:E1331. [PMID: 31661896 PMCID: PMC6912416 DOI: 10.3390/cells8111331] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 12/19/2022] Open
Abstract
Human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CM) have been intensively used in drug development and disease modeling. Since iPSC-cardiomyocyte (CM) was first generated, their characterization has become a major focus of research. Multi-/micro-electrode array (MEA) systems provide a non-invasive user-friendly platform for detailed electrophysiological analysis of iPSC cardiomyocytes including drug testing to identify potential targets and the assessment of proarrhythmic risk. Here, we provide a systematical overview about the physiological and technical background of micro-electrode array measurements of iPSC-CM. We introduce the similarities and differences between action- and field potential and the advantages and drawbacks of MEA technology. In addition, we present current studies focusing on proarrhythmic side effects of novel and established compounds combining MEA systems and iPSC-CM. MEA technology will help to open a new gateway for novel therapies in cardiovascular diseases while reducing animal experiments at the same time.
Collapse
Affiliation(s)
- Sophie Kussauer
- Department Cardiac Surgery, Medical Center, University of Rostock, 18057 Rostock, Germany.
| | - Robert David
- Department Cardiac Surgery, Medical Center, University of Rostock, 18057 Rostock, Germany.
| | - Heiko Lemcke
- Department Cardiac Surgery, Medical Center, University of Rostock, 18057 Rostock, Germany.
| |
Collapse
|