151
|
Wagner KT, Nash TR, Liu B, Vunjak-Novakovic G, Radisic M. Extracellular Vesicles in Cardiac Regeneration: Potential Applications for Tissues-on-a-Chip. Trends Biotechnol 2021; 39:755-773. [PMID: 32958383 PMCID: PMC7969481 DOI: 10.1016/j.tibtech.2020.08.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/26/2022]
Abstract
Strategies to regenerate cardiac tissue postinjury are limited and heart transplantation remains the only 'cure' for a failing heart. Extracellular vesicles (EVs), membrane-bound cell secretions important in intercellular signaling, have been shown to play a crucial role in regulating heart function. A mechanistic understanding of the role of EVs in the heart remains elusive due to the challenges in studying the native human heart. Tissue-on-a-chip platforms, comprising functional, physiologically relevant human tissue models, are an emerging technology that has yet to be fully applied to the study of EVs. In this review, we summarize recent advances in cardiac tissue-on-a-chip (CTC) platforms and discuss how they are uniquely situated to advance our understanding of EVs in cardiac disease and regeneration.
Collapse
Affiliation(s)
- Karl T Wagner
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Trevor R Nash
- Department of Medicine, Columbia University, New York, NY, USA; Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Bohao Liu
- Department of Medicine, Columbia University, New York, NY, USA; Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Gordana Vunjak-Novakovic
- Department of Medicine, Columbia University, New York, NY, USA; Department of Biomedical Engineering, Columbia University, New York, NY, USA.
| | - Milica Radisic
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Toronto General Research Institute, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
152
|
Epicardially Placed Bioengineered Cardiomyocyte Xenograft in Immune-Competent Rat Model of Heart Failure. Stem Cells Int 2021; 2021:9935679. [PMID: 34341667 PMCID: PMC8325579 DOI: 10.1155/2021/9935679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/09/2021] [Accepted: 06/29/2021] [Indexed: 11/17/2022] Open
Abstract
Background Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are under preclinical investigation as a cell-based therapy for heart failure post-myocardial infarction. In a previous study, tissue-engineered cardiac grafts were found to improve hosts' cardiac electrical and mechanical functions. However, the durability of effect, immune response, and in vitro properties of the tissue graft remained uncharacterized. This present study is aimed at confirming the graft therapeutic efficacy in an immune-competent chronic heart failure (CHF) model and providing evaluation of the in vitro properties of the tissue graft. Methods hiPSC-CMs and human dermal fibroblasts were cultured into a synthetic bioabsorbable scaffold. The engineered grafts underwent epicardial implantation in infarcted immune-competent male Sprague-Dawley rats. Plasma samples were collected throughout the study to quantify antibody titers. At the study endpoint, all cohorts underwent echocardiographic, hemodynamic, electrophysiologic, and histopathologic assessments. Results The epicardially placed tissue graft therapy improved (p < 0.05) in vivo and ex vivo cardiac function compared to the untreated CHF cohort. Total IgM and IgG increased for both the untreated and graft-treated CHF cohorts. An immune response to the grafts was detected after seven days in graft-treated CHF rats only. In vitro, engineered grafts exhibited responsiveness to beta-adrenergic receptor agonism/antagonism and SERCA inhibition and elicited complex molecular profiles. Conclusions This hiPSC-CM-derived cardiac graft improved systolic and diastolic cardiac function in immune-competent CHF rats. The improvements were detectable at seven weeks post-graft implantation despite an antibody response beginning at week one and peaking at week three. This suggests that non-integrating cell-based therapy delivered by a bioengineered tissue graft for ischemic cardiomyopathy is a viable treatment option.
Collapse
|
153
|
Psaras Y, Margara F, Cicconet M, Sparrow AJ, Repetti GG, Schmid M, Steeples V, Wilcox JA, Bueno-Orovio A, Redwood CS, Watkins HC, Robinson P, Rodriguez B, Seidman JG, Seidman CE, Toepfer CN. CalTrack: High-Throughput Automated Calcium Transient Analysis in Cardiomyocytes. Circ Res 2021; 129:326-341. [PMID: 34018815 PMCID: PMC8260473 DOI: 10.1161/circresaha.121.318868] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 05/06/2021] [Accepted: 05/20/2021] [Indexed: 11/21/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Yiangos Psaras
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine (Y.P., F.M., A.J.S., M.S., V.S., C.S.R., H.C.W., P.R., C.N.T.), University of Oxford, United Kingdom
| | - Francesca Margara
- Computer Science (F.M., A.B.-O., B.R.), University of Oxford, United Kingdom
| | - Marcelo Cicconet
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine (Y.P., F.M., A.J.S., M.S., V.S., C.S.R., H.C.W., P.R., C.N.T.), University of Oxford, United Kingdom
- Computer Science (F.M., A.B.-O., B.R.), University of Oxford, United Kingdom
- Wellcome Centre for Human Genetics (H.C.W., C.N.T.), University of Oxford, United Kingdom
- Image and Data Analysis Core (M.C.), Harvard Medical School, Boston, MA
- Genetics (G.G.R., J.A.L.W., J.G.S., C.E.S., C.N.T.), Harvard Medical School, Boston, MA
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA (C.E.S.)
- Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.)
| | - Alexander J. Sparrow
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine (Y.P., F.M., A.J.S., M.S., V.S., C.S.R., H.C.W., P.R., C.N.T.), University of Oxford, United Kingdom
- Computer Science (F.M., A.B.-O., B.R.), University of Oxford, United Kingdom
- Wellcome Centre for Human Genetics (H.C.W., C.N.T.), University of Oxford, United Kingdom
- Image and Data Analysis Core (M.C.), Harvard Medical School, Boston, MA
- Genetics (G.G.R., J.A.L.W., J.G.S., C.E.S., C.N.T.), Harvard Medical School, Boston, MA
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA (C.E.S.)
- Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.)
| | - Giuliana G. Repetti
- Genetics (G.G.R., J.A.L.W., J.G.S., C.E.S., C.N.T.), Harvard Medical School, Boston, MA
| | - Manuel Schmid
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine (Y.P., F.M., A.J.S., M.S., V.S., C.S.R., H.C.W., P.R., C.N.T.), University of Oxford, United Kingdom
- Computer Science (F.M., A.B.-O., B.R.), University of Oxford, United Kingdom
- Wellcome Centre for Human Genetics (H.C.W., C.N.T.), University of Oxford, United Kingdom
- Image and Data Analysis Core (M.C.), Harvard Medical School, Boston, MA
- Genetics (G.G.R., J.A.L.W., J.G.S., C.E.S., C.N.T.), Harvard Medical School, Boston, MA
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA (C.E.S.)
- Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.)
| | - Violetta Steeples
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine (Y.P., F.M., A.J.S., M.S., V.S., C.S.R., H.C.W., P.R., C.N.T.), University of Oxford, United Kingdom
- Computer Science (F.M., A.B.-O., B.R.), University of Oxford, United Kingdom
- Wellcome Centre for Human Genetics (H.C.W., C.N.T.), University of Oxford, United Kingdom
- Image and Data Analysis Core (M.C.), Harvard Medical School, Boston, MA
- Genetics (G.G.R., J.A.L.W., J.G.S., C.E.S., C.N.T.), Harvard Medical School, Boston, MA
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA (C.E.S.)
- Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.)
| | - Jonathan A.L. Wilcox
- Genetics (G.G.R., J.A.L.W., J.G.S., C.E.S., C.N.T.), Harvard Medical School, Boston, MA
| | | | - Charles S. Redwood
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine (Y.P., F.M., A.J.S., M.S., V.S., C.S.R., H.C.W., P.R., C.N.T.), University of Oxford, United Kingdom
- Computer Science (F.M., A.B.-O., B.R.), University of Oxford, United Kingdom
- Wellcome Centre for Human Genetics (H.C.W., C.N.T.), University of Oxford, United Kingdom
- Image and Data Analysis Core (M.C.), Harvard Medical School, Boston, MA
- Genetics (G.G.R., J.A.L.W., J.G.S., C.E.S., C.N.T.), Harvard Medical School, Boston, MA
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA (C.E.S.)
- Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.)
| | - Hugh C. Watkins
- Wellcome Centre for Human Genetics (H.C.W., C.N.T.), University of Oxford, United Kingdom
| | - Paul Robinson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine (Y.P., F.M., A.J.S., M.S., V.S., C.S.R., H.C.W., P.R., C.N.T.), University of Oxford, United Kingdom
| | - Blanca Rodriguez
- Computer Science (F.M., A.B.-O., B.R.), University of Oxford, United Kingdom
| | - Jonathan G. Seidman
- Genetics (G.G.R., J.A.L.W., J.G.S., C.E.S., C.N.T.), Harvard Medical School, Boston, MA
| | - Christine E. Seidman
- Genetics (G.G.R., J.A.L.W., J.G.S., C.E.S., C.N.T.), Harvard Medical School, Boston, MA
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA (C.E.S.)
- Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.)
| | - Christopher N. Toepfer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine (Y.P., F.M., A.J.S., M.S., V.S., C.S.R., H.C.W., P.R., C.N.T.), University of Oxford, United Kingdom
- Wellcome Centre for Human Genetics (H.C.W., C.N.T.), University of Oxford, United Kingdom
- Genetics (G.G.R., J.A.L.W., J.G.S., C.E.S., C.N.T.), Harvard Medical School, Boston, MA
| |
Collapse
|
154
|
Andrysiak K, Stępniewski J, Dulak J. Human-induced pluripotent stem cell-derived cardiomyocytes, 3D cardiac structures, and heart-on-a-chip as tools for drug research. Pflugers Arch 2021; 473:1061-1085. [PMID: 33629131 PMCID: PMC8245367 DOI: 10.1007/s00424-021-02536-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/01/2021] [Accepted: 02/03/2021] [Indexed: 12/13/2022]
Abstract
Development of new drugs is of high interest for the field of cardiac and cardiovascular diseases, which are a dominant cause of death worldwide. Before being allowed to be used and distributed, every new potentially therapeutic compound must be strictly validated during preclinical and clinical trials. The preclinical studies usually involve the in vitro and in vivo evaluation. Due to the increasing reporting of discrepancy in drug effects in animal and humans and the requirement to reduce the number of animals used in research, improvement of in vitro models based on human cells is indispensable. Primary cardiac cells are difficult to access and maintain in cell culture for extensive experiments; therefore, the human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) became an excellent alternative. This technology enables a production of high number of patient- and disease-specific cardiomyocytes and other cardiac cell types for a large-scale research. The drug effects can be extensively evaluated in the context of electrophysiological responses with a use of well-established tools, such as multielectrode array (MEA), patch clamp, or calcium ion oscillation measurements. Cardiotoxicity, which is a common reason for withdrawing drugs from marketing or rejection at final stages of clinical trials, can be easily verified with a use of hiPSC-CM model providing a prediction of human-specific responses and higher safety of clinical trials involving patient cohort. Abovementioned studies can be performed using two-dimensional cell culture providing a high-throughput and relatively lower costs. On the other hand, more complex structures, such as engineered heart tissue, organoids, or spheroids, frequently applied as co-culture systems, represent more physiological conditions and higher maturation rate of hiPSC-derived cells. Furthermore, heart-on-a-chip technology has recently become an increasingly popular tool, as it implements controllable culture conditions, application of various stimulations and continuous parameters read-out. This paper is an overview of possible use of cardiomyocytes and other cardiac cell types derived from hiPSC as in vitro models of heart in drug research area prepared on the basis of latest scientific reports and providing thorough discussion regarding their advantages and limitations.
Collapse
Affiliation(s)
- Kalina Andrysiak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Jacek Stępniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
155
|
Comparison of 10 Control hPSC Lines for Drug Screening in an Engineered Heart Tissue Format. Stem Cell Reports 2021; 15:983-998. [PMID: 33053362 PMCID: PMC7561618 DOI: 10.1016/j.stemcr.2020.09.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 12/19/2022] Open
Abstract
Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) are commercially available, and cardiac differentiation established routine. Systematic evaluation of several control hiPSC-CM is lacking. We investigated 10 different control hiPSC-CM lines and analyzed function and suitability for drug screening. Five commercial and 5 academic hPSC-CM lines were casted in engineered heart tissue (EHT) format. Spontaneous and stimulated EHT contractions were analyzed, and 7 inotropic indicator compounds investigated on 8 cell lines. Baseline contractile force, kinetics, and rate varied widely among the different lines (e.g., relaxation time range: 118-471 ms). In contrast, the qualitative correctness of responses to BayK-8644, nifedipine, EMD-57033, isoprenaline, and digoxin in terms of force and kinetics varied only between 80% and 93%. Large baseline differences between control cell lines support the request for isogenic controls in disease modeling. Variability appears less relevant for drug screening but needs to be considered, arguing for studies with more than one line.
Collapse
|
156
|
Teles D, Kim Y, Ronaldson-Bouchard K, Vunjak-Novakovic G. Machine Learning Techniques to Classify Healthy and Diseased Cardiomyocytes by Contractility Profile. ACS Biomater Sci Eng 2021; 7:3043-3052. [PMID: 34152732 DOI: 10.1021/acsbiomaterials.1c00418] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cardiomyocytes derived from human induced pluripotent stem (iPS) cells enable the study of cardiac physiology and the developmental testing of new therapeutic drugs in a human setting. In parallel, machine learning methods are being applied to biomedical science in unprecedented ways. Machine learning has been used to distinguish healthy from diseased cardiomyocytes using calcium (Ca2+) transient signals. Most Ca2+ transient signals are obtained via terminal assays that do not permit longitudinal studies, although some recently developed options can circumvent these concerns. Here, we describe the use of machine learning to identify healthy and diseased cardiomyocytes according to their contractility profiles, which are derived from brightfield videos. This noncontact, label-free approach allows for the continued cultivation of cells after they have been evaluated for use in other assays and can be readily extended to organs-on-chip. To demonstrate utility, we assessed contractility profiles of cardiomyocytes obtained from patients with Timothy Syndrome (TS), a long QT disease which can lead to fatal arrhythmias, and from healthy individuals. The videos were processed and classified using machine learning methods and their performance was evaluated according to several parameters. The trained algorithms were able to distinguish the TS cardiomyocytes from healthy controls and classify two different healthy controls. The proposed computational machine learning evaluation of human iPS cell-derived cardiomyocytes' contractility profiles has the potential to identify other genetic proarrhythmic events, screen therapeutic agents for inducing or suppressing long QT events, and predict drug-target interactions. The same approach could be readily extended to the evaluation of engineered cardiac tissues within single-tissue and multi-tissue organs-on-chip.
Collapse
Affiliation(s)
- Diogo Teles
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States.,Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, Braga/Guimara̅es, Braga, Portugal
| | - Youngbin Kim
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| | - Kacey Ronaldson-Bouchard
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States.,Department of Medicine, Columbia University, New York, New York 10032, United States
| |
Collapse
|
157
|
Saleem U, van Meer BJ, Katili PA, Mohd Yusof NAN, Mannhardt I, Garcia AK, Tertoolen L, de Korte T, Vlaming MLH, McGlynn K, Nebel J, Bahinski A, Harris K, Rossman E, Xu X, Burton FL, Smith GL, Clements P, Mummery CL, Eschenhagen T, Hansen A, Denning C. Blinded, Multicenter Evaluation of Drug-induced Changes in Contractility Using Human-induced Pluripotent Stem Cell-derived Cardiomyocytes. Toxicol Sci 2021; 176:103-123. [PMID: 32421822 PMCID: PMC7357169 DOI: 10.1093/toxsci/kfaa058] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Animal models are 78% accurate in determining whether drugs will alter contractility of the human heart. To evaluate the suitability of human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) for predictive safety pharmacology, we quantified changes in contractility, voltage, and/or Ca2+ handling in 2D monolayers or 3D engineered heart tissues (EHTs). Protocols were unified via a drug training set, allowing subsequent blinded multicenter evaluation of drugs with known positive, negative, or neutral inotropic effects. Accuracy ranged from 44% to 85% across the platform-cell configurations, indicating the need to refine test conditions. This was achieved by adopting approaches to reduce signal-to-noise ratio, reduce spontaneous beat rate to ≤ 1 Hz or enable chronic testing, improving accuracy to 85% for monolayers and 93% for EHTs. Contraction amplitude was a good predictor of negative inotropes across all the platform-cell configurations and of positive inotropes in the 3D EHTs. Although contraction- and relaxation-time provided confirmatory readouts forpositive inotropes in 3D EHTs, these parameters typically served as the primary source of predictivity in 2D. The reliance of these “secondary” parameters to inotropy in the 2D systems was not automatically intuitive and may be a quirk of hiPSC-CMs, hence require adaptations in interpreting the data from this model system. Of the platform-cell configurations, responses in EHTs aligned most closely to the free therapeutic plasma concentration. This study adds to the notion that hiPSC-CMs could add value to drug safety evaluation.
Collapse
Affiliation(s)
- Umber Saleem
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, 20246 Hamburg, and DZHK (German Center for Cardiovascular Research), Partner Site, Hamburg/Kiel/Lübeck, Germany
| | - Berend J van Meer
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZD, Leiden, The Netherlands
| | - Puspita A Katili
- Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Nurul A N Mohd Yusof
- Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Ingra Mannhardt
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, 20246 Hamburg, and DZHK (German Center for Cardiovascular Research), Partner Site, Hamburg/Kiel/Lübeck, Germany
| | - Ana Krotenberg Garcia
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZD, Leiden, The Netherlands
| | - Leon Tertoolen
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZD, Leiden, The Netherlands
| | - Tessa de Korte
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZD, Leiden, The Netherlands.,Ncardia, 2333 BD, Leiden, The Netherlands
| | | | - Karen McGlynn
- Clyde Biosciences Ltd, Biocity Scotland, Newhouse, Lanarkshire ML1 5HU, UK
| | - Jessica Nebel
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, 20246 Hamburg, and DZHK (German Center for Cardiovascular Research), Partner Site, Hamburg/Kiel/Lübeck, Germany
| | | | | | - Eric Rossman
- GlaxoSmithKline, Collegeville, Pennsylvania 19426
| | - Xiaoping Xu
- GlaxoSmithKline, Collegeville, Pennsylvania 19426
| | - Francis L Burton
- Clyde Biosciences Ltd, Biocity Scotland, Newhouse, Lanarkshire ML1 5HU, UK.,Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Godfrey L Smith
- Clyde Biosciences Ltd, Biocity Scotland, Newhouse, Lanarkshire ML1 5HU, UK.,Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Peter Clements
- GlaxoSmithKline, David Jack Centre for R&D, Ware, Hertfordshire SG12 0DP, UK
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZD, Leiden, The Netherlands.,Department Applied Stem Cell Technologies, University of Twente, 7500 EA Enschede, The Netherlands
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, 20246 Hamburg, and DZHK (German Center for Cardiovascular Research), Partner Site, Hamburg/Kiel/Lübeck, Germany
| | - Arne Hansen
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, 20246 Hamburg, and DZHK (German Center for Cardiovascular Research), Partner Site, Hamburg/Kiel/Lübeck, Germany
| | - Chris Denning
- Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| |
Collapse
|
158
|
Homan T, Delanoë-Ayari H, Meli AC, Cazorla O, Gergely C, Mejat A, Chevalier P, Moreau A. MorphoScript: a dedicated analysis to assess the morphology and contractile structures of cardiomyocytes derived from stem cells. Bioinformatics 2021; 37:4209-4215. [PMID: 34048539 DOI: 10.1093/bioinformatics/btab400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/19/2021] [Accepted: 05/27/2021] [Indexed: 01/07/2023] Open
Abstract
MOTIVATION Cardiomyocytes derived from stem cells are closely followed, notably since the discovery in 2007 of human induced pluripotent stem cells (hiPSC). Cardiomyocytes (hiPSC-CM) derived from hiPSC are indeed more and more used to study specific cardiac diseases as well as for developing novel applications such as drug safety experiments. Robust dedicated tools to characterize hiPSC-CM are now required. The hiPSC-CM morphology constitutes an important parameter since these cells do not demonstrate the expected rod shape, characteristic of native human cardiomyocytes. Similarly, the presence, the density and the organization of contractile structures would be a valuable parameter to study. Precise measurements of such characteristics would be useful in many situations: for describing pathological conditions, for pharmacological screens or even for studies focused on the hiPSC-CM maturation process. RESULTS For this purpose, we developed a MATLAB based image analysis toolbox, which gives accurate values for cellular morphology parameters as well as for the contractile cell organization. IMPLEMENTATION To demonstrate the power of this automated image analysis, we used a commercial maturation medium intended to promote the maturation status of hiPSC-CM, and compare the parameters with the ones obtained with standard culture medium, and with freshly dissociated mouse cardiomyocytes. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Tess Homan
- Claude Bernard University, Lyon 1, Université de Lyon, Lyon; Institut lumière matière, (Lyon, France)
| | - Hélène Delanoë-Ayari
- Claude Bernard University, Lyon 1, Université de Lyon, Lyon; Institut lumière matière, (Lyon, France)
| | - Albano C Meli
- PhyMedExp, INSERM, CNRS, University of Montpellier, Montpellier
| | - Olivier Cazorla
- PhyMedExp, INSERM, CNRS, University of Montpellier, Montpellier
| | - Csilla Gergely
- L2C, University of Montpellier, CNRS, Montpellier, France
| | - Alexandre Mejat
- Claude Bernard University, Lyon 1, Université de Lyon, Lyon; Neuromyogene Institut, (Lyon, France)
| | - Philippe Chevalier
- Claude Bernard University, Lyon 1, Université de Lyon, Lyon; Neuromyogene Institut, (Lyon, France).,Hospices civils de Lyon, Service de Rythmologie, (Bron, France)
| | - Adrien Moreau
- PhyMedExp, INSERM, CNRS, University of Montpellier, Montpellier
| |
Collapse
|
159
|
Esser T, Trossmann V, Lentz S, Engel F, Scheibel T. Designing of spider silk proteins for human induced pluripotent stem cell-based cardiac tissue engineering. Mater Today Bio 2021; 11:100114. [PMID: 34169268 PMCID: PMC8209670 DOI: 10.1016/j.mtbio.2021.100114] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/01/2021] [Accepted: 05/08/2021] [Indexed: 12/25/2022] Open
Abstract
Materials made of recombinant spider silk proteins are promising candidates for cardiac tissue engineering, and their suitability has so far been investigated utilizing primary rat cardiomyocytes. Herein, we expanded the tool box of available spider silk variants and demonstrated for the first time that human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes attach, contract, and respond to pharmacological treatment using phenylephrine and verapamil on explicit spider silk films. The hiPSC-cardiomyocytes contracted for at least 14 days on films made of positively charged engineered Araneus diadematus fibroin 4 (eADF4(κ16)) and three different arginyl-glycyl-aspartic acid (RGD)-tagged spider silk variants (positively or negatively charged and uncharged). Notably, hiPSC-cardiomyocytes exhibited different morphologies depending on the spider silk variant used, with less spreading and being smaller on films made of eADF4(κ16) than on RGD-tagged spider silk films. These results indicate that spider silk engineering is a powerful tool to provide new materials suitable for hiPSC-based cardiac tissue engineering. hiPSC-cardiomyocytes attach and contract on positively charged and/or RGD-tagged spider silk variants. hiPSC-cardiomyocytes exhibit spider silk variant-dependent morphology upon adhesion. Explicit spider silk variants promote long-term contractility of hiPSC-cardiomyocytes. hiPSC-cardiomyocytes grown on spider silk materials respond to pharmacological treatment.
Collapse
Key Words
- AFM, atomic force microscopy
- APTES, (3-aminopropyl) triethoxysilane
- ATR, attenuated total reflection
- DPBS, Dulbecco's phosphate-buffered saline
- EthHD1, ethidium homodimer 1
- FT-IR, Fourier-transform infrared (spectroscopy)
- IPTG, isopropyl-β-D-thiogalactoside
- MALDI-TOF, matrix-assisted laser desorption/ionization time-of-flight
- SDS-PAGE, sodium dodecyl sulfate-polyacrylamide gel electrophoresis
- eADF4, Engineered Araneus diadematus fibroin 4
- hiPSC, human-induced pluripotent stem cell
Collapse
Affiliation(s)
- T.U. Esser
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, 91054, Germany
| | - V.T. Trossmann
- Lehrstuhl Biomaterialien, Prof.-Rüdiger-Bormann Straße 1, Bayreuth, 95447, Germany
| | - S. Lentz
- Lehrstuhl Biomaterialien, Prof.-Rüdiger-Bormann Straße 1, Bayreuth, 95447, Germany
| | - F.B. Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, 91054, Germany
- MURCE, Muscle Research Center Erlangen, Erlangen, Germany
- Corresponding author. Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, 91054, Germany.
| | - T. Scheibel
- Lehrstuhl Biomaterialien, Prof.-Rüdiger-Bormann Straße 1, Bayreuth, 95447, Germany
- Bayreuther Zentrum für Kolloide und Grenzflächen (BZKG), Bayerisches Polymerinstitut (BPI), Bayreuther Zentrum für Molekulare Biowissenschaften (BZMB), Bayreuther Materialzentrum (BayMAT), Universitätsstraße 30, Universität Bayreuth, Bayreuth, D-95447, Germany
- Corresponding author. Lehrstuhl Biomaterialien, Prof.-Rüdiger-Bormann Straße 1, Bayreuth, 95447, Germany.
| |
Collapse
|
160
|
Liu X, Wang S, Guo X, Li Y, Ogurlu R, Lu F, Prondzynski M, Buzon SDLS, Ma Q, Zhang D, Wang G, Cotton J, Guo Y, Xiao L, Milan DJ, Xu Y, Schlame M, Bezzerides VJ, Pu WT. Increased Reactive Oxygen Species-Mediated Ca 2+/Calmodulin-Dependent Protein Kinase II Activation Contributes to Calcium Handling Abnormalities and Impaired Contraction in Barth Syndrome. Circulation 2021; 143:1894-1911. [PMID: 33793303 PMCID: PMC8691127 DOI: 10.1161/circulationaha.120.048698] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Mutations in tafazzin (TAZ), a gene required for biogenesis of cardiolipin, the signature phospholipid of the inner mitochondrial membrane, causes Barth syndrome (BTHS). Cardiomyopathy and risk of sudden cardiac death are prominent features of BTHS, but the mechanisms by which impaired cardiolipin biogenesis causes cardiac muscle weakness and arrhythmia are poorly understood. METHODS We performed in vivo electrophysiology to define arrhythmia vulnerability in cardiac-specific TAZ knockout mice. Using cardiomyocytes derived from human induced pluripotent stem cells and cardiac-specific TAZ knockout mice as model systems, we investigated the effect of TAZ inactivation on Ca2+ handling. Through genome editing and pharmacology, we defined a molecular link between TAZ mutation and abnormal Ca2+ handling and contractility. RESULTS A subset of mice with cardiac-specific TAZ inactivation developed arrhythmias, including bidirectional ventricular tachycardia, atrial tachycardia, and complete atrioventricular block. Compared with wild-type controls, BTHS-induced pluripotent stem cell-derived cardiomyocytes had increased diastolic Ca2+ and decreased Ca2+ transient amplitude. BTHS-induced pluripotent stem cell-derived cardiomyocytes had higher levels of mitochondrial and cellular reactive oxygen species than wild-type controls, which activated CaMKII (Ca2+/calmodulin-dependent protein kinase II). Activated CaMKII phosphorylated the RYR2 (ryanodine receptor 2) on serine 2814, increasing Ca2+ leak through RYR2. Inhibition of this reactive oxygen species-CaMKII-RYR2 pathway through pharmacological inhibitors or genome editing normalized aberrant Ca2+ handling in BTHS-induced pluripotent stem cell-derived cardiomyocytes and improved their contractile function. Murine Taz knockout cardiomyocytes also exhibited elevated diastolic Ca2+ and decreased Ca2+ transient amplitude. These abnormalities were ameliorated by Ca2+/calmodulin-dependent protein kinase II or reactive oxygen species inhibition. CONCLUSIONS This study identified a molecular pathway that links TAZ mutation with abnormal Ca2+ handling and decreased cardiomyocyte contractility. This pathway may offer therapeutic opportunities to treat BTHS and potentially other diseases with elevated mitochondrial reactive oxygen species production.
Collapse
Affiliation(s)
- Xujie Liu
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Radiology, Basic Medical School, Chongqing Medical University, Chongqing, 400016, China
| | - Suya Wang
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Xiaoling Guo
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Center of Scientific Research, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Roza Ogurlu
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Fujian Lu
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | | | | | - Qing Ma
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Donghui Zhang
- State key laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, Hubei 430062, China
| | - Gang Wang
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Justin Cotton
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard College, Cambridge, MA 02138, USA
| | - Yuxuan Guo
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Ling Xiao
- Department of Cardiology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - David J. Milan
- Department of Cardiology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Yang Xu
- Department of Anesthesiology, New York University School of Medicine, New York, New York
| | - Michael Schlame
- Department of Anesthesiology, New York University School of Medicine, New York, New York
| | | | - William T. Pu
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
161
|
Defining optimal enzyme and matrix combination for replating of human induced pluripotent stem cell-derived cardiomyocytes at different levels of maturity. Exp Cell Res 2021; 403:112599. [PMID: 33848551 DOI: 10.1016/j.yexcr.2021.112599] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 03/25/2021] [Accepted: 04/04/2021] [Indexed: 11/24/2022]
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) create an unlimited cell source for basic and translational research. Depending on the maturity of cardiac cultures and the intended applications, obtaining hiPSC-CMs as a single-cell, monolayer or three-dimensional clusters can be challenging. Here, we defined strategies to replate hiPSC-CMs on early days (D15-30) or later more mature (D60-150) differentiation cultures. After generation of hiPSCs and derivation of cardiomyocytes, four dissociation reagents Collagenase A/B, Collagenase II, TrypLE, EDTA and five different extracellular matrix materials Laminin, iMatrix-511, Fibronectin, Matrigel, and Geltrex were comparatively evaluated by imaging, cell viability, and contraction analysis. For early cardiac differentiation cultures mimicking mostly the embryonic stage, the highest adhesion, cell viability, and beating frequencies were achieved by treatment with the TrypLE enzyme. Video-based contraction analysis demonstrated higher beating rates after replating compared to before treatment. For later differentiation days of more mature cardiac cultures, dissociation with EDTA and replating cells on Geltrex or Laminin-derivatives yielded better recovery. Cardiac clusters at various sizes were detected in several groups treated with collagenases. Collectively, our findings revealed the selection criteria of the dissociation approach and coating matrix for replating iPSC-CMs based on the maturity and the requirements of further downstream applications.
Collapse
|
162
|
Campostrini G, Meraviglia V, Giacomelli E, van Helden RW, Yiangou L, Davis RP, Bellin M, Orlova VV, Mummery CL. Generation, functional analysis and applications of isogenic three-dimensional self-aggregating cardiac microtissues from human pluripotent stem cells. Nat Protoc 2021; 16:2213-2256. [PMID: 33772245 PMCID: PMC7611409 DOI: 10.1038/s41596-021-00497-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 01/11/2021] [Indexed: 02/01/2023]
Abstract
Tissue-like structures from human pluripotent stem cells containing multiple cell types are transforming our ability to model and understand human development and disease. Here we describe a protocol to generate cardiomyocytes (CMs), cardiac fibroblasts (CFs) and cardiac endothelial cells (ECs), the three principal cell types in the heart, from human induced pluripotent stem cells (hiPSCs) and combine them in three-dimensional (3D) cardiac microtissues (MTs). We include details of how to differentiate, isolate, cryopreserve and thaw the component cells and how to construct and analyze the MTs. The protocol supports hiPSC-CM maturation and allows replacement of one or more of the three heart cell types in the MTs with isogenic variants bearing disease mutations. Differentiation of each cell type takes ~30 d, while MT formation and maturation requires another 20 d. No specialist equipment is needed and the method is inexpensive, requiring just 5,000 cells per MT.
Collapse
Affiliation(s)
- Giulia Campostrini
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Viviana Meraviglia
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Elisa Giacomelli
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Ruben W.J. van Helden
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Loukia Yiangou
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Richard P. Davis
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands,Department of Biology, University of Padua, 35121 Padua, Italy,Veneto Institute of Molecular Medicine, 35129 Padua, Italy,Correspondence to , or
| | - Valeria V. Orlova
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands,Correspondence to , or
| | - Christine L. Mummery
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands,Department of Applied Stem Cell Technologies, University of Twente, The Netherlands,Correspondence to , or
| |
Collapse
|
163
|
Gacita AM, Fullenkamp DE, Ohiri J, Pottinger T, Puckelwartz MJ, Nobrega MA, McNally EM. Genetic Variation in Enhancers Modifies Cardiomyopathy Gene Expression and Progression. Circulation 2021; 143:1302-1316. [PMID: 33478249 PMCID: PMC8009836 DOI: 10.1161/circulationaha.120.050432] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND Inherited cardiomyopathy associates with a range of phenotypes, mediated by genetic and nongenetic factors. Noninherited cardiomyopathy also displays varying progression and outcomes. Expression of cardiomyopathy genes is under the regulatory control of promoters and enhancers, and human genetic variation in promoters and enhancers may contribute to this variability. METHODS We superimposed epigenomic profiling from hearts and cardiomyocytes, including promoter-capture chromatin conformation information, to identify enhancers for 2 cardiomyopathy genes, MYH7 and LMNA. Enhancer function was validated in human cardiomyocytes derived from induced pluripotent stem cells. We also conducted a genome-wide search to ascertain genomic variation in enhancers positioned to alter cardiac expression and correlated one of these variants to cardiomyopathy progression using biobank data. RESULTS Multiple enhancers were identified and validated for LMNA and MYH7, including a key enhancer that regulates the switch from MYH6 expression to MYH7 expression. Deletion of this enhancer resulted in a dose-dependent increase in MYH6 and faster contractile rate in engineered heart tissues. We searched for genomic variation in enhancer sequences across the genome, with a focus on nucleotide changes that create or interrupt transcription factor binding sites. The sequence variant, rs875908, disrupts a T-Box Transcription Factor 5 binding motif and maps to an enhancer region 2 kilobases from the transcriptional start site of MYH7. Gene editing to remove the enhancer that harbors this variant markedly reduced MYH7 expression in human cardiomyocytes. Using biobank-derived data, rs875908 associated with longitudinal echocardiographic features of cardiomyopathy. CONCLUSIONS Enhancers regulate cardiomyopathy gene expression, and genomic variation within these enhancer regions associates with cardiomyopathic progression over time. This integrated approach identified noncoding modifiers of cardiomyopathy and is applicable to other cardiac genes.
Collapse
Affiliation(s)
- Anthony M. Gacita
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Dominic E. Fullenkamp
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Joyce Ohiri
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Tess Pottinger
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Megan J. Puckelwartz
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | | | - Elizabeth M. McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| |
Collapse
|
164
|
Fang J, Wei X, Li H, Hu N, Liu X, Xu D, Zhang T, Wan H, Wang P, Xie X. Cardiomyocyte electrical-mechanical synchronized model for high-content, dose-quantitative and time-dependent drug assessment. MICROSYSTEMS & NANOENGINEERING 2021; 7:26. [PMID: 34567740 PMCID: PMC8433219 DOI: 10.1038/s41378-021-00247-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 01/13/2021] [Accepted: 01/31/2021] [Indexed: 05/16/2023]
Abstract
Cardiovascular diseases have emerged as a significant threat to human health. However, drug development is a time-consuming and costly process, and few drugs pass the preclinical assessment of safety and efficacy. The existing patch-clamp, Ca2+ imaging, and microelectrode array technologies in cardiomyocyte models for drug preclinical screening have suffered from issues of low throughput, limited long-term assessment, or inability to synchronously and correlatively analyze electrical and mechanical signals. Here, we develop a high-content, dose-quantitative and time-dependent drug assessment platform based on an electrical-mechanical synchronized (EMS) biosensing system. This microfabricated EMS can record both firing potential (FP) and mechanical beating (MB) signals from cardiomyocytes and extract a variety of characteristic parameters from these two signals (FP-MB) for further analysis. This system was applied to test typical ion channel drugs (lidocaine and isradipine), and the dynamic responses of cardiomyocytes to the tested drugs were recorded and analyzed. The high-throughput characteristics of the system can facilitate simultaneous experiments on a large number of samples. Furthermore, a database of various cardiac drugs can be established by heat map analysis for rapid and effective screening of drugs. The EMS biosensing system is highly promising as a powerful tool for the preclinical development of new medicines.
Collapse
Affiliation(s)
- Jiaru Fang
- The First Affiliated Hospital of Sun Yat-Sen University; School of Electronics and Information Technology, Guangdong Province Key Laboratory of Display Material and Technology, Sun Yat-sen University, Guangzhou, 510006 China
| | - Xinwei Wei
- Biosensor National Special Laboratory, Key Laboratory of Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027 China
| | - Hongbo Li
- The First Affiliated Hospital of Sun Yat-Sen University; School of Electronics and Information Technology, Guangdong Province Key Laboratory of Display Material and Technology, Sun Yat-sen University, Guangzhou, 510006 China
| | - Ning Hu
- The First Affiliated Hospital of Sun Yat-Sen University; School of Electronics and Information Technology, Guangdong Province Key Laboratory of Display Material and Technology, Sun Yat-sen University, Guangzhou, 510006 China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050 China
| | - Xingxing Liu
- The First Affiliated Hospital of Sun Yat-Sen University; School of Electronics and Information Technology, Guangdong Province Key Laboratory of Display Material and Technology, Sun Yat-sen University, Guangzhou, 510006 China
| | - Dongxin Xu
- The First Affiliated Hospital of Sun Yat-Sen University; School of Electronics and Information Technology, Guangdong Province Key Laboratory of Display Material and Technology, Sun Yat-sen University, Guangzhou, 510006 China
| | - Tao Zhang
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006 China
| | - Hao Wan
- Biosensor National Special Laboratory, Key Laboratory of Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027 China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050 China
| | - Ping Wang
- Biosensor National Special Laboratory, Key Laboratory of Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027 China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050 China
| | - Xi Xie
- The First Affiliated Hospital of Sun Yat-Sen University; School of Electronics and Information Technology, Guangdong Province Key Laboratory of Display Material and Technology, Sun Yat-sen University, Guangzhou, 510006 China
| |
Collapse
|
165
|
Campostrini G, Windt LM, van Meer BJ, Bellin M, Mummery CL. Cardiac Tissues From Stem Cells: New Routes to Maturation and Cardiac Regeneration. Circ Res 2021; 128:775-801. [PMID: 33734815 PMCID: PMC8410091 DOI: 10.1161/circresaha.121.318183] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The ability of human pluripotent stem cells to form all cells of the body has provided many opportunities to study disease and produce cells that can be used for therapy in regenerative medicine. Even though beating cardiomyocytes were among the first cell types to be differentiated from human pluripotent stem cell, cardiac applications have advanced more slowly than those, for example, for the brain, eye, and pancreas. This is, in part, because simple 2-dimensional human pluripotent stem cell cardiomyocyte cultures appear to need crucial functional cues normally present in the 3-dimensional heart structure. Recent tissue engineering approaches combined with new insights into the dialogue between noncardiomyocytes and cardiomyocytes have addressed and provided solutions to issues such as cardiomyocyte immaturity and inability to recapitulate adult heart values for features like contraction force, electrophysiology, or metabolism. Three-dimensional bioengineered heart tissues are thus poised to contribute significantly to disease modeling, drug discovery, and safety pharmacology, as well as provide new modalities for heart repair. Here, we review the current status of 3-dimensional engineered heart tissues.
Collapse
Affiliation(s)
- Giulia Campostrini
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands (G.C., L.M.W., B.J.v.M., M.B., C.L.M.)
| | - Laura M. Windt
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands (G.C., L.M.W., B.J.v.M., M.B., C.L.M.)
| | - Berend J. van Meer
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands (G.C., L.M.W., B.J.v.M., M.B., C.L.M.)
- MESA+ Institute (B.J.v.M.), University of Twente, Enschede, the Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands (G.C., L.M.W., B.J.v.M., M.B., C.L.M.)
- Department of Biology, University of Padua, Italy (M.B.)
- Veneto Institute of Molecular Medicine, Padua, Padua, Italy (M.B.)
| | - Christine L. Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands (G.C., L.M.W., B.J.v.M., M.B., C.L.M.)
- Department of Applied Stem Cell Technologies (C.L.M.), University of Twente, Enschede, the Netherlands
| |
Collapse
|
166
|
Hnatiuk AP, Briganti F, Staudt DW, Mercola M. Human iPSC modeling of heart disease for drug development. Cell Chem Biol 2021; 28:271-282. [PMID: 33740432 PMCID: PMC8054828 DOI: 10.1016/j.chembiol.2021.02.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/26/2021] [Accepted: 02/19/2021] [Indexed: 02/08/2023]
Abstract
Human induced pluripotent stem cells (hiPSCs) have emerged as a promising platform for pharmacogenomics and drug development. In cardiology, they make it possible to produce unlimited numbers of patient-specific human cells that reproduce hallmark features of heart disease in the culture dish. Their potential applications include the discovery of mechanism-specific therapeutics, the evaluation of safety and efficacy in a human context before a drug candidate reaches patients, and the stratification of patients for clinical trials. Although this new technology has the potential to revolutionize drug discovery, translational hurdles have hindered its widespread adoption for pharmaceutical development. Here we discuss recent progress in overcoming these hurdles that should facilitate the use of hiPSCs to develop new medicines and individualize therapies for heart disease.
Collapse
Affiliation(s)
- Anna P Hnatiuk
- Stanford Cardiovascular Institute, 240 Pasteur Drive, Biomedical Innovation Building, Palo Alto, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Francesca Briganti
- Stanford Cardiovascular Institute, 240 Pasteur Drive, Biomedical Innovation Building, Palo Alto, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - David W Staudt
- Stanford Cardiovascular Institute, 240 Pasteur Drive, Biomedical Innovation Building, Palo Alto, CA 94305, USA; Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Mark Mercola
- Stanford Cardiovascular Institute, 240 Pasteur Drive, Biomedical Innovation Building, Palo Alto, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
167
|
Querdel E, Reinsch M, Castro L, Köse D, Bähr A, Reich S, Geertz B, Ulmer B, Schulze M, Lemoine MD, Krause T, Lemme M, Sani J, Shibamiya A, Stüdemann T, Köhne M, Bibra CV, Hornaschewitz N, Pecha S, Nejahsie Y, Mannhardt I, Christ T, Reichenspurner H, Hansen A, Klymiuk N, Krane M, Kupatt C, Eschenhagen T, Weinberger F. Human Engineered Heart Tissue Patches Remuscularize the Injured Heart in a Dose-Dependent Manner. Circulation 2021; 143:1991-2006. [PMID: 33648345 PMCID: PMC8126500 DOI: 10.1161/circulationaha.120.047904] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Supplemental Digital Content is available in the text. Human engineered heart tissue (EHT) transplantation represents a potential regenerative strategy for patients with heart failure and has been successful in preclinical models. Clinical application requires upscaling, adaptation to good manufacturing practices, and determination of the effective dose.
Collapse
Affiliation(s)
- Eva Querdel
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Marina Reinsch
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Liesa Castro
- Department of Cardiovascular Surgery, University Heart Center (L.C., S.P., H.R.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.).,Now with Department of Cardiology and Intensive Care Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Germany (L.C.)
| | - Deniz Köse
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Andrea Bähr
- I. Medizinische Klinik & Poliklinik, University Clinic Rechts der Isar (A.B., N.H., N.K., C.K.), Technical University Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance Munich (A.B., N.H., N.K., C.K.).,Center for Innovative Medical Models, LMU Munich, Oberschleissheim, Germany (A.B., N.K.)
| | - Svenja Reich
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany
| | - Birgit Geertz
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany
| | - Bärbel Ulmer
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Mirja Schulze
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Marc D Lemoine
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.).,Department of Cardiology-Electrophysiology (M.D.L.), University Heart Center, Hamburg, Germany
| | - Tobias Krause
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Marta Lemme
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Jascha Sani
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Aya Shibamiya
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Tim Stüdemann
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Maria Köhne
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.).,Department of Pediatric Cardiac Surgery (M. Köhne), University Heart Center, Hamburg, Germany
| | - Constantin von Bibra
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Nadja Hornaschewitz
- I. Medizinische Klinik & Poliklinik, University Clinic Rechts der Isar (A.B., N.H., N.K., C.K.), Technical University Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance Munich (A.B., N.H., N.K., C.K.)
| | - Simon Pecha
- Department of Cardiovascular Surgery, University Heart Center (L.C., S.P., H.R.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Yusuf Nejahsie
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany
| | - Ingra Mannhardt
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Torsten Christ
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Hermann Reichenspurner
- Department of Cardiovascular Surgery, University Heart Center (L.C., S.P., H.R.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Arne Hansen
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Nikolai Klymiuk
- I. Medizinische Klinik & Poliklinik, University Clinic Rechts der Isar (A.B., N.H., N.K., C.K.), Technical University Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance Munich (A.B., N.H., N.K., C.K.).,Center for Innovative Medical Models, LMU Munich, Oberschleissheim, Germany (A.B., N.K.)
| | - M Krane
- Department of Cardiovascular Surgery, German Heart Centre Munich (M. Krane), Technical University Munich, Germany.,INSURE (Institute for Translational Cardiac Surgery), Cardiovascular Surgery, Munich, Germany (M. Krane)
| | - C Kupatt
- I. Medizinische Klinik & Poliklinik, University Clinic Rechts der Isar (A.B., N.H., N.K., C.K.), Technical University Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance Munich (A.B., N.H., N.K., C.K.)
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Florian Weinberger
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| |
Collapse
|
168
|
Hortigon-Vinagre MP, Zamora V, Burton FL, Smith GL. The Use of Voltage Sensitive Dye di-4-ANEPPS and Video-Based Contractility Measurements to Assess Drug Effects on Excitation-Contraction Coupling in Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes. J Cardiovasc Pharmacol 2021; 77:280-290. [PMID: 33109927 DOI: 10.1097/fjc.0000000000000937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/09/2020] [Indexed: 12/21/2022]
Abstract
ABSTRACT Because cardiotoxicity is one of the leading causes of drug failure and attrition, the design of new protocols and technologies to assess proarrhythmic risks on cardiac cells is in continuous development by different laboratories. Current methodologies use electrical, intracellular Ca2+, or contractility assays to evaluate cardiotoxicity. Increasingly, the human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are the in vitro tissue model used in commercial assays because it is believed to recapitulate many aspects of human cardiac physiology. In this work, we demonstrate that the combination of a contractility and voltage measurements, using video-based imaging and fluorescence microscopy, on hiPSC-CMs allows the investigation of mechanistic links between electrical and mechanical effects in an assay design that can address medium throughput scales necessary for drug screening, offering a view of the mechanisms underlying drug toxicity. To assess the accuracy of this novel technique, 10 commercially available inotropic drugs were tested (5 positive and 5 negative). Included were drugs with simple and specific mechanisms, such as nifedipine, Bay K8644, and blebbistatin, and others with a more complex action such as isoproterenol, pimobendan, digoxin, and amrinone, among others. In addition, the results provide a mechanism for the toxicity of itraconazole in a human model, a drug with reported side effects on the heart. The data demonstrate a strong negative inotropic effect because of the blockade of L-type Ca2+ channels and additional action on the cardiac myofilaments. We can conclude that the combination of contractility and action potential measurements can provide wider mechanistic knowledge of drug cardiotoxicity for preclinical assays.
Collapse
MESH Headings
- Action Potentials/drug effects
- Arrhythmias, Cardiac/chemically induced
- Arrhythmias, Cardiac/metabolism
- Arrhythmias, Cardiac/physiopathology
- Calcium Channels, L-Type/drug effects
- Calcium Channels, L-Type/metabolism
- Cardiotoxicity
- Cell Differentiation
- Cells, Cultured
- Excitation Contraction Coupling/drug effects
- Fluorescent Dyes/chemistry
- Humans
- Induced Pluripotent Stem Cells/drug effects
- Induced Pluripotent Stem Cells/metabolism
- Induced Pluripotent Stem Cells/pathology
- Microscopy, Fluorescence
- Microscopy, Video
- Myocardial Contraction/drug effects
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Myofibrils/drug effects
- Myofibrils/metabolism
- Myofibrils/pathology
- Pyridinium Compounds/chemistry
- Risk Assessment
- Time Factors
- Toxicity Tests
Collapse
Affiliation(s)
- Maria Pura Hortigon-Vinagre
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universdad de Extremadura, Badajoz, Spain
| | - Victor Zamora
- Departamento de Ingeniería Mecánica, Energética y de los Materiales, Escuela de Ingerierias Industriales, Universidad de Extremadura, Badajoz, Spain
| | - Francis L Burton
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Science, University of Glasgow, Glasgow, United Kingdom ; and
- Clyde Biosciences Ltd, BioCity Scotland, Newhouse, United Kingdom
| | - Godfrey L Smith
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Science, University of Glasgow, Glasgow, United Kingdom ; and
- Clyde Biosciences Ltd, BioCity Scotland, Newhouse, United Kingdom
| |
Collapse
|
169
|
Tijsen AJ, Cócera Ortega L, Reckman YJ, Zhang X, van der Made I, Aufiero S, Li J, Kamps SC, van den Bout A, Devalla HD, van Spaendonck-Zwarts KY, Engelhardt S, Gepstein L, Ware JS, Pinto YM. Titin Circular RNAs Create a Back-Splice Motif Essential for SRSF10 Splicing. Circulation 2021; 143:1502-1512. [PMID: 33583186 PMCID: PMC8032209 DOI: 10.1161/circulationaha.120.050455] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Supplemental Digital Content is available in the text. Background: TTN (Titin), the largest protein in humans, forms the molecular spring that spans half of the sarcomere to provide passive elasticity to the cardiomyocyte. Mutations that disrupt the TTN transcript are the most frequent cause of hereditary heart failure. We showed before that TTN produces a class of circular RNAs (circRNAs) that depend on RBM20 to be formed. In this study, we show that the back-splice junction formed by this class of circRNAs creates a unique motif that binds SRSF10 to enable it to regulate splicing. Furthermore, we show that one of these circRNAs (cTTN1) distorts both localization of and splicing by RBM20. Methods: We calculated genetic constraint of the identified motif in 125 748 exomes collected from the gnomAD database. Furthermore, we focused on the highest expressed RBM20-dependent circRNA in the human heart, which we named cTTN1. We used shRNAs directed to the back-splice junction to induce selective loss of cTTN1 in human induced pluripotent stem cell–derived cardiomyocytes. Results: Human genetics suggests reduced genetic tolerance of the generated motif, indicating that mutations in this motif might lead to disease. RNA immunoprecipitation confirmed binding of circRNAs with this motif to SRSF10. Selective loss of cTTN1 in human induced pluripotent stem cell–derived cardiomyocytes induced structural abnormalities, apoptosis, and reduced contractile force in engineered heart tissue. In line with its SRSF10 binding, loss of cTTN1 caused abnormal splicing of important cardiomyocyte SRSF10 targets such as MEF2A and CASQ2. Strikingly, loss of cTTN1 also caused abnormal splicing of TTN itself. Mechanistically, we show that loss of cTTN1 distorts both localization of and splicing by RBM20. Conclusions: We demonstrate that circRNAs formed from the TTN transcript are essential for normal splicing of key muscle genes by enabling splice regulators RBM20 and SRSF10. This shows that the TTN transcript also has regulatory roles, besides its well-known signaling and structural function. In addition, we demonstrate that the specific sequence created by the back-splice junction of these circRNAs has important functions. This highlights the existence of functionally important sequences that cannot be recognized as such in the human genome but provides an as-yet unrecognized source for functional sequence variation.
Collapse
Affiliation(s)
- Anke J Tijsen
- Amsterdam UMC, University of Amsterdam, Departments of Experimental Cardiology, Amsterdam Cardiovascular Sciences (A.J.T., L.C.O., Y.J.R., I.v.d.M., S.A., S.C.K., A.v.d.B., Y.M.P.), Amsterdam, The Netherlands
| | - Lucía Cócera Ortega
- Amsterdam UMC, University of Amsterdam, Departments of Experimental Cardiology, Amsterdam Cardiovascular Sciences (A.J.T., L.C.O., Y.J.R., I.v.d.M., S.A., S.C.K., A.v.d.B., Y.M.P.), Amsterdam, The Netherlands
| | - Yolan J Reckman
- Amsterdam UMC, University of Amsterdam, Departments of Experimental Cardiology, Amsterdam Cardiovascular Sciences (A.J.T., L.C.O., Y.J.R., I.v.d.M., S.A., S.C.K., A.v.d.B., Y.M.P.), Amsterdam, The Netherlands
| | - Xiaolei Zhang
- Imperial College London, South Kensington Campus, London, UK (X.Z., J.S.W.)
| | - Ingeborg van der Made
- Amsterdam UMC, University of Amsterdam, Departments of Experimental Cardiology, Amsterdam Cardiovascular Sciences (A.J.T., L.C.O., Y.J.R., I.v.d.M., S.A., S.C.K., A.v.d.B., Y.M.P.), Amsterdam, The Netherlands
| | - Simona Aufiero
- Amsterdam UMC, University of Amsterdam, Departments of Experimental Cardiology, Amsterdam Cardiovascular Sciences (A.J.T., L.C.O., Y.J.R., I.v.d.M., S.A., S.C.K., A.v.d.B., Y.M.P.), Amsterdam, The Netherlands
| | - Jiuru Li
- Medical Biology, Amsterdam Cardiovascular Sciences (J.L., H.D.D.), Amsterdam, The Netherlands
| | - Selina C Kamps
- Amsterdam UMC, University of Amsterdam, Departments of Experimental Cardiology, Amsterdam Cardiovascular Sciences (A.J.T., L.C.O., Y.J.R., I.v.d.M., S.A., S.C.K., A.v.d.B., Y.M.P.), Amsterdam, The Netherlands
| | - Anouk van den Bout
- Amsterdam UMC, University of Amsterdam, Departments of Experimental Cardiology, Amsterdam Cardiovascular Sciences (A.J.T., L.C.O., Y.J.R., I.v.d.M., S.A., S.C.K., A.v.d.B., Y.M.P.), Amsterdam, The Netherlands
| | - Harsha D Devalla
- Medical Biology, Amsterdam Cardiovascular Sciences (J.L., H.D.D.), Amsterdam, The Netherlands
| | | | - Stefan Engelhardt
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (S.E.).,Institut für Pharmakologie und Toxikologie, Technische Universität München, Munich, Germany (S.E.)
| | - Lior Gepstein
- The Sohnis Family Laboratory for Cardiac Electrophysiology and Regenerative Medicine, Rappaport Faculty of Medicine and Research Institute, Technion-Institute of Technology, Haifa, Israel (L.G.)
| | - James S Ware
- Imperial College London, South Kensington Campus, London, UK (X.Z., J.S.W.)
| | - Yigal M Pinto
- Amsterdam UMC, University of Amsterdam, Departments of Experimental Cardiology, Amsterdam Cardiovascular Sciences (A.J.T., L.C.O., Y.J.R., I.v.d.M., S.A., S.C.K., A.v.d.B., Y.M.P.), Amsterdam, The Netherlands
| |
Collapse
|
170
|
Eroglu TE, Mohr GH, Blom MT, Verkerk AO, Souverein PC, Torp-Pedersen C, Folke F, Wissenberg M, van den Brink L, Davis RP, de Boer A, Gislason GH, Tan HL. Differential effects on out-of-hospital cardiac arrest of dihydropyridines: real-world data from population-based cohorts across two European countries. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2021; 6:347-355. [PMID: 31504369 PMCID: PMC8061029 DOI: 10.1093/ehjcvp/pvz038] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/20/2019] [Accepted: 08/08/2019] [Indexed: 12/20/2022]
Abstract
AIMS Various drugs increase the risk of out-of-hospital cardiac arrest (OHCA) in the general population by impacting cardiac ion channels, thereby causing ventricular tachycardia/fibrillation (VT/VF). Dihydropyridines block L-type calcium channels, but their association with OHCA risk is unknown. We aimed to study whether nifedipine and/or amlodipine, often-used dihydropyridines, are associated with increased OHCA risk, and how these drugs impact on cardiac electrophysiology. METHODS AND RESULTS We conducted a case-control study with VT/VF-documented OHCA cases with presumed cardiac cause from ongoing population-based OHCA registries in the Netherlands and Denmark, and age/sex/index date-matched non-OHCA controls (Netherlands: PHARMO Database Network, Denmark: Danish Civil Registration System). We included 2503 OHCA cases, 10 543 non-OHCA controls in Netherlands, and 8101 OHCA cases, 40 505 non-OHCA controls in Denmark. To examine drug effects on cardiac electrophysiology, we performed single-cell patch-clamp studies in human-induced pluripotent stem cell-derived cardiomyocytes. Use of high-dose nifedipine (≥60 mg/day), but not low-dose nifedipine (<60 mg/day) or amlodipine (any-dose), was associated with higher OHCA risk than non-use of dihydropyridines [Netherlands: adjusted odds ratios (ORadj) 1.45 (95% confidence interval 1.02-2.07), Denmark: 1.96 (1.18-3.25)] or use of amlodipine [Netherlands: 2.31 (1.54-3.47), Denmark: 2.20 (1.32-3.67)]. Out-of-hospital cardiac arrest risk of (high-dose) nifedipine use was not further increased in patients using nitrates, or with a history of ischaemic heart disease. Nifedipine and amlodipine blocked L-type calcium channels at similar concentrations, but, at clinically used concentrations, nifedipine caused more L-type calcium current block, resulting in more action potential shortening. CONCLUSION High-dose nifedipine, but not low-dose nifedipine or any-dose amlodipine, is associated with increased OHCA risk in the general population. Careful titration of nifedipine dose should be considered.
Collapse
Affiliation(s)
- Talip E Eroglu
- Department of Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Grimur H Mohr
- Department of Cardiology, Copenhagen University Hospital Gentofte, Kildegårdsvej 28, 2900 Hellerup, Denmark
| | - Marieke T Blom
- Department of Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Arie O Verkerk
- Department of Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.,Department of Medical Biology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Patrick C Souverein
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, PO Box 80082, Utrecht 3508 TB, The Netherlands
| | - Christian Torp-Pedersen
- Department of Cardiology, Copenhagen University Hospital Gentofte, Kildegårdsvej 28, 2900 Hellerup, Denmark.,Unit of Epidemiology and Biostatistics, Aalborg University Hospital, Sdr. Skovvej 15, 9000 Aalborg, Denmark.,Department of Cardiology, Aalborg University Hospital, Sdr. Skovvej 15, 9000 Aalborg, Denmark.,Department of Health Science and Technology, Aalborg University Hospital, Sdr. Skovvej 15, 9000 Aalborg, Denmark
| | - Fredrik Folke
- Department of Cardiology, Copenhagen University Hospital Gentofte, Kildegårdsvej 28, 2900 Hellerup, Denmark
| | - Mads Wissenberg
- Department of Cardiology, Copenhagen University Hospital Gentofte, Kildegårdsvej 28, 2900 Hellerup, Denmark
| | - Lettine van den Brink
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2300 RC Leiden, The Netherlands
| | - Richard P Davis
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2300 RC Leiden, The Netherlands
| | - Anthonius de Boer
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, PO Box 80082, Utrecht 3508 TB, The Netherlands
| | - Gunnar H Gislason
- Department of Cardiology, Copenhagen University Hospital Gentofte, Kildegårdsvej 28, 2900 Hellerup, Denmark.,National Institute of Public Health, University of Southern Denmark, Studiestræde 6, 1455 Copenhagen, Denmark.,The Danish Heart Foundation, Vognmagergade 7, 1120 Copenhagen, Denmark
| | - Hanno L Tan
- Department of Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
171
|
3D bioprinting of high cell-density heterogeneous tissue models through spheroid fusion within self-healing hydrogels. Nat Commun 2021; 12:753. [PMID: 33531489 PMCID: PMC7854667 DOI: 10.1038/s41467-021-21029-2] [Citation(s) in RCA: 265] [Impact Index Per Article: 66.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 01/07/2021] [Indexed: 02/06/2023] Open
Abstract
Cellular models are needed to study human development and disease in vitro, and to screen drugs for toxicity and efficacy. Current approaches are limited in the engineering of functional tissue models with requisite cell densities and heterogeneity to appropriately model cell and tissue behaviors. Here, we develop a bioprinting approach to transfer spheroids into self-healing support hydrogels at high resolution, which enables their patterning and fusion into high-cell density microtissues of prescribed spatial organization. As an example application, we bioprint induced pluripotent stem cell-derived cardiac microtissue models with spatially controlled cardiomyocyte and fibroblast cell ratios to replicate the structural and functional features of scarred cardiac tissue that arise following myocardial infarction, including reduced contractility and irregular electrical activity. The bioprinted in vitro model is combined with functional readouts to probe how various pro-regenerative microRNA treatment regimes influence tissue regeneration and recovery of function as a result of cardiomyocyte proliferation. This method is useful for a range of biomedical applications, including the development of precision models to mimic diseases and the screening of drugs, particularly where high cell densities and heterogeneity are important.
Collapse
|
172
|
Hang C, Song Y, Li Y, Zhang S, Chang Y, Bai R, Saleem A, Jiang M, Lu W, Lan F, Cui M. Knockout of MYOM1 in human cardiomyocytes leads to myocardial atrophy via impairing calcium homeostasis. J Cell Mol Med 2021; 25:1661-1676. [PMID: 33452765 PMCID: PMC7875908 DOI: 10.1111/jcmm.16268] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/14/2020] [Accepted: 12/22/2020] [Indexed: 12/27/2022] Open
Abstract
Myomesin-1 (encoded by MYOM1 gene) is expressed in almost all cross-striated muscles, whose family (together with myomesin-2 and myomesin-3) helps to cross-link adjacent myosin to form the M-line in myofibrils. However, little is known about its biological function, causal relationship and mechanisms underlying the MYOM1-related myopathies (especially in the heart). Regrettably, there is no MYMO1 knockout model for its study so far. A better and further understanding of MYOM1 biology is urgently needed. Here, we used CRISPR/Cas9 gene-editing technology to establish an MYOM1 knockout human embryonic stem cell line (MYOM1-/- hESC), which was then differentiated into myomesin-1 deficient cardiomyocytes (MYOM1-/- hESC-CMs) in vitro. We found that myomesin-1 plays an important role in sarcomere assembly, contractility regulation and cardiomyocytes development. Moreover, myomesin-1-deficient hESC-CMs can recapitulate myocardial atrophy phenotype in vitro. Based on this model, not only the biological function of MYOM1, but also the aetiology, pathogenesis, and potential treatments of myocardial atrophy caused by myomesin-1 deficiency can be studied.
Collapse
Affiliation(s)
- Chengwen Hang
- Department of CardiologyPeking University Third HospitalBeijingChina
| | - Yuanxiu Song
- Department of CardiologyPeking University Third HospitalBeijingChina
| | - Ya’nan Li
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Siyao Zhang
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Yun Chang
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Rui Bai
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Amina Saleem
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Mengqi Jiang
- Department of CardiologyPeking University Third HospitalBeijingChina
| | - Wenjing Lu
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Feng Lan
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Ming Cui
- Department of CardiologyPeking University Third HospitalBeijingChina
| |
Collapse
|
173
|
Lamotte JDD, Roqueviere S, Gautier H, Raban E, Bouré C, Fonfria E, Krupp J, Nicoleau C. hiPSC-Derived Neurons Provide a Robust and Physiologically Relevant In Vitro Platform to Test Botulinum Neurotoxins. Front Pharmacol 2021; 11:617867. [PMID: 33519485 PMCID: PMC7840483 DOI: 10.3389/fphar.2020.617867] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022] Open
Abstract
Botulinum neurotoxins (BoNTs) are zinc metalloproteases that block neurotransmitter release at the neuromuscular junction (NMJ). Their high affinity for motor neurons combined with a high potency have made them extremely effective drugs for the treatment of a variety of neurological diseases as well as for aesthetic applications. Current in vitro assays used for testing and developing BoNT therapeutics include primary rodent cells and immortalized cell lines. Both models have limitations concerning accuracy and physiological relevance. In order to improve the translational value of preclinical data there is a clear need to use more accurate models such as human induced Pluripotent Stem Cells (hiPSC)-derived neuronal models. In this study we have assessed the potential of four different human iPSC-derived neuronal models including Motor Neurons for BoNT testing. We have characterized these models in detail and found that all models express all proteins needed for BoNT intoxication and showed that all four hiPSC-derived neuronal models are sensitive to both serotype A and E BoNT with Motor Neurons being the most sensitive. We showed that hiPSC-derived Motor Neurons expressed authentic markers after only 7 days of culture, are functional and able to form active synapses. When cultivated with myotubes, we demonstrated that they can innervate myotubes and induce contraction, generating an in vitro model of NMJ showing dose-responsive sensitivity BoNT intoxication. Together, these data demonstrate the promise of hiPSC-derived neurons, especially Motor Neurons, for pharmaceutical BoNT testing and development.
Collapse
|
174
|
Wang H, Li H, Wei X, Zhang T, Xiang Y, Fang J, Wu P, Xie X, Wang P, Hu N. Recognition of high-specificity hERG K+ channel inhibitor-induced arrhythmia in cardiomyocytes by automated template matching. MICROSYSTEMS & NANOENGINEERING 2021; 7:24. [PMID: 34567738 PMCID: PMC8433465 DOI: 10.1038/s41378-021-00251-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 01/15/2021] [Accepted: 02/04/2021] [Indexed: 05/11/2023]
Abstract
Cardiovascular disease (CVD) is the number one cause of death in humans. Arrhythmia induced by gene mutations, heart disease, or hERG K+ channel inhibitors is a serious CVD that can lead to sudden death or heart failure. Conventional cardiomyocyte-based biosensors can record extracellular potentials and mechanical beating signals. However, parameter extraction and examination by the naked eye are the traditional methods for analyzing arrhythmic beats, and it is difficult to achieve automated and efficient arrhythmic recognition with these methods. In this work, we developed a unique automated template matching (ATM) cardiomyocyte beating model to achieve arrhythmic recognition at the single beat level with an interdigitated electrode impedance detection system. The ATM model was established based on a rhythmic template with a data length that was dynamically adjusted to match the data length of the target beat by spline interpolation. The performance of the ATM model under long-term astemizole, droperidol, and sertindole treatment at different doses was determined. The results indicated that the ATM model based on a random rhythmic template of a signal segment obtained after astemizole treatment presented a higher recognition accuracy (100% for astemizole treatment and 99.14% for droperidol and sertindole treatment) than the ATM model based on arrhythmic multitemplates. We believe this highly specific ATM method based on a cardiomyocyte beating model has the potential to be used for arrhythmia screening in the fields of cardiology and pharmacology.
Collapse
Affiliation(s)
- Hao Wang
- The First Affiliated Hospital of Sun Yat-sen University, School of Electronics and Information Technology, State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, Sun Yat-sen University, Guangzhou, 510006 China
| | - Hongbo Li
- The First Affiliated Hospital of Sun Yat-sen University, School of Electronics and Information Technology, State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, Sun Yat-sen University, Guangzhou, 510006 China
| | - Xinwei Wei
- Biosensor National Special Laboratory, Key Laboratory of Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027 China
| | - Tao Zhang
- The First Affiliated Hospital of Sun Yat-sen University, School of Electronics and Information Technology, State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, Sun Yat-sen University, Guangzhou, 510006 China
| | - Yuting Xiang
- The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510655 China
| | - Jiaru Fang
- The First Affiliated Hospital of Sun Yat-sen University, School of Electronics and Information Technology, State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, Sun Yat-sen University, Guangzhou, 510006 China
| | - Peiran Wu
- The First Affiliated Hospital of Sun Yat-sen University, School of Electronics and Information Technology, State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, Sun Yat-sen University, Guangzhou, 510006 China
| | - Xi Xie
- The First Affiliated Hospital of Sun Yat-sen University, School of Electronics and Information Technology, State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, Sun Yat-sen University, Guangzhou, 510006 China
| | - Ping Wang
- Biosensor National Special Laboratory, Key Laboratory of Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027 China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050 China
| | - Ning Hu
- The First Affiliated Hospital of Sun Yat-sen University, School of Electronics and Information Technology, State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, Sun Yat-sen University, Guangzhou, 510006 China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050 China
| |
Collapse
|
175
|
Dal Lin C, Radu CM, Vitiello G, Romano P, Polcari A, Iliceto S, Simioni P, Tona F. Sounds Stimulation on In Vitro HL1 Cells: A Pilot Study and a Theoretical Physical Model. Int J Mol Sci 2020; 22:ijms22010156. [PMID: 33375749 PMCID: PMC7796405 DOI: 10.3390/ijms22010156] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/19/2020] [Accepted: 12/20/2020] [Indexed: 02/07/2023] Open
Abstract
Mechanical vibrations seem to affect the behaviour of different cell types and the functions of different organs. Pressure waves, including acoustic waves (sounds), could affect cytoskeletal molecules via coherent changes in their spatial organization and mechano-transduction signalling. We analyzed the sounds spectra and their fractal features. Cardiac muscle HL1 cells were exposed to different sounds, were stained for cytoskeletal markers (phalloidin, beta-actin, alpha-tubulin, alpha-actinin-1), and studied with multifractal analysis (using FracLac for ImageJ). A single cell was live-imaged and its dynamic contractility changes in response to each different sound were analysed (using Musclemotion for ImageJ). Different sound stimuli seem to influence the contractility and the spatial organization of HL1 cells, resulting in a different localization and fluorescence emission of cytoskeletal proteins. Since the cellular behaviour seems to correlate with the fractal structure of the sound used, we speculate that it can influence the cells by virtue of the different sound waves’ geometric properties that we have photographed and filmed. A theoretical physical model is proposed to explain our results, based on the coherent molecular dynamics. We stress the role of the systemic view in the understanding of the biological activity.
Collapse
Affiliation(s)
- Carlo Dal Lin
- Department of Cardiac, Thoracic and Vascular Sciences, Padua University Medical School, 35100 Padua, Italy; (S.I.); (F.T.)
- Correspondence: ; Tel.: +39-049-8218642; Fax: +39-049-8211802
| | - Claudia Maria Radu
- Department of Women’s and Children’s Health, University of Padua, 35100 Padua, Italy;
- Department of Medicine, Thrombotic and Haemorrhagic Diseases Unit, Veneto Region Haemophilia and Thrombophilia Centre, University of Padua Medical School, 35100 Padua, Italy;
| | - Giuseppe Vitiello
- Department of Physics “E.R. Caianiello”, Salerno University, Fisciano, 84084 Salerno, Italy;
| | - Paola Romano
- Department of Sciences and Technologies, Sannio University, 82100 Benevento, Italy;
- CNR-SPIN Salerno, Baronissi, 84084 Salerno, Italy
| | | | - Sabino Iliceto
- Department of Cardiac, Thoracic and Vascular Sciences, Padua University Medical School, 35100 Padua, Italy; (S.I.); (F.T.)
| | - Paolo Simioni
- Department of Medicine, Thrombotic and Haemorrhagic Diseases Unit, Veneto Region Haemophilia and Thrombophilia Centre, University of Padua Medical School, 35100 Padua, Italy;
| | - Francesco Tona
- Department of Cardiac, Thoracic and Vascular Sciences, Padua University Medical School, 35100 Padua, Italy; (S.I.); (F.T.)
| |
Collapse
|
176
|
Abstract
Inherited cardiac arrhythmias contribute substantially to sudden cardiac death in the young. The underlying pathophysiology remains incompletely understood because of the lack of representative study models and the labour-intensive nature of electrophysiological patch clamp experiments. Whereas patch clamp is still considered the gold standard for investigating electrical properties in a cell, optical mapping of voltage and calcium transients has paved the way for high-throughput studies. Moreover, the development of human-induced pluripotent stem-cell-derived cardiomyocytes (hiPSC-CMs) has enabled the study of patient specific cell lines capturing the full genomic background. Nevertheless, hiPSC-CMs do not fully address the complex interactions between various cell types in the heart. Studies using in vivo models, are therefore necessary. Given the analogies between the human and zebrafish cardiovascular system, zebrafish has emerged as a cost-efficient model for arrhythmogenic diseases. In this review, we describe how hiPSC-CM and zebrafish are employed as models to study primary electrical disorders. We provide an overview of the contemporary electrophysiological phenotyping tools and discuss in more depth the different strategies available for optical mapping. We consider the current advantages and disadvantages of both hiPSC-CM and zebrafish as a model and optical mapping as phenotyping tool and propose strategies for further improvement. Overall, the combination of experimental readouts at cellular (hiPSC-CM) and whole organ (zebrafish) level can raise our understanding of the complexity of inherited cardiac arrhythmia disorders to the next level.
Collapse
|
177
|
Andersen J, Revah O, Miura Y, Thom N, Amin ND, Kelley KW, Singh M, Chen X, Thete MV, Walczak EM, Vogel H, Fan HC, Paşca SP. Generation of Functional Human 3D Cortico-Motor Assembloids. Cell 2020; 183:1913-1929.e26. [PMID: 33333020 DOI: 10.1016/j.cell.2020.11.017] [Citation(s) in RCA: 312] [Impact Index Per Article: 62.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 05/27/2020] [Accepted: 11/04/2020] [Indexed: 12/21/2022]
Abstract
Neurons in the cerebral cortex connect through descending pathways to hindbrain and spinal cord to activate muscle and generate movement. Although components of this pathway have been previously generated and studied in vitro, the assembly of this multi-synaptic circuit has not yet been achieved with human cells. Here, we derive organoids resembling the cerebral cortex or the hindbrain/spinal cord and assemble them with human skeletal muscle spheroids to generate 3D cortico-motor assembloids. Using rabies tracing, calcium imaging, and patch-clamp recordings, we show that corticofugal neurons project and connect with spinal spheroids, while spinal-derived motor neurons connect with muscle. Glutamate uncaging or optogenetic stimulation of cortical spheroids triggers robust contraction of 3D muscle, and assembloids are morphologically and functionally intact for up to 10 weeks post-fusion. Together, this system highlights the remarkable self-assembly capacity of 3D cultures to form functional circuits that could be used to understand development and disease.
Collapse
Affiliation(s)
- Jimena Andersen
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Omer Revah
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Yuki Miura
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Nicholas Thom
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Neal D Amin
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Kevin W Kelley
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Mandeep Singh
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Xiaoyu Chen
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Mayuri Vijay Thete
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | | | - Hannes Vogel
- Departments of Pathology and Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - H Christina Fan
- BD Biosciences, 4040 Campbell Ave Suite 110, Menlo Park, CA 94025, USA
| | - Sergiu P Paşca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
178
|
Wei X, Zhuang L, Li H, He C, Wan H, Hu N, Wang P. Advances in Multidimensional Cardiac Biosensing Technologies: From Electrophysiology to Mechanical Motion and Contractile Force. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2005828. [PMID: 33230867 DOI: 10.1002/smll.202005828] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Indexed: 06/11/2023]
Abstract
Cardiovascular disease is currently a leading killer to human, while drug-induced cardiotoxicity remains the main cause of the withdrawal and attrition of drugs. Taking clinical correlation and throughput into account, cardiomyocyte is perfect as in vitro cardiac model for heart disease modeling, drug discovery, and cardiotoxicity assessment by accurately measuring the physiological multiparameters of cardiomyocytes. Remarkably, cardiomyocytes present both electrophysiological and biomechanical characteristics due to the unique excitation-contraction coupling, which plays a significant role in studying the cardiomyocytes. This review mainly focuses on the recent advances of biosensing technologies for the 2D and 3D cardiac models with three special properties: electrophysiology, mechanical motion, and contractile force. These high-performance multidimensional cardiac models are popular and effective to rebuild and mimic the heart in vitro. To help understand the high-quality and accurate physiologies, related detection techniques are highly demanded, from microtechnology to nanotechnology, from extracellular to intracellular recording, from multiple cells to single cell, and from planar to 3D models. Furthermore, the characteristics, advantages, limitations, and applications of these cardiac biosensing technologies, as well as the future development prospects should contribute to the systematization and expansion of knowledge.
Collapse
Affiliation(s)
- Xinwei Wei
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Liujing Zhuang
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Hongbo Li
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou, 510006, China
| | - Chuanjiang He
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
| | - Hao Wan
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Ning Hu
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou, 510006, China
| | - Ping Wang
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| |
Collapse
|
179
|
Li J, Hua Y, Miyagawa S, Zhang J, Li L, Liu L, Sawa Y. hiPSC-Derived Cardiac Tissue for Disease Modeling and Drug Discovery. Int J Mol Sci 2020; 21:E8893. [PMID: 33255277 PMCID: PMC7727666 DOI: 10.3390/ijms21238893] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 12/20/2022] Open
Abstract
Relevant, predictive normal, or disease model systems are of vital importance for drug development. The difference between nonhuman models and humans could contribute to clinical trial failures despite ideal nonhuman results. As a potential substitute for animal models, human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs) provide a powerful tool for drug toxicity screening, modeling cardiovascular diseases, and drug discovery. Here, we review recent hiPSC-CM disease models and discuss the features of hiPSC-CMs, including subtype and maturation and the tissue engineering technologies for drug assessment. Updates from the international multisite collaborators/administrations for development of novel drug discovery paradigms are also summarized.
Collapse
Affiliation(s)
- Junjun Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
- Department of Cell Design for Tissue Construction, Faculty of Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ying Hua
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Jingbo Zhang
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Lingjun Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Li Liu
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
- Department of Design for Tissue Regeneration, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| |
Collapse
|
180
|
Yadav V, Chong N, Ellis B, Ren X, Senapati S, Chang HC, Zorlutuna P. Constant-potential environment for activating and synchronizing cardiomyocyte colonies with on-chip ion-depleting perm-selective membranes. LAB ON A CHIP 2020; 20:4273-4284. [PMID: 33090162 DOI: 10.1039/d0lc00809e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
In this study, an ion depleted zone created by an ion-selective membrane was used to impose a high and uniform constant extracellular potential over an entire ∼1000 cell rat cardiomyocyte (rCM) colony on-a-chip to trigger synchronized voltage-gated ion channel activities while preserving cell viability, thus extending single-cell voltage-clamp ion channel studies to an entire normalized colony. Image analysis indicated that rCM beating was strengthened and accelerated (by a factor of ∼2) within minutes of ion depletion and the duration of contraction and relaxation phases was significantly reduced. After the initial synchronization, the entire colony responds collectively to external potential changes such that beating over the entire colony can be activated or deactivated within 0.1 s. These newly observed collective dynamic responses, due to simultaneous ion channel activation/deactivation by a uniform constant-potential extracellular environment, suggest that perm-selective membrane modules on cell culture chips can facilitate studies of extracellular cardiac cell electrical communication and how ion-channel related pathologies affect cardiac cell synchronization. The future applications of this new technology can lead to better drug screening platforms for cardiotoxicity as well as platforms that can facilitate synchronized maturation of pluripotent stem cells into colonies with high electrical connectivity.
Collapse
Affiliation(s)
- Vivek Yadav
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA. and Center for Microfluidics and Medical Diagnostics, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Nicholas Chong
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Bradley Ellis
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Xiang Ren
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Satyajyoti Senapati
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA. and Center for Microfluidics and Medical Diagnostics, University of Notre Dame, Notre Dame, IN 46556, USA and Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Hsueh-Chia Chang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA. and Center for Microfluidics and Medical Diagnostics, University of Notre Dame, Notre Dame, IN 46556, USA and Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA and Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Pinar Zorlutuna
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA. and Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA and Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
181
|
Abulaiti M, Yalikun Y, Murata K, Sato A, Sami MM, Sasaki Y, Fujiwara Y, Minatoya K, Shiba Y, Tanaka Y, Masumoto H. Establishment of a heart-on-a-chip microdevice based on human iPS cells for the evaluation of human heart tissue function. Sci Rep 2020. [DOI: 10.1201/9781420010138] [Citation(s) in RCA: 1419] [Impact Index Per Article: 283.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Abstract
Human iPS cell (iPSC)-derived cardiomyocytes (CMs) hold promise for drug discovery for heart diseases and cardiac toxicity tests. To utilize human iPSC-derived CMs, the establishment of three-dimensional (3D) heart tissues from iPSC-derived CMs and other heart cells, and a sensitive bioassay system to depict physiological heart function are anticipated. We have developed a heart-on-a-chip microdevice (HMD) as a novel system consisting of dynamic culture-based 3D cardiac microtissues derived from human iPSCs and microelectromechanical system (MEMS)-based microfluidic chips. The HMDs could visualize the kinetics of cardiac microtissue pulsations by monitoring particle displacement, which enabled us to quantify the physiological parameters, including fluidic output, pressure, and force. The HMDs demonstrated a strong correlation between particle displacement and the frequency of external electrical stimulation. The transition patterns were validated by a previously reported versatile video-based system to evaluate contractile function. The patterns are also consistent with oscillations of intracellular calcium ion concentration of CMs, which is a fundamental biological component of CM contraction. The HMDs showed a pharmacological response to isoproterenol, a β-adrenoceptor agonist, that resulted in a strong correlation between beating rate and particle displacement. Thus, we have validated the basic performance of HMDs as a resource for human iPSC-based pharmacological investigations.
Collapse
|
182
|
Abulaiti M, Yalikun Y, Murata K, Sato A, Sami MM, Sasaki Y, Fujiwara Y, Minatoya K, Shiba Y, Tanaka Y, Masumoto H. Establishment of a heart-on-a-chip microdevice based on human iPS cells for the evaluation of human heart tissue function. Sci Rep 2020; 10:19201. [PMID: 33154509 PMCID: PMC7645446 DOI: 10.1038/s41598-020-76062-w] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 10/23/2020] [Indexed: 12/04/2022] Open
Abstract
Human iPS cell (iPSC)-derived cardiomyocytes (CMs) hold promise for drug discovery for heart diseases and cardiac toxicity tests. To utilize human iPSC-derived CMs, the establishment of three-dimensional (3D) heart tissues from iPSC-derived CMs and other heart cells, and a sensitive bioassay system to depict physiological heart function are anticipated. We have developed a heart-on-a-chip microdevice (HMD) as a novel system consisting of dynamic culture-based 3D cardiac microtissues derived from human iPSCs and microelectromechanical system (MEMS)-based microfluidic chips. The HMDs could visualize the kinetics of cardiac microtissue pulsations by monitoring particle displacement, which enabled us to quantify the physiological parameters, including fluidic output, pressure, and force. The HMDs demonstrated a strong correlation between particle displacement and the frequency of external electrical stimulation. The transition patterns were validated by a previously reported versatile video-based system to evaluate contractile function. The patterns are also consistent with oscillations of intracellular calcium ion concentration of CMs, which is a fundamental biological component of CM contraction. The HMDs showed a pharmacological response to isoproterenol, a β-adrenoceptor agonist, that resulted in a strong correlation between beating rate and particle displacement. Thus, we have validated the basic performance of HMDs as a resource for human iPSC-based pharmacological investigations.
Collapse
Affiliation(s)
- Mosha Abulaiti
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima Minami-machi, Chuo-Ku, Kobe, 650-0047, Japan.,Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Laboratory for Integrated Biodevice, RIKEN Center for Biosystems Dynamics Research, Suita, Japan
| | - Yaxiaer Yalikun
- Laboratory for Integrated Biodevice, RIKEN Center for Biosystems Dynamics Research, Suita, Japan
| | - Kozue Murata
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima Minami-machi, Chuo-Ku, Kobe, 650-0047, Japan.,Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Japan
| | - Asako Sato
- Laboratory for Integrated Biodevice, RIKEN Center for Biosystems Dynamics Research, Suita, Japan
| | - Mustafa M Sami
- Laboratory for Morphogenetic Signaling, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Yuko Sasaki
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima Minami-machi, Chuo-Ku, Kobe, 650-0047, Japan
| | - Yasue Fujiwara
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima Minami-machi, Chuo-Ku, Kobe, 650-0047, Japan.,Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kenji Minatoya
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuji Shiba
- Department of Regenerative Science and Medicine, Institute for Biomedical Sciences, Shinshu University, Matsumoto, Japan
| | - Yo Tanaka
- Laboratory for Integrated Biodevice, RIKEN Center for Biosystems Dynamics Research, Suita, Japan
| | - Hidetoshi Masumoto
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima Minami-machi, Chuo-Ku, Kobe, 650-0047, Japan. .,Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
183
|
Gintant G, Kaushik EP, Feaster T, Stoelzle-Feix S, Kanda Y, Osada T, Smith G, Czysz K, Kettenhofen R, Lu HR, Cai B, Shi H, Herron TJ, Dang Q, Burton F, Pang L, Traebert M, Abassi Y, Pierson JB, Blinova K. Repolarization studies using human stem cell-derived cardiomyocytes: Validation studies and best practice recommendations. Regul Toxicol Pharmacol 2020; 117:104756. [PMID: 32822771 DOI: 10.1016/j.yrtph.2020.104756] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/24/2020] [Accepted: 07/31/2020] [Indexed: 12/20/2022]
Abstract
Human stem cell-derived cardiomyocytes (hSC-CMs) hold great promise as in vitro models to study the electrophysiological effects of novel drug candidates on human ventricular repolarization. Two recent large validation studies have demonstrated the ability of hSC-CMs to detect drug-induced delayed repolarization and "cellrhythmias" (interrupted repolarization or irregular spontaneous beating of myocytes) linked to Torsade-de-Pointes proarrhythmic risk. These (and other) studies have also revealed variability of electrophysiological responses attributable to differences in experimental approaches and experimenter, protocols, technology platforms used, and pharmacologic sensitivity of different human-derived models. Thus, when evaluating drug-induced repolarization effects, there is a need to consider 1) the advantages and disadvantages of different approaches, 2) the need for robust functional characterization of hSC-CM preparations to define "fit for purpose" applications, and 3) adopting standardized best practices to guide future studies with evolving hSC-CM preparations. Examples provided and suggested best practices are instructional in defining consistent, reproducible, and interpretable "fit for purpose" hSC-CM-based applications. Implementation of best practices should enhance the clinical translation of hSC-CM-based cell and tissue preparations in drug safety evaluations and support their growing role in regulatory filings.
Collapse
Affiliation(s)
- Gary Gintant
- Department of Integrative Pharmacology, Integrated Sciences and Technology, AbbVie, North Chicago, IL, 60064, USA.
| | | | - Tromondae Feaster
- Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, MD, 20993, USA.
| | | | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, Kanagawa, Japan.
| | | | - Godfrey Smith
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Scotland, UK; Clyde Biosciences Ltd., Scotland, UK.
| | | | - Ralf Kettenhofen
- Fraunhofer-Institute for Biomed Engineering IBMT, Sulzbach, Germany.
| | - Hua Rong Lu
- Nonclinical Safety, Johnson & Johnson R&D, Beerse, Belgium.
| | - Beibei Cai
- Takeda California, Inc., San Diego, CA, 92121, USA.
| | - Hong Shi
- Bristol-Myers Squibb, New York, NY, 10016, USA.
| | - Todd Joseph Herron
- Frankel Cardiovascular Regeneration Core Laboratory, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Qianyu Dang
- Office of Biostatistics, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, 20993, USA.
| | - Francis Burton
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Scotland, UK; Clyde Biosciences Ltd., Scotland, UK.
| | - Li Pang
- Division of Systems Biology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA.
| | | | - Yama Abassi
- Agilent Technologies, San Diego, CA, 92121, USA.
| | | | - Ksenia Blinova
- Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, MD, 20993, USA.
| |
Collapse
|
184
|
Majid QA, Fricker ATR, Gregory DA, Davidenko N, Hernandez Cruz O, Jabbour RJ, Owen TJ, Basnett P, Lukasiewicz B, Stevens M, Best S, Cameron R, Sinha S, Harding SE, Roy I. Natural Biomaterials for Cardiac Tissue Engineering: A Highly Biocompatible Solution. Front Cardiovasc Med 2020; 7:554597. [PMID: 33195451 PMCID: PMC7644890 DOI: 10.3389/fcvm.2020.554597] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases (CVD) constitute a major fraction of the current major global diseases and lead to about 30% of the deaths, i.e., 17.9 million deaths per year. CVD include coronary artery disease (CAD), myocardial infarction (MI), arrhythmias, heart failure, heart valve diseases, congenital heart disease, and cardiomyopathy. Cardiac Tissue Engineering (CTE) aims to address these conditions, the overall goal being the efficient regeneration of diseased cardiac tissue using an ideal combination of biomaterials and cells. Various cells have thus far been utilized in pre-clinical studies for CTE. These include adult stem cell populations (mesenchymal stem cells) and pluripotent stem cells (including autologous human induced pluripotent stem cells or allogenic human embryonic stem cells) with the latter undergoing differentiation to form functional cardiac cells. The ideal biomaterial for cardiac tissue engineering needs to have suitable material properties with the ability to support efficient attachment, growth, and differentiation of the cardiac cells, leading to the formation of functional cardiac tissue. In this review, we have focused on the use of biomaterials of natural origin for CTE. Natural biomaterials are generally known to be highly biocompatible and in addition are sustainable in nature. We have focused on those that have been widely explored in CTE and describe the original work and the current state of art. These include fibrinogen (in the context of Engineered Heart Tissue, EHT), collagen, alginate, silk, and Polyhydroxyalkanoates (PHAs). Amongst these, fibrinogen, collagen, alginate, and silk are isolated from natural sources whereas PHAs are produced via bacterial fermentation. Overall, these biomaterials have proven to be highly promising, displaying robust biocompatibility and, when combined with cells, an ability to enhance post-MI cardiac function in pre-clinical models. As such, CTE has great potential for future clinical solutions and hence can lead to a considerable reduction in mortality rates due to CVD.
Collapse
Affiliation(s)
- Qasim A. Majid
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Annabelle T. R. Fricker
- Department of Material Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| | - David A. Gregory
- Department of Material Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Natalia Davidenko
- Department of Materials Science and Metallurgy, Cambridge Centre for Medical Materials, University of Cambridge, Cambridge, United Kingdom
| | - Olivia Hernandez Cruz
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Bioengineering, Department of Materials, IBME, Faculty of Engineering, Imperial College London, United Kingdom
| | - Richard J. Jabbour
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Thomas J. Owen
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Pooja Basnett
- Applied Biotechnology Research Group, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London, United Kingdom
| | - Barbara Lukasiewicz
- Applied Biotechnology Research Group, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London, United Kingdom
| | - Molly Stevens
- Department of Bioengineering, Department of Materials, IBME, Faculty of Engineering, Imperial College London, United Kingdom
| | - Serena Best
- Department of Materials Science and Metallurgy, Cambridge Centre for Medical Materials, University of Cambridge, Cambridge, United Kingdom
| | - Ruth Cameron
- Department of Materials Science and Metallurgy, Cambridge Centre for Medical Materials, University of Cambridge, Cambridge, United Kingdom
| | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Sian E. Harding
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Ipsita Roy
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Material Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
185
|
Pappas MP, Peifer LN, Chan SSK. Dual TGFβ and Wnt inhibition promotes Mesp1-mediated mouse pluripotent stem cell differentiation into functional cardiomyocytes. Dev Growth Differ 2020; 62:487-494. [PMID: 33048365 DOI: 10.1111/dgd.12694] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 08/26/2020] [Accepted: 09/11/2020] [Indexed: 12/15/2022]
Abstract
Efficient derivation of cardiomyocytes from mouse pluripotent stem cells has proven challenging, and existing approaches rely on expensive supplementation or extensive manipulation. Mesp1 is a transcription factor that regulates cardiovascular specification during embryo development, and its overexpression has been shown to promote cardiogenesis. Here, we utilize a doxycycline-inducible Mesp1-expressing mouse embryonic stem cell system to develop an efficient differentiation protocol to generate functional cardiomyocytes. Our cardiac differentiation method involves transient Mesp1 induction following by subsequent dual inhibition of TGFβ and Wnt signaling pathways using small molecules. We discovered that whereas TGFβ inhibition promoted Mesp1-induced cardiac differentiation, Wnt inhibition was ineffective. Nevertheless, a combined inhibition of both pathways was superior to either inhibition alone in generating cardiomyocytes. These observations suggested a potential interaction between TGFβ and Wnt signaling pathways in the context of Mesp1-induced cardiac differentiation. Using a step-by-step approach, we have further optimized the windows of Mesp1 induction, TGFβ inhibition and Wnt inhibition to yield a maximal cardiomyocyte output - Mesp1 was induced first, followed by dual inhibition of TGFβ and Wnt signaling. Our protocol is capable of producing approximately 50% of cardiomyocytes in 12 days, which is comparable to existing methods, and have the advantages of being technically simple and inexpensive. Moreover, cardiomyocytes thus derived are functional, displaying intrinsic contractile capacity and contraction in response to electric stimulus. Derivation of mouse cardiomyocytes without the use of growth factors or other costly supplementation provides an accessible cell source for future applications.
Collapse
Affiliation(s)
- Matthew P Pappas
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Lindsay N Peifer
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Sunny S K Chan
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA.,Paul and Shelia Wellstone Muscular Dystrophy Center, Stem Cell Institute, Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
186
|
Effects of fibrillin mutations on the behavior of heart muscle cells in Marfan syndrome. Sci Rep 2020; 10:16756. [PMID: 33028885 PMCID: PMC7542175 DOI: 10.1038/s41598-020-73802-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 09/21/2020] [Indexed: 12/29/2022] Open
Abstract
Marfan syndrome (MFS) is a systemic disorder of connective tissue caused by pathogenic variants in the fibrillin-1 (FBN1) gene. Myocardial dysfunction has been demonstrated in MFS patients and mouse models, but little is known about the intrinsic effect on the cardiomyocytes (CMs). In this study, both induced pluripotent stem cells derived from a MFS-patient and the line with the corrected FBN1 mutation were differentiated to CMs. Several functional analyses are performed on this model to study MFS related cardiomyopathy. Atomic force microscopy revealed that MFS CMs are stiffer compared to corrected CMs. The contraction amplitude of MFS CMs is decreased compared to corrected CMs. Under normal culture conditions, MFS CMs show a lower beat-to-beat variability compared to corrected CMs using multi electrode array. Isoproterenol-induced stress or cyclic strain demonstrates lack of support from the matrix in MFS CMs. This study reports the first cardiac cell culture model for MFS, revealing abnormalities in the behavior of MFS CMs that are related to matrix defects. Based on these results, we postulate that impaired support from the extracellular environment plays a key role in the improper functioning of CMs in MFS.
Collapse
|
187
|
Aalders J, Léger L, Tuerlings T, Ledda S, van Hengel J. Liquid marble technology to create cost-effective 3D cardiospheres as a platform for in vitro drug testing and disease modelling. MethodsX 2020; 7:101065. [PMID: 33005571 PMCID: PMC7509398 DOI: 10.1016/j.mex.2020.101065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 09/09/2020] [Indexed: 01/28/2023] Open
Abstract
Three-dimensional (3D) cell culturing has several advantages over 2D cultures. 3D cell cultures more accurately mimic the in vivo environment, which is vital to obtain reliable results in disease modelling and toxicity testing. With the introduction of the Yamanaka factors, reprogramming of somatic cells to induced pluripotent stem cells (iPSCs) became available. This iPSC technology provides a scalable source of differentiated cells. iPSCs can be programmed to differentiate into any cell type of the body, including cardiomyocytes. These heart-specific muscle cells, can then serve as a model for therapeutic drug screening or assay development. Current methods to achieve multicellular spheroids by 3D cell cultures, such as hanging drop and spinner flasks are expensive, time-consuming and require specialized materials and training. Hydrophobic powders can be used to create a micro environment for cell cultures, which are termed liquid marbles (LM). In this procedure we describe the first use of the LM technology for 3D culturing in vitro derived human cardiomyocytes which results in the formation of cardiospheres within 24h. The cardiospheres could be used for several in depth and high-throughput analyses.
Collapse
Affiliation(s)
- Jeffrey Aalders
- Medical Cell Biology research group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Corneel Heymanslaan 10, Building B, Entrance 36, 9000 Ghent, Belgium
| | - Laurens Léger
- Medical Cell Biology research group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Corneel Heymanslaan 10, Building B, Entrance 36, 9000 Ghent, Belgium
| | - Tim Tuerlings
- Medical Cell Biology research group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Corneel Heymanslaan 10, Building B, Entrance 36, 9000 Ghent, Belgium
| | - Sergio Ledda
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy
| | - Jolanda van Hengel
- Medical Cell Biology research group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Corneel Heymanslaan 10, Building B, Entrance 36, 9000 Ghent, Belgium
| |
Collapse
|
188
|
Ito M, Nomura S, Morita H, Komuro I. Trends and Limitations in the Assessment of the Contractile Properties of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes From Patients With Dilated Cardiomyopathy. Front Cardiovasc Med 2020; 7:154. [PMID: 33102534 PMCID: PMC7494730 DOI: 10.3389/fcvm.2020.00154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/22/2020] [Indexed: 12/14/2022] Open
Abstract
The application of human induced pluripotent stem cell-derived cardiomyocytes (hiPSCMs) from patients is expected in disease modeling and drug screening in vitro. Dilated cardiomyopathy (DCM) is an intractable disease characterized by the impairment of systolic function and leads to severe heart failure. A number of researchers have focused on disease modeling of DCM and reproduced its pathologic phenotypes in hiPSCMs, but a robust method to evaluate the contractile properties of cardiomyocytes in vitro has not been standardized. In addition, it is unknown whether the throughput of measurements and analyses could be increased sufficiently for compound screening. Here, we reviewed the articles in which the contractile abnormalities of DCM hiPSCMs were recapitulated and assessed the trends and problems in sample preparation and evaluation. We found that single-cell level analysis was ineffective in some cases, and a tissue engineering approach has become dominant recently because of its increased efficiency in reproducing impaired contractility. We also examined two commercially available automated measurement devices with moderate throughput for motion analysis using two-dimensional hiPSCM sheets composed of originally established DCM hiPSCMs. As a result, both of the tested devices, an impedance analyzer and a video image-based cell motion analyzer, were not effective in detecting the expected reduction of contractility in the DCM clone. These findings collectively suggest that a tissue engineering approach could expand the potential of disease modeling with hiPSCMs, and so far, appropriate methods for in vitro force measurement with sufficient throughput, but without sacrificing physiological fidelity, are awaited.
Collapse
Affiliation(s)
- Masamichi Ito
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Seitaro Nomura
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Morita
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
189
|
Video-based assessment of drug-induced effects on contractile motion properties using human induced pluripotent stem cell-derived cardiomyocytes. J Pharmacol Toxicol Methods 2020; 105:106893. [DOI: 10.1016/j.vascn.2020.106893] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/28/2020] [Accepted: 06/21/2020] [Indexed: 02/08/2023]
|
190
|
Krishnamoorthi MK, Sarig U, Baruch L, Ting S, Reuveny S, Oh S, Goldfracht I, Gepstein L, Venkatraman SS, Tan LP, Machluf M. Robust Fabrication of Composite 3D Scaffolds with Tissue-Specific Bioactivity: A Proof-of-Concept Study. ACS APPLIED BIO MATERIALS 2020; 3:4974-4986. [PMID: 35021675 DOI: 10.1021/acsabm.0c00310] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The basic requirement of any engineered scaffold is to mimic the native tissue extracellular matrix (ECM). Despite substantial strides in understanding the ECM, scaffold fabrication processes of sufficient product robustness and bioactivity require further investigation, owing to the complexity of the natural ECM. A promising bioacive platform for cardiac tissue engineering is that of decellularized porcine cardiac ECM (pcECM, used here as a soft tissue representative model). However, this platform's complexity and batch-to-batch variability serve as processing limitations in attaining a robust and tunable cardiac tissue-specific bioactive scaffold. To address these issues, we fabricated 3D composite scaffolds (3DCSs) that demonstrate comparable physical and biochemical properties to the natural pcECM using wet electrospinning and functionalization with a pcECM hydrogel. The fabricated 3DCSs are non-immunogenic in vitro and support human mesenchymal stem cells' proliferation. Most importantly, the 3DCSs demonstrate tissue-specific bioactivity in inducing spontaneous cardiac lineage differentiation in human induced pluripotent stem cells (hiPSC) and further support the viability, functionality, and maturation of hiPSC-derived cardiomyocytes. Overall, this work illustrates the technology to fabricate robust yet tunable 3D scaffolds of tissue-specific bioactivity (with a proof of concept provided for cardiac tissues) as a platform for basic materials science studies and possible future R&D application in regenerative medicine.
Collapse
Affiliation(s)
- Muthu Kumar Krishnamoorthi
- School of Materials Science & Engineering, Nanyang Technological University (NTU), 50 Nanyang Avenue, 639798 Singapore.,Faculty of Biotechnology & Food Engineering, Technion-Israel Institute of Technology (IIT), Haifa 32000, Israel
| | - Udi Sarig
- School of Materials Science & Engineering, Nanyang Technological University (NTU), 50 Nanyang Avenue, 639798 Singapore.,Faculty of Biotechnology & Food Engineering, Technion-Israel Institute of Technology (IIT), Haifa 32000, Israel.,Biotechnology & Food Engineering, Technion-Israel Institute of Technology (IIT), Guangdong-Technion Israel Institute of Technology (GTIIT), Shantou, Guangdong Province, 515063 P.R. China
| | - Limor Baruch
- Faculty of Biotechnology & Food Engineering, Technion-Israel Institute of Technology (IIT), Haifa 32000, Israel
| | - Sherwin Ting
- Bioprocessing Technology Institute, A*STAR, 20 Biopolis Way, 138668 Singapore
| | - Shaul Reuveny
- Bioprocessing Technology Institute, A*STAR, 20 Biopolis Way, 138668 Singapore
| | - Steve Oh
- Bioprocessing Technology Institute, A*STAR, 20 Biopolis Way, 138668 Singapore
| | - Idit Goldfracht
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Efron St 1, Haifa 31096, Israel
| | - Lior Gepstein
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Efron St 1, Haifa 31096, Israel
| | - Subramanian S Venkatraman
- School of Materials Science & Engineering, Nanyang Technological University (NTU), 50 Nanyang Avenue, 639798 Singapore
| | - Lay Poh Tan
- School of Materials Science & Engineering, Nanyang Technological University (NTU), 50 Nanyang Avenue, 639798 Singapore
| | - Marcelle Machluf
- Faculty of Biotechnology & Food Engineering, Technion-Israel Institute of Technology (IIT), Haifa 32000, Israel
| |
Collapse
|
191
|
Branco MA, Cabral JM, Diogo MM. From Human Pluripotent Stem Cells to 3D Cardiac Microtissues: Progress, Applications and Challenges. Bioengineering (Basel) 2020; 7:E92. [PMID: 32785039 PMCID: PMC7552661 DOI: 10.3390/bioengineering7030092] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/30/2020] [Accepted: 08/06/2020] [Indexed: 12/19/2022] Open
Abstract
The knowledge acquired throughout the years concerning the in vivo regulation of cardiac development has promoted the establishment of directed differentiation protocols to obtain cardiomyocytes (CMs) and other cardiac cells from human pluripotent stem cells (hPSCs), which play a crucial role in the function and homeostasis of the heart. Among other developments in the field, the transition from homogeneous cultures of CMs to more complex multicellular cardiac microtissues (MTs) has increased the potential of these models for studying cardiac disorders in vitro and for clinically relevant applications such as drug screening and cardiotoxicity tests. This review addresses the state of the art of the generation of different cardiac cells from hPSCs and the impact of transitioning CM differentiation from 2D culture to a 3D environment. Additionally, current methods that may be employed to generate 3D cardiac MTs are reviewed and, finally, the adoption of these models for in vitro applications and their adaptation to medium- to high-throughput screening settings are also highlighted.
Collapse
Affiliation(s)
| | | | - Maria Margarida Diogo
- iBB-Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; (M.A.B.); (J.M.S.C.)
| |
Collapse
|
192
|
Goßmann M, Linder P, Thomas U, Juhasz K, Lemme M, George M, Fertig N, Dragicevic E, Stoelzle-Feix S. Integration of mechanical conditioning into a high throughput contractility assay for cardiac safety assessment. J Pharmacol Toxicol Methods 2020; 105:106892. [PMID: 32629160 DOI: 10.1016/j.vascn.2020.106892] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/29/2020] [Accepted: 06/18/2020] [Indexed: 01/10/2023]
Abstract
INDUCTION Despite increasing acceptance of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in safety pharmacology, controversy remains about the physiological relevance of existing in vitro models for their mechanical testing. We hypothesize that existing signs of immaturity of the cell models result from an improper mechanical environment. With the presented study, we aimed at validating the newly developed FLEXcyte96 technology with respect to physiological responses of hiPSC-CMs to pharmacological compounds with known inotropic and/or cardiotoxic effects. METHODS hiPSC-CMs were cultured in a 96-well format on hyperelastic silicone membranes imitating their native mechanical environment. Cardiomyocyte contractility was measured contact-free by application of capacitive displacement sensing of the cell-membrane biohybrids. Acute effects of positive inotropic compounds with distinct mechanisms of action were examined. Additionally, cardiotoxic effects of tyrosine kinase inhibitors and anthracyclines were repetitively examined during repeated exposure to drug concentrations for up to 5 days. RESULTS hiPSC-CMs grown on biomimetic membranes displayed increased contractility responses to isoproterenol, S-Bay K8644 and omecamtiv mecarbil without the need for additional stimulation. Tyrosine kinase inhibitor erlotinib, vandetanib, nilotinib, gefitinib, A-674563 as well as anthracycline idarubicin showed the expected cardiotoxic effects, including negative inotropy and induction of proarrhythmic events. DISCUSSION We conclude that the FLEXcyte 96 system is a reliable high throughput tool for invitro cardiac contractility research, providing the user with data obtained under physiological conditions which resemble the native environment of human heart tissue. We showed that the results obtained for both acute and sub-chronic compound administration are consistent with the respective physiological responses in humans.
Collapse
Affiliation(s)
| | - Peter Linder
- innoVitro GmbH, Artilleriestr 2, 52428 Jülich, Germany
| | - Ulrich Thomas
- Nanion Technologies GmbH, Ganghoferstr 70A, 80339 Munich, Germany
| | - Krisztina Juhasz
- Nanion Technologies GmbH, Ganghoferstr 70A, 80339 Munich, Germany; Institute for Nanoelectronics, Technische Universität München, Arcisstrasse 21, 80333 Munich, Germany
| | - Marta Lemme
- Nanion Technologies GmbH, Ganghoferstr 70A, 80339 Munich, Germany
| | - Michael George
- Nanion Technologies GmbH, Ganghoferstr 70A, 80339 Munich, Germany
| | - Niels Fertig
- Nanion Technologies GmbH, Ganghoferstr 70A, 80339 Munich, Germany
| | - Elena Dragicevic
- Nanion Technologies GmbH, Ganghoferstr 70A, 80339 Munich, Germany
| | | |
Collapse
|
193
|
Hanses U, Kleinsorge M, Roos L, Yigit G, Li Y, Barbarics B, El-Battrawy I, Lan H, Tiburcy M, Hindmarsh R, Lenz C, Salinas G, Diecke S, Müller C, Adham I, Altmüller J, Nürnberg P, Paul T, Zimmermann WH, Hasenfuss G, Wollnik B, Cyganek L. Intronic CRISPR Repair in a Preclinical Model of Noonan Syndrome-Associated Cardiomyopathy. Circulation 2020; 142:1059-1076. [PMID: 32623905 DOI: 10.1161/circulationaha.119.044794] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Noonan syndrome (NS) is a multisystemic developmental disorder characterized by common, clinically variable symptoms, such as typical facial dysmorphisms, short stature, developmental delay, intellectual disability as well as cardiac hypertrophy. The underlying mechanism is a gain-of-function of the RAS-mitogen-activated protein kinase signaling pathway. However, our understanding of the pathophysiological alterations and mechanisms, especially of the associated cardiomyopathy, remains limited and effective therapeutic options are lacking. METHODS Here, we present a family with two siblings displaying an autosomal recessive form of NS with massive hypertrophic cardiomyopathy as clinically the most prevalent symptom caused by biallelic mutations within the leucine zipper-like transcription regulator 1 (LZTR1). We generated induced pluripotent stem cell-derived cardiomyocytes of the affected siblings and investigated the patient-specific cardiomyocytes on the molecular and functional level. RESULTS Patients' induced pluripotent stem cell-derived cardiomyocytes recapitulated the hypertrophic phenotype and uncovered a so-far-not-described causal link between LZTR1 dysfunction, RAS-mitogen-activated protein kinase signaling hyperactivity, hypertrophic gene response and cellular hypertrophy. Calcium channel blockade and MEK inhibition could prevent some of the disease characteristics, providing a molecular underpinning for the clinical use of these drugs in patients with NS, but might not be a sustainable therapeutic option. In a proof-of-concept approach, we explored a clinically translatable intronic CRISPR (clustered regularly interspaced short palindromic repeats) repair and demonstrated a rescue of the hypertrophic phenotype. CONCLUSIONS Our study revealed the human cardiac pathogenesis in patient-specific induced pluripotent stem cell-derived cardiomyocytes from NS patients carrying biallelic variants in LZTR1 and identified a unique disease-specific proteome signature. In addition, we identified the intronic CRISPR repair as a personalized and in our view clinically translatable therapeutic strategy to treat NS-associated hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Ulrich Hanses
- Clinic for Cardiology and Pneumology (U.H., M.K., L.R., R.H., G.H., L.C.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Mandy Kleinsorge
- Clinic for Cardiology and Pneumology (U.H., M.K., L.R., R.H., G.H., L.C.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Lennart Roos
- Clinic for Cardiology and Pneumology (U.H., M.K., L.R., R.H., G.H., L.C.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Gökhan Yigit
- Institute of Human Genetics (G.Y., Y.L., G.S., C.M., I.A., B.W.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Yun Li
- Institute of Human Genetics (G.Y., Y.L., G.S., C.M., I.A., B.W.)
| | - Boris Barbarics
- Clinic for Pediatric Cardiology and Intensive Care Medicine (B.B., T.P.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Ibrahim El-Battrawy
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.).,First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany (I.E-B., H.L.)
| | - Huan Lan
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany (I.E-B., H.L.)
| | - Malte Tiburcy
- Institute of Pharmacology and Toxicology (M.T., W-H.Z.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Robin Hindmarsh
- Clinic for Cardiology and Pneumology (U.H., M.K., L.R., R.H., G.H., L.C.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Christof Lenz
- Institute for Clinical Chemistry (C.L.), University Medical Center Göttingen, Germany.,Bioanalytical Mass Spectrometry, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany (C.L.)
| | - Gabriela Salinas
- Institute of Human Genetics (G.Y., Y.L., G.S., C.M., I.A., B.W.)
| | - Sebastian Diecke
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.).,Stem Cell Core Facility, Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.D.).,Berlin Institute of Health, Germany (S.D.)
| | - Christian Müller
- Institute of Human Genetics (G.Y., Y.L., G.S., C.M., I.A., B.W.)
| | - Ibrahim Adham
- Institute of Human Genetics (G.Y., Y.L., G.S., C.M., I.A., B.W.)
| | - Janine Altmüller
- Cologne Center for Genomics, University of Cologne, Germany (J.A., P.N.)
| | - Peter Nürnberg
- Cologne Center for Genomics, University of Cologne, Germany (J.A., P.N.)
| | - Thomas Paul
- Clinic for Pediatric Cardiology and Intensive Care Medicine (B.B., T.P.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology (M.T., W-H.Z.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.).,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W-H.Z., G.H., B.W.)
| | - Gerd Hasenfuss
- Clinic for Cardiology and Pneumology (U.H., M.K., L.R., R.H., G.H., L.C.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.).,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W-H.Z., G.H., B.W.)
| | - Bernd Wollnik
- Institute of Human Genetics (G.Y., Y.L., G.S., C.M., I.A., B.W.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.).,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W-H.Z., G.H., B.W.)
| | - Lukas Cyganek
- Clinic for Cardiology and Pneumology (U.H., M.K., L.R., R.H., G.H., L.C.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| |
Collapse
|
194
|
Portillo‐Esquivel LE, Nanduri V, Zhang F, Liang W, Zhang B. z-Wire: A Microscaffold That Supports Guided Tissue Assembly and Intramyocardium Delivery for Cardiac Repair. Adv Healthc Mater 2020; 9:e2000358. [PMID: 32543115 DOI: 10.1002/adhm.202000358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/29/2020] [Indexed: 12/13/2022]
Abstract
Tissue engineering holds promise to replace damaged tissues for repair of vital organs in the human body. In cardiac repairs specifically, approaches are developed for intramyocardial delivery of cells and the epicardial delivery of tissue-engineered cardiac patches, providing benefit of cell localization and tissue structure, respectively. However, to improve cell retention and integration, there is a need for the intramyocardial delivery of functional tissues while preserving anisotropic muscle alignment. Here, a biodegradable z-wire scaffold that supports the scalable gel-free production of an array of functional cardiac tissues in a 384-well plate format is developed. The z-wire scaffold design supports cellular alignment, provides tunable mechanical support, and allows for tissue contraction. When the scaffold is imparted with magnetic properties, individual tissues can be assembled with macroscopic alignment under magnetic guidance. When used in combination with a customized surgical delivery tool, z-wire tissues can be injected directly into the myocardial wall, with controlled tissue orientation according to the injection path. This modular tissue engineering approach, in combination with the use of smart scaffolds, can expand opportunity in functional tissue delivery.
Collapse
Affiliation(s)
| | - Vibudha Nanduri
- Department of Chemical EngineeringMcMaster University 1280 Main Street West Hamilton ON L8S 4L8 Canada
| | - Feng Zhang
- School of Biomedical EngineeringMcMaster University 1280 Main Street West Hamilton ON L8S 4L8 Canada
| | - Wenbin Liang
- University of Ottawa Heart InstituteDepartment of Cellular and Molecular MedicineUniversity of Ottawa 40 Ruskin Street Ottawa ON K1Y 4W7 Canada
| | - Boyang Zhang
- Department of Chemical EngineeringMcMaster University 1280 Main Street West Hamilton ON L8S 4L8 Canada
- School of Biomedical EngineeringMcMaster University 1280 Main Street West Hamilton ON L8S 4L8 Canada
| |
Collapse
|
195
|
Qiao X, van der Zanden SY, Wander DPA, Borràs DM, Song JY, Li X, van Duikeren S, van Gils N, Rutten A, van Herwaarden T, van Tellingen O, Giacomelli E, Bellin M, Orlova V, Tertoolen LGJ, Gerhardt S, Akkermans JJ, Bakker JM, Zuur CL, Pang B, Smits AM, Mummery CL, Smit L, Arens R, Li J, Overkleeft HS, Neefjes J. Uncoupling DNA damage from chromatin damage to detoxify doxorubicin. Proc Natl Acad Sci U S A 2020; 117:15182-15192. [PMID: 32554494 DOI: 10.1073/pnas.1922072117/suppl_file/pnas.1922072117.sm04.mp4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023] Open
Abstract
The anthracycline doxorubicin (Doxo) and its analogs daunorubicin (Daun), epirubicin (Epi), and idarubicin (Ida) have been cornerstones of anticancer therapy for nearly five decades. However, their clinical application is limited by severe side effects, especially dose-dependent irreversible cardiotoxicity. Other detrimental side effects of anthracyclines include therapy-related malignancies and infertility. It is unclear whether these side effects are coupled to the chemotherapeutic efficacy. Doxo, Daun, Epi, and Ida execute two cellular activities: DNA damage, causing double-strand breaks (DSBs) following poisoning of topoisomerase II (Topo II), and chromatin damage, mediated through histone eviction at selected sites in the genome. Here we report that anthracycline-induced cardiotoxicity requires the combination of both cellular activities. Topo II poisons with either one of the activities fail to induce cardiotoxicity in mice and human cardiac microtissues, as observed for aclarubicin (Acla) and etoposide (Etop). Further, we show that Doxo can be detoxified by chemically separating these two activities. Anthracycline variants that induce chromatin damage without causing DSBs maintain similar anticancer potency in cell lines, mice, and human acute myeloid leukemia patients, implying that chromatin damage constitutes a major cytotoxic mechanism of anthracyclines. With these anthracyclines abstained from cardiotoxicity and therapy-related tumors, we thus uncoupled the side effects from anticancer efficacy. These results suggest that anthracycline variants acting primarily via chromatin damage may allow prolonged treatment of cancer patients and will improve the quality of life of cancer survivors.
Collapse
Affiliation(s)
- Xiaohang Qiao
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands;
- Department of Head and Neck Oncology and Surgery, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Sabina Y van der Zanden
- Department of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Dennis P A Wander
- Leiden Institute of Chemistry, Leiden University, 2300 RA Leiden, The Netherlands
| | - Daniel M Borràs
- Department of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Ji-Ying Song
- Division of Experimental Animal Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Xiaoyang Li
- Department of Hematology, Shanghai Institute of Hematology, National Research Center for Translational Medicine, RuiJin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Suzanne van Duikeren
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Noortje van Gils
- Department of Hematology, Vrije Universiteit Medical Center, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Arjo Rutten
- Department of Hematology, Vrije Universiteit Medical Center, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Tessa van Herwaarden
- Department of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Olaf van Tellingen
- Division of Diagnostic Oncology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Elisa Giacomelli
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Valeria Orlova
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Leon G J Tertoolen
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Sophie Gerhardt
- Central Laboratory Animal Facility, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Jimmy J Akkermans
- Department of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Jeroen M Bakker
- Department of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Charlotte L Zuur
- Department of Head and Neck Oncology and Surgery, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Baoxu Pang
- Department of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Anke M Smits
- Department of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Linda Smit
- Department of Hematology, Vrije Universiteit Medical Center, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Ramon Arens
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Junmin Li
- Department of Hematology, Shanghai Institute of Hematology, National Research Center for Translational Medicine, RuiJin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China;
| | - Hermen S Overkleeft
- Leiden Institute of Chemistry, Leiden University, 2300 RA Leiden, The Netherlands
| | - Jacques Neefjes
- Department of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands;
| |
Collapse
|
196
|
Abstract
The anthracycline doxorubicin (Doxo) and its analogs daunorubicin (Daun), epirubicin (Epi), and idarubicin (Ida) have been cornerstones of anticancer therapy for nearly five decades. However, their clinical application is limited by severe side effects, especially dose-dependent irreversible cardiotoxicity. Other detrimental side effects of anthracyclines include therapy-related malignancies and infertility. It is unclear whether these side effects are coupled to the chemotherapeutic efficacy. Doxo, Daun, Epi, and Ida execute two cellular activities: DNA damage, causing double-strand breaks (DSBs) following poisoning of topoisomerase II (Topo II), and chromatin damage, mediated through histone eviction at selected sites in the genome. Here we report that anthracycline-induced cardiotoxicity requires the combination of both cellular activities. Topo II poisons with either one of the activities fail to induce cardiotoxicity in mice and human cardiac microtissues, as observed for aclarubicin (Acla) and etoposide (Etop). Further, we show that Doxo can be detoxified by chemically separating these two activities. Anthracycline variants that induce chromatin damage without causing DSBs maintain similar anticancer potency in cell lines, mice, and human acute myeloid leukemia patients, implying that chromatin damage constitutes a major cytotoxic mechanism of anthracyclines. With these anthracyclines abstained from cardiotoxicity and therapy-related tumors, we thus uncoupled the side effects from anticancer efficacy. These results suggest that anthracycline variants acting primarily via chromatin damage may allow prolonged treatment of cancer patients and will improve the quality of life of cancer survivors.
Collapse
|
197
|
Selective Induction of Human Autonomic Neurons Enables Precise Control of Cardiomyocyte Beating. Sci Rep 2020; 10:9464. [PMID: 32528170 PMCID: PMC7289887 DOI: 10.1038/s41598-020-66303-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022] Open
Abstract
The autonomic nervous system (ANS) regulates tissue homeostasis and remodelling through antagonistic effects of noradrenergic sympathetic and cholinergic parasympathetic signalling. Despite numerous reports on the induction of sympathetic neurons from human pluripotent stem cells (hPSCs), no induction methods have effectively derived cholinergic parasympathetic neurons from hPSCs. Considering the antagonistic effects of noradrenergic and cholinergic inputs on target organs, both sympathetic and parasympathetic neurons are expected to be induced. This study aimed to develop a stepwise chemical induction method to induce sympathetic-like and parasympathetic-like ANS neurons. Autonomic specification was achieved through restricting signals inducing sensory or enteric neurogenesis and activating bone morphogenetic protein (BMP) signals. Global mRNA expression analyses after stepwise induction, including single-cell RNA-seq analysis of induced neurons and functional assays revealed that each induced sympathetic-like or parasympathetic-like neuron acquired pharmacological and electrophysiological functional properties with distinct marker expression. Further, we identified selective induction methods using appropriate seeding cell densities and neurotrophic factor concentrations. Neurons were individually induced, facilitating the regulation of the beating rates of hiPSC-derived cardiomyocytes in an antagonistic manner. The induction methods yield specific neuron types, and their influence on various tissues can be studied by co-cultured assays.
Collapse
|
198
|
Drug response analysis for scaffold-free cardiac constructs fabricated using bio-3D printer. Sci Rep 2020; 10:8972. [PMID: 32487993 PMCID: PMC7265390 DOI: 10.1038/s41598-020-65681-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 05/05/2020] [Indexed: 12/04/2022] Open
Abstract
Cardiac constructs fabricated using human induced pluripotent stem cells-derived cardiomyocytes (iPSCs-CMs) are useful for evaluating the cardiotoxicity of and cardiac response to new drugs. Previously, we fabricated scaffold-free three-dimensional (3D) tubular cardiac constructs using a bio-3D printer, which can load cardiac spheroids onto a needle array. In this study, we developed a method to measure the contractile force and to evaluate the drug response in cardiac constructs. Specifically, we measured the movement of the needle tip upon contraction of the cardiac constructs on the needle array. The contractile force and beating rate of the cardiac constructs were evaluated by analysing changes in the movement of the needle tip. To evaluate the drug response, contractile properties were measured following treatment with isoproterenol, propranolol, or blebbistatin, in which the movement of the needle tip was increased following isoproterenol treatment, but was decreased following propranolol or blebbistain, treatments. To evaluate cardiotoxicity, contraction and cell viability of the cardiac constructs were measured following doxorubicin treatment. Cell viability was found to decrease with decreasing movement of the needle tip following doxorubicin treatment. Collectively, our results show that this method can aid in evaluating the contractile force of cardiac constructs.
Collapse
|
199
|
Kramer JPM, Aigner TB, Petzold J, Roshanbinfar K, Scheibel T, Engel FB. Recombinant spider silk protein eADF4(C16)-RGD coatings are suitable for cardiac tissue engineering. Sci Rep 2020; 10:8789. [PMID: 32472031 PMCID: PMC7260369 DOI: 10.1038/s41598-020-65786-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/08/2020] [Indexed: 01/05/2023] Open
Abstract
Cardiac tissue engineering is a promising approach to treat cardiovascular diseases, which are a major socio-economic burden worldwide. An optimal material for cardiac tissue engineering, allowing cardiomyocyte attachment and exhibiting proper immunocompatibility, biocompatibility and mechanical characteristics, has not yet emerged. An additional challenge is to develop a fabrication method that enables the generation of proper hierarchical structures and constructs with a high density of cardiomyocytes for optimal contractility. Thus, there is a focus on identifying suitable materials for cardiac tissue engineering. Here, we investigated the interaction of neonatal rat heart cells with engineered spider silk protein (eADF4(C16)) tagged with the tripeptide arginyl-glycyl-aspartic acid cell adhesion motif RGD, which can be used as coating, but can also be 3D printed. Cardiomyocytes, fibroblasts, and endothelial cells attached well to eADF4(C16)-RGD coatings, which did not induce hypertrophy in cardiomyocytes, but allowed response to hypertrophic as well as proliferative stimuli. Furthermore, Kymograph and MUSCLEMOTION analyses showed proper cardiomyocyte beating characteristics on spider silk coatings, and cardiomyocytes formed compact cell aggregates, exhibiting markedly higher speed of contraction than cardiomyocyte mono-layers on fibronectin. The results suggest that eADF4(C16)-RGD is a promising material for cardiac tissue engineering.
Collapse
Affiliation(s)
- Johannes P M Kramer
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 12, 91054, Erlangen, Germany
| | - Tamara B Aigner
- Lehrstuhl Biomaterialien, Prof.-Rüdiger-Bormann Straße 1, 95447, Bayreuth, Germany
| | - Jana Petzold
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 12, 91054, Erlangen, Germany
| | - Kaveh Roshanbinfar
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 12, 91054, Erlangen, Germany
| | - Thomas Scheibel
- Lehrstuhl Biomaterialien, Prof.-Rüdiger-Bormann Straße 1, 95447, Bayreuth, Germany.
- Bayreuther Zentrum für Kolloide und Grenzflächen (BZKG), Bayerisches Polymerinstitut (BPI), Bayreuther Zentrum für Molekulare Biowissenschaften (BZMB), Bayreuther Materialzentrum (BayMAT), Universitätsstraße 30, Universität Bayreuth, Bayreuth, D-95447, Germany.
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 12, 91054, Erlangen, Germany.
- MURCE, Muscle Research Center Erlangen, Erlangen, Germany.
| |
Collapse
|
200
|
Giacomelli E, Meraviglia V, Campostrini G, Cochrane A, Cao X, van Helden RWJ, Krotenberg Garcia A, Mircea M, Kostidis S, Davis RP, van Meer BJ, Jost CR, Koster AJ, Mei H, Míguez DG, Mulder AA, Ledesma-Terrón M, Pompilio G, Sala L, Salvatori DCF, Slieker RC, Sommariva E, de Vries AAF, Giera M, Semrau S, Tertoolen LGJ, Orlova VV, Bellin M, Mummery CL. Human-iPSC-Derived Cardiac Stromal Cells Enhance Maturation in 3D Cardiac Microtissues and Reveal Non-cardiomyocyte Contributions to Heart Disease. Cell Stem Cell 2020; 26:862-879.e11. [PMID: 32459996 PMCID: PMC7284308 DOI: 10.1016/j.stem.2020.05.004] [Citation(s) in RCA: 378] [Impact Index Per Article: 75.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/05/2020] [Accepted: 05/01/2020] [Indexed: 12/12/2022]
Abstract
Cardiomyocytes (CMs) from human induced pluripotent stem cells (hiPSCs) are functionally immature, but this is improved by incorporation into engineered tissues or forced contraction. Here, we showed that tri-cellular combinations of hiPSC-derived CMs, cardiac fibroblasts (CFs), and cardiac endothelial cells also enhance maturation in easily constructed, scaffold-free, three-dimensional microtissues (MTs). hiPSC-CMs in MTs with CFs showed improved sarcomeric structures with T-tubules, enhanced contractility, and mitochondrial respiration and were electrophysiologically more mature than MTs without CFs. Interactions mediating maturation included coupling between hiPSC-CMs and CFs through connexin 43 (CX43) gap junctions and increased intracellular cyclic AMP (cAMP). Scaled production of thousands of hiPSC-MTs was highly reproducible across lines and differentiated cell batches. MTs containing healthy-control hiPSC-CMs but hiPSC-CFs from patients with arrhythmogenic cardiomyopathy strikingly recapitulated features of the disease. Our MT model is thus a simple and versatile platform for modeling multicellular cardiac diseases that will facilitate industry and academic engagement in high-throughput molecular screening. Cardiac fibroblasts and endothelial cells induce hiPSC-cardiomyocyte maturation CX43 gap junctions form between cardiac fibroblasts and cardiomyocytes cAMP-pathway activation contributes to hiPSC-cardiomyocyte maturation Patient-derived hiPSC-cardiac fibroblasts cause arrhythmia in microtissues
Collapse
Affiliation(s)
- Elisa Giacomelli
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Viviana Meraviglia
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Giulia Campostrini
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Amy Cochrane
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Xu Cao
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Ruben W J van Helden
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Ana Krotenberg Garcia
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Maria Mircea
- Leiden Institute of Physics, Leiden University, 2333 Leiden, the Netherlands
| | - Sarantos Kostidis
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Richard P Davis
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Berend J van Meer
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Carolina R Jost
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Abraham J Koster
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - David G Míguez
- Centro de Biologia Molecular Severo Ochoa, Departamento de Física de la Materia Condensada, Instituto Nicolas Cabrera and Condensed Matter Physics Center (IFIMAC), Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Aat A Mulder
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Mario Ledesma-Terrón
- Centro de Biologia Molecular Severo Ochoa, Departamento de Física de la Materia Condensada, Instituto Nicolas Cabrera and Condensed Matter Physics Center (IFIMAC), Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Giulio Pompilio
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Luca Sala
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Daniela C F Salvatori
- Central Laboratory Animal Facility, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Roderick C Slieker
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 Leiden, the Netherlands; Department of Epidemiology and Biostatistics, Amsterdam Public Health Institute, VU University Medical Center, 1007 Amsterdam, the Netherlands
| | - Elena Sommariva
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Antoine A F de Vries
- Department of Cardiology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Stefan Semrau
- Leiden Institute of Physics, Leiden University, 2333 Leiden, the Netherlands
| | - Leon G J Tertoolen
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Valeria V Orlova
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands.
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands; Department of Biology, University of Padua, 35121 Padua, Italy; Veneto Institute of Molecular Medicine, 35129 Padua, Italy.
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands; Department of Applied Stem Cell Technologies, University of Twente, 7500 Enschede, the Netherlands.
| |
Collapse
|