1
|
Aghavali R, Roberts EG, Kurokawa YK, Mak E, Chan MYC, Wong AOT, Li RA, Costa KD. Enhanced drug classification using machine learning with multiplexed cardiac contractility assays. Pharmacol Res 2024; 209:107459. [PMID: 39396765 DOI: 10.1016/j.phrs.2024.107459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 09/04/2024] [Accepted: 10/08/2024] [Indexed: 10/15/2024]
Abstract
Cardiac screening of newly discovered drugs remains a longstanding challenge for the pharmaceutical industry. While therapeutic efficacy and cardiotoxicity are evaluated through preclinical biochemical and animal testing, 90 % of lead compounds fail to meet safety and efficacy benchmarks during human clinical trials. A preclinical model more representative of the human cardiac response is needed; heart tissue engineered from human pluripotent stem cell derived cardiomyocytes offers such a platform. In this study, three functionally distinct and independently validated engineered cardiac tissue assays are exposed to increasing concentrations of known compounds representing 5 classes of mechanistic action, creating a robust electrophysiology and contractility dataset. Combining results from six individual models, the resulting ensemble algorithm can classify the mechanistic action of unknown compounds with 86.2 % predictive accuracy. This outperforms single-assay models and offers a strategy to enhance future clinical trial success aligned with the recent FDA Modernization Act 2.0.
Collapse
Affiliation(s)
- Reza Aghavali
- Novoheart, Medera Inc., 6 Tide St., Boston, MA 02210, USA.
| | - Erin G Roberts
- Novoheart, Medera Inc., 6 Tide St., Boston, MA 02210, USA.
| | | | - Erica Mak
- Novoheart, Medera Inc., 6 Tide St., Boston, MA 02210, USA.
| | | | - Andy O T Wong
- Novoheart, Medera Inc., 6 Tide St., Boston, MA 02210, USA.
| | - Ronald A Li
- Novoheart, Medera Inc., 6 Tide St., Boston, MA 02210, USA.
| | - Kevin D Costa
- Novoheart, Medera Inc., 6 Tide St., Boston, MA 02210, USA.
| |
Collapse
|
2
|
Sun YH, Kao HKJ, Thai PN, Smithers R, Chang CW, Pretto D, Yechikov S, Oppenheimer S, Bedolla A, Chalker BA, Ghobashy R, Nolta JA, Chan JW, Chiamvimonvat N, Lieu DK. The sinoatrial node extracellular matrix promotes pacemaker phenotype and protects automaticity in engineered heart tissues from cyclic strain. Cell Rep 2023; 42:113505. [PMID: 38041810 PMCID: PMC10790625 DOI: 10.1016/j.celrep.2023.113505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 10/17/2023] [Accepted: 11/13/2023] [Indexed: 12/04/2023] Open
Abstract
The composite material-like extracellular matrix (ECM) in the sinoatrial node (SAN) supports the native pacemaking cardiomyocytes (PCMs). To test the roles of SAN ECM in the PCM phenotype and function, we engineered reconstructed-SAN heart tissues (rSANHTs) by recellularizing porcine SAN ECMs with hiPSC-derived PCMs. The hiPSC-PCMs in rSANHTs self-organized into clusters resembling the native SAN and displayed higher expression of pacemaker-specific genes and a faster automaticity compared with PCMs in reconstructed-left ventricular heart tissues (rLVHTs). To test the protective nature of SAN ECMs under strain, rSANHTs and rLVHTs were transplanted onto the murine thoracic diaphragm to undergo constant cyclic strain. All strained-rSANHTs preserved automaticity, whereas 66% of strained-rLVHTs lost their automaticity. In contrast to the strained-rLVHTs, PCMs in strained-rSANHTs maintained high expression of key pacemaker genes (HCN4, TBX3, and TBX18). These findings highlight the promotive and protective roles of the composite SAN ECM and provide valuable insights for pacemaking tissue engineering.
Collapse
Affiliation(s)
- Yao-Hui Sun
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95616, USA; Institute for Regenerative Cures and Stem Cell Program, University of California, Davis, Sacramento, CA 95817, USA
| | - Hillary K J Kao
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95616, USA; Institute for Regenerative Cures and Stem Cell Program, University of California, Davis, Sacramento, CA 95817, USA
| | - Phung N Thai
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Regan Smithers
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95616, USA; Institute for Regenerative Cures and Stem Cell Program, University of California, Davis, Sacramento, CA 95817, USA
| | - Che-Wei Chang
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Dalyir Pretto
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95616, USA; Institute for Regenerative Cures and Stem Cell Program, University of California, Davis, Sacramento, CA 95817, USA
| | - Sergey Yechikov
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95616, USA; Institute for Regenerative Cures and Stem Cell Program, University of California, Davis, Sacramento, CA 95817, USA
| | - Sarah Oppenheimer
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95616, USA; Institute for Regenerative Cures and Stem Cell Program, University of California, Davis, Sacramento, CA 95817, USA; Bridges to Stem Cell Research Program, California State University, Sacramento, Sacramento, CA 95817, USA
| | - Amanda Bedolla
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95616, USA; Institute for Regenerative Cures and Stem Cell Program, University of California, Davis, Sacramento, CA 95817, USA; Bridges to Stem Cell Research Program, California State University, Sacramento, Sacramento, CA 95817, USA
| | - Brooke A Chalker
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95616, USA; Institute for Regenerative Cures and Stem Cell Program, University of California, Davis, Sacramento, CA 95817, USA; Bridges to Stem Cell Research Program, Cal Poly Humboldt, Humboldt, CA 95521, USA
| | - Rana Ghobashy
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95616, USA; Institute for Regenerative Cures and Stem Cell Program, University of California, Davis, Sacramento, CA 95817, USA; Bridges to Stem Cell Research Program, California State University, Sacramento, Sacramento, CA 95817, USA
| | - Jan A Nolta
- Institute for Regenerative Cures and Stem Cell Program, University of California, Davis, Sacramento, CA 95817, USA
| | - James W Chan
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95616, USA; Department of Veterans Affairs, Northern California Health Care System, Mather, CA 95655, USA
| | - Deborah K Lieu
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95616, USA; Institute for Regenerative Cures and Stem Cell Program, University of California, Davis, Sacramento, CA 95817, USA.
| |
Collapse
|
3
|
English EJ, Samolyk BL, Gaudette GR, Pins GD. Micropatterned fibrin scaffolds increase cardiomyocyte alignment and contractility for the fabrication of engineered myocardial tissue. J Biomed Mater Res A 2023; 111:1309-1321. [PMID: 36932841 PMCID: PMC11128133 DOI: 10.1002/jbm.a.37530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 02/09/2023] [Accepted: 03/01/2023] [Indexed: 03/19/2023]
Abstract
Cardiovascular disease is the leading cause of death in the United States, which can result in blockage of a coronary artery, triggering a myocardial infarction (MI), scar tissue formation in the myocardium, and ultimately heart failure. Currently, the gold-standard solution for total heart failure is a heart transplantation. An alternative to total-organ transplantation is surgically remodeling the ventricle with the implantation of a cardiac patch. Acellular cardiac patches have previously been investigated using synthetic or decellularized native materials to improve cardiac function. However, a limitation of this strategy is that acellular cardiac patches only reshape the ventricle and do not increase cardiac contractile function. Toward the development of a cardiac patch, our laboratory previously developed a cell-populated composite fibrin scaffold and aligned microthreads to recapitulate the mechanical properties of native myocardium. In this study, we explore micropatterning the surfaces of fibrin gels to mimic anisotropic native tissue architecture and promote cellular alignment of human induced pluripotent stem cell cardiomyocytes (hiPS-CM), which is crucial for increasing scaffold contractile properties. hiPS-CMs seeded on micropatterned surfaces exhibit cellular elongation, distinct sarcomere alignment, and circumferential connexin-43 staining at 14 days of culture, which are necessary for mature contractile properties. Constructs were also subject to electrical stimulation during culture to promote increased contractile properties. After 7 days of stimulation, contractile strains of micropatterned constructs were significantly higher than unpatterned controls. These results suggest that the use of micropatterned topographic cues on fibrin scaffolds may be a promising strategy for creating engineered cardiac tissue.
Collapse
Affiliation(s)
- Elizabeth J. English
- Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
- Tessera Therapeutics, Somerville, Massachusetts, USA
| | - Bryanna L. Samolyk
- Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Glenn R. Gaudette
- Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
- Department of Engineering, Boston College, Newton, Massachusetts, USA
| | - George D. Pins
- Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| |
Collapse
|
4
|
Cheng S, Brenière-Letuffe D, Ahola V, Wong AO, Keung HY, Gurung B, Zheng Z, Costa KD, Lieu DK, Keung W, Li RA. Single-cell RNA sequencing reveals maturation trajectory in human pluripotent stem cell-derived cardiomyocytes in engineered tissues. iScience 2023; 26:106302. [PMID: 36950112 PMCID: PMC10025988 DOI: 10.1016/j.isci.2023.106302] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 01/04/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023] Open
Abstract
Cardiac in vitro models have become increasingly obtainable and affordable with the optimization of human pluripotent stem cell-derived cardiomyocyte (hPSC-CM) differentiation. However, these CMs are immature compared to their in vivo counterparts. Here we study the cellular phenotype of hPSC-CMs by comparing their single-cell gene expression and functional profiles in three engineered cardiac tissue configurations: human ventricular (hv) cardiac anisotropic sheet, cardiac tissue strip, and cardiac organoid chamber (hvCOC), with spontaneously aggregated 3D cardiac spheroids (CS) as control. The CM maturity was found to increase with increasing levels of complexity of the engineered tissues from CS to hvCOC. The contractile components are the first function to mature, followed by electrophysiology and oxidative metabolism. Notably, the 2D tissue constructs show a higher cellular organization whereas metabolic maturity preferentially increases in the 3D constructs. We conclude that the tissue engineering models resembling configurations of native tissues may be reliable for drug screening or disease modeling.
Collapse
Affiliation(s)
- Shangli Cheng
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Hong Kong SAR, China
| | - David Brenière-Letuffe
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Hong Kong SAR, China
- Department of Clinical Sciences, Intervention and Technology, CLINTEC, Karolinska Institutet, 141 52 Stockholm, Sweden
- Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, 141 86 Stockholm, Sweden
| | - Virpi Ahola
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Hong Kong SAR, China
| | | | - Hoi Yee Keung
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Hong Kong SAR, China
| | - Bimal Gurung
- Novoheart, Irvine, CA 92617, USA
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zongli Zheng
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Hong Kong SAR, China
| | - Kevin D. Costa
- Novoheart, Irvine, CA 92617, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Deborah K. Lieu
- Novoheart, Irvine, CA 92617, USA
- Institute for Regenerative Cures and Stem Cell Program, University of California, Davis, Sacramento, CA 95817, USA
| | - Wendy Keung
- Novoheart, Irvine, CA 92617, USA
- Dr. Li Dak Sum Research Centre, The University of Hong Kong, Hong Kong SAR, China
| | | |
Collapse
|
5
|
Lin Y, Zhang F, Chen S, Zhu X, Jiao J, Zhang Y, Li Z, Lin J, Ma B, Chen M, Wang PY, Cui C. Binary Colloidal Crystals Promote Cardiac Differentiation of Human Pluripotent Stem Cells via Nuclear Accumulation of SETDB1. ACS NANO 2023; 17:3181-3193. [PMID: 36655945 PMCID: PMC9933589 DOI: 10.1021/acsnano.3c00009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 01/17/2023] [Indexed: 06/17/2023]
Abstract
Biophysical cues can facilitate the cardiac differentiation of human pluripotent stem cells (hPSCs), yet the mechanism is far from established. One of the binary colloidal crystals, composed of 5 μm Si and 400 nm poly(methyl methacrylate) particles named 5PM, has been applied as a substrate for hPSCs cultivation and cardiac differentiation. In this study, cell nucleus, cytoskeleton, and epigenetic states of human induced pluripotent stem cells on the 5PM were analyzed using atomic force microscopy, molecular biology assays, and the assay for transposase-accessible chromatin sequencing (ATAC-seq). Cells were more spherical with stiffer cell nuclei on the 5PM compared to the flat control. ATAC-seq revealed that chromatin accessibility decreased on the 5PM, caused by the increased entry of histone lysine methyltransferase SETDB1 into the cell nuclei and the amplified level of histone H3K9me3 modification. Reducing cytoskeleton tension using a ROCK inhibitor attenuated the nuclear accumulation of SETDB1 on the 5PM, indicating that the effect is cytoskeleton-dependent. In addition, the knockdown of SETDB1 reversed the promotive effects of the 5PM on cardiac differentiation, demonstrating that biophysical cue-induced cytoskeletal tension, cell nucleus deformation, and then SETDB1 accumulation are critical outside-in signal transformations in cardiac differentiation. Human embryonic stem cells showed similar results, indicating that the biophysical impact of the 5PM surfaces on cardiac differentiation could be universal. These findings contribute to our understanding of material-assistant hPSC differentiation, which benefits materiobiology and stem cell bioengineering.
Collapse
Affiliation(s)
- Yongping Lin
- Department
of Cardiology, The First Affiliated Hospital
of Nanjing Medical University, Nanjing210000, Jiangsu, China
- Department
of Cardiology, The Affiliated Taizhou People’s Hospital of
Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou225300, Jiangsu, China
| | - Feng Zhang
- Department
of Cardiology, The First Affiliated Hospital
of Nanjing Medical University, Nanjing210000, Jiangsu, China
| | - Shaojie Chen
- Department
of Cardiology, The First Affiliated Hospital
of Nanjing Medical University, Nanjing210000, Jiangsu, China
| | - Xiyu Zhu
- Department
of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical
School, Nanjing210000, China
| | - Jincheng Jiao
- State
Key Laboratory of Bioelectronics, School of Biological Science and
Medical Engineering, Southeast University, Nanjing210000, Jiangsu, China
| | - Yike Zhang
- Department
of Cardiology, The First Affiliated Hospital
of Nanjing Medical University, Nanjing210000, Jiangsu, China
| | - Zhaomin Li
- Department
of Cardiology, The First Affiliated Hospital
of Nanjing Medical University, Nanjing210000, Jiangsu, China
| | - Jiao Lin
- Shenzhen
Key Laboratory of Biomimetic Materials and Cellular Immunomodulation,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen518000, Guangdong, China
| | - Biao Ma
- State
Key Laboratory of Bioelectronics, School of Biological Science and
Medical Engineering, Southeast University, Nanjing210000, Jiangsu, China
| | - Minglong Chen
- Department
of Cardiology, The First Affiliated Hospital
of Nanjing Medical University, Nanjing210000, Jiangsu, China
- Department
of Cardiology, The Affiliated Taizhou People’s Hospital of
Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou225300, Jiangsu, China
- Key
Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative
Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing210000, Jiangsu, China
| | - Peng-Yuan Wang
- Oujiang
Laboratory; Key Laboratory of Alzheimer’s Disease of Zhejiang
Province, Institute of Aging, Wenzhou Medical
University, Wenzhou325000, Zhejiang, China
| | - Chang Cui
- Department
of Cardiology, The First Affiliated Hospital
of Nanjing Medical University, Nanjing210000, Jiangsu, China
| |
Collapse
|
6
|
Gurung B, Tse G, Keung W, Li RA, Wong WT. Arrhythmic Risk Assessment of Hypokalaemia Using Human Pluripotent Stem Cell-Derived Cardiac Anisotropic Sheets. Front Cell Dev Biol 2021; 9:681665. [PMID: 34938727 PMCID: PMC8685904 DOI: 10.3389/fcell.2021.681665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 11/01/2021] [Indexed: 11/27/2022] Open
Abstract
Introduction: Hypokalaemia, defined as an extracellular concentration of K+ below 3.5 mM, can cause cardiac arrhythmias by triggered or re-entrant mechanisms. Whilst these effects have been reported in animal and human stem cell-based models, to date there has been no investigation in more complex structures such as the human ventricular cardiac anisotropic sheet (hvCAS). Here, we investigated arrhythmogenicity, electrophysiological, and calcium transient (CaT) changes induced by hypokalaemia using this bioengineered platform. Methods: An optical mapping technique was applied on hvCAS derived from human pluripotent stem cells to visualize electrophysiological and CaT changes under normokalaemic (5 mM KCl) and hypokalaemic (3 mM KCl) conditions. Results: Hypokalaemia significantly increased the proportion of preparations showing spontaneous arrhythmias from 0/14 to 7/14 (Fisher’s exact test, p = 0.003). Hypokalaemia reduced longitudinal conduction velocity (CV) from 7.81 to 7.18 cm⋅s−1 (n = 9, 7; p = 0.036), transverse CV from 5.72 to 4.69 cm⋅s−1 (n = 12, 11; p = 0.030), prolonged action potential at 90% repolarization (APD90) from 83.46 to 97.45 ms (n = 13, 15; p < 0.001), increased action potential amplitude from 0.888 to 1.195 ΔF (n = 12, 14; p < 0.001) and CaT amplitude from 0.76 to 1.37 ΔF (n = 12, 13; p < 0.001), and shortened effective refractory periods from 242 to 165 ms (n = 12, 13; p < 0.001). Conclusion: Hypokalaemia exerts pro-arrhythmic effects on hvCAS, which are associated with alterations in CV, repolarization, refractoriness, and calcium handling. These preparations provide a useful platform for investigating electrophysiological substrates and for conducting arrhythmia screening.
Collapse
Affiliation(s)
- Bimal Gurung
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Gary Tse
- Cardiac Electrophysiology Unit, Cardiovascular Analytics Group, China-UK Collaboration, Hong Kong SAR, China.,Kent and Medway Medical School, Canterbury, Kent, United Kingdom.,Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Wendy Keung
- Novoheart, Irvine, CA, United States.,Dr. Li Dak-Sum Research Centre, HKU-Karolinska Institutet Collaboration in Regenerative Medicine, The University of Hong Kong, Hong Kong SAR, China
| | | | - Wing Tak Wong
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.,State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
7
|
Esfahani SN, Resto Irizarry AM, Xue X, Lee SBD, Shao Y, Fu J. Micro/nanoengineered technologies for human pluripotent stem cells maintenance and differentiation. NANO TODAY 2021; 41:101310. [PMID: 34745321 PMCID: PMC8570530 DOI: 10.1016/j.nantod.2021.101310] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Human pluripotent stem cells (hPSCs) are a promising source of cells for cell replacement-based therapies as well as modeling human development and diseases in vitro. However, achieving fate control of hPSC with a high yield and specificity remains challenging. The fate specification of hPSCs is regulated by biochemical and biomechanical cues in their environment. Driven by this knowledge, recent exciting advances in micro/nanoengineering have been leveraged to develop a broad range of tools for the generation of extracellular biomechanical and biochemical signals that determine the behavior of hPSCs. In this review, we summarize such micro/nanoengineered technologies for controlling hPSC fate and highlight the role of biochemical and biomechanical cues such as substrate rigidity, surface topography, and cellular confinement in the hPSC-based technologies that are on the horizon.
Collapse
Affiliation(s)
- Sajedeh Nasr Esfahani
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Xufeng Xue
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Samuel Byung-Deuk Lee
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yue Shao
- Department of Engineering Mechanics, Tsinghua University, Beijing, China
| | - Jiangping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
8
|
He W, Ye X, Cui T. Progress of shrink polymer micro- and nanomanufacturing. MICROSYSTEMS & NANOENGINEERING 2021; 7:88. [PMID: 34790360 PMCID: PMC8566528 DOI: 10.1038/s41378-021-00312-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/29/2021] [Accepted: 09/16/2021] [Indexed: 05/31/2023]
Abstract
Traditional lithography plays a significant role in the fabrication of micro- and nanostructures. Nevertheless, the fabrication process still suffers from the limitations of manufacturing devices with a high aspect ratio or three-dimensional structure. Recent findings have revealed that shrink polymers attain a certain potential in micro- and nanostructure manufacturing. This technique, denoted as heat-induced shrink lithography, exhibits inherent merits, including an improved fabrication resolution by shrinking, controllable shrinkage behavior, and surface wrinkles, and an efficient fabrication process. These merits unfold new avenues, compensating for the shortcomings of traditional technologies. Manufacturing using shrink polymers is investigated in regard to its mechanism and applications. This review classifies typical applications of shrink polymers in micro- and nanostructures into the size-contraction feature and surface wrinkles. Additionally, corresponding shrinkage mechanisms and models for shrinkage, and wrinkle parameter control are examined. Regarding the size-contraction feature, this paper summarizes the progress on high-aspect-ratio devices, microchannels, self-folding structures, optical antenna arrays, and nanowires. Regarding surface wrinkles, this paper evaluates the development of wearable sensors, electrochemical sensors, energy-conversion technology, cell-alignment structures, and antibacterial surfaces. Finally, the limitations and prospects of shrink lithography are analyzed.
Collapse
Affiliation(s)
- Wenzheng He
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instruments, Tsinghua University, Beijing, 100084 China
| | - Xiongying Ye
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instruments, Tsinghua University, Beijing, 100084 China
| | - Tianhong Cui
- Department of Mechanical Engineering, University of Minnesota, 111 Church Street S.E., Minneapolis, MN 55455 USA
| |
Collapse
|
9
|
Vinje JB, Guadagno NA, Progida C, Sikorski P. Analysis of Actin and Focal Adhesion Organisation in U2OS Cells on Polymer Nanostructures. NANOSCALE RESEARCH LETTERS 2021; 16:143. [PMID: 34524556 PMCID: PMC8443752 DOI: 10.1186/s11671-021-03598-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/28/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND In this work, we explore how U2OS cells are affected by arrays of polymer nanopillars fabricated on flat glass surfaces. We focus on describing changes to the organisation of the actin cytoskeleton and in the location, number and shape of focal adhesions. From our findings we identify that the cells can be categorised into different regimes based on their spreading and adhesion behaviour on nanopillars. A quantitative analysis suggests that cells seeded on dense nanopillar arrays are suspended on top of the pillars with focal adhesions forming closer to the cell periphery compared to flat surfaces or sparse pillar arrays. This change is analogous to similar responses for cells seeded on soft substrates. RESULTS In this work, we explore how U2OS cells are affected by arrays of polymer nanopillars fabricated on flat glass surfaces. We focus on describing changes to the organisation of the actin cytoskeleton and in the location, number and shape of focal adhesions. From our findings we identify that the cells can be categorised into different regimes based on their spreading and adhesion behaviour on nanopillars. A quantitative analysis suggests that cells seeded on dense nanopillar arrays are suspended on top of the pillars with focal adhesions forming closer to the cell periphery compared to flat surfaces or sparse pillar arrays. This change is analogous to similar responses for cells seeded on soft substrates. CONCLUSION Overall, we show that the combination of high throughput nanofabrication, advanced optical microscopy, molecular biology tools to visualise cellular processes and data analysis can be used to investigate how cells interact with nanostructured surfaces and will in the future help to create culture substrates that induce particular cell function.
Collapse
Affiliation(s)
- Jakob B Vinje
- Department of Physics, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
- Department of Electronic Systems, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| | | | - Cinzia Progida
- Department of Biosciences, University of Oslo (UiO), Oslo, Norway
| | - Pawel Sikorski
- Department of Physics, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
10
|
Zhang XD, Thai PN, Lieu DK, Chiamvimonvat N. Model Systems for Addressing Mechanism of Arrhythmogenesis in Cardiac Repair. Curr Cardiol Rep 2021; 23:72. [PMID: 34050853 PMCID: PMC8164614 DOI: 10.1007/s11886-021-01498-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/09/2021] [Indexed: 11/09/2022]
Abstract
PURPOSE OF REVIEW Cardiac cell-based therapy represents a promising approach for cardiac repair. However, one of the main challenges is cardiac arrhythmias associated with stem cell transplantation. The current review summarizes the recent progress in model systems for addressing mechanisms of arrhythmogenesis in cardiac repair. RECENT FINDINGS Animal models have been extensively developed for mechanistic studies of cardiac arrhythmogenesis. Advances in human induced pluripotent stem cells (hiPSCs), patient-specific disease models, tissue engineering, and gene editing have greatly enhanced our ability to probe the mechanistic bases of cardiac arrhythmias. Additionally, recent development in multiscale computational studies and machine learning provides yet another powerful tool to quantitatively decipher the mechanisms of cardiac arrhythmias. Advancing efforts towards the integrations of experimental and computational studies are critical to gain insights into novel mitigation strategies for cardiac arrhythmias in cell-based therapy.
Collapse
Affiliation(s)
- Xiao-Dong Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA 95616 USA
- Department of Veterans Affairs, Veterans Affairs Northern California Health Care System, Mather, CA 95655 USA
| | - Phung N. Thai
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA 95616 USA
- Department of Veterans Affairs, Veterans Affairs Northern California Health Care System, Mather, CA 95655 USA
| | - Deborah K. Lieu
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA 95616 USA
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA 95616 USA
- Department of Veterans Affairs, Veterans Affairs Northern California Health Care System, Mather, CA 95655 USA
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA 95616 USA
| |
Collapse
|
11
|
Liu T, Zhang S, Huang C, Ma S, Bai R, Li Y, Chang Y, Hang C, Saleem A, Dong T, Guo T, Jiang Y, Lu W, Zhang L, Jianwen L, Jiang H, Lan F. Microscale grooves regulate maturation development of hPSC-CMs by the transient receptor potential channels (TRP channels). J Cell Mol Med 2021; 25:3469-3483. [PMID: 33689230 PMCID: PMC8034460 DOI: 10.1111/jcmm.16429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 02/07/2021] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
The use of human pluripotent stem cell‐derived cardiomyocytes (hPSC‐CMs) is limited in drug discovery and cardiac disease mechanism studies due to cell immaturity. Micro‐scaled grooves can promote the maturation of cardiomyocytes by aligning them in order, but the mechanism of cardiomyocytes alignment has not been studied. From the level of calcium activity, gene expression and cell morphology, we verified that the W20H5 grooves can effectively promote the maturation of cardiomyocytes. The transient receptor potential channels (TRP channels) also play an important role in the maturation and development of cardiomyocytes. These findings support the engineered hPSC‐CMs as a powerful model to study cardiac disease mechanism and partly mimic the myocardial morphological development. The important role of the TRP channels in the maturation and development of myocardium is first revealed.
Collapse
Affiliation(s)
- Taoyan Liu
- Beijing Lab for Cardiovascular Precision Medicine, Anzhen Hospital, Capital Medical University, Beijing, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Siyao Zhang
- Beijing Lab for Cardiovascular Precision Medicine, Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chenwu Huang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Shuhong Ma
- Beijing Lab for Cardiovascular Precision Medicine, Anzhen Hospital, Capital Medical University, Beijing, China
| | - Rui Bai
- Beijing Lab for Cardiovascular Precision Medicine, Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yanan Li
- Beijing Lab for Cardiovascular Precision Medicine, Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yun Chang
- Beijing Lab for Cardiovascular Precision Medicine, Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chenwen Hang
- Department of Cardiology, Peking University Third Hospital, Beijing, China
| | - Amina Saleem
- Beijing Lab for Cardiovascular Precision Medicine, Anzhen Hospital, Capital Medical University, Beijing, China
| | - Tao Dong
- Beijing Lab for Cardiovascular Precision Medicine, Anzhen Hospital, Capital Medical University, Beijing, China
| | - Tianwei Guo
- Beijing Lab for Cardiovascular Precision Medicine, Anzhen Hospital, Capital Medical University, Beijing, China
| | - Youxu Jiang
- Beijing Lab for Cardiovascular Precision Medicine, Anzhen Hospital, Capital Medical University, Beijing, China
| | - Wenjing Lu
- Beijing Lab for Cardiovascular Precision Medicine, Anzhen Hospital, Capital Medical University, Beijing, China
| | - Lina Zhang
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China
| | - Luo Jianwen
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Hongfeng Jiang
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Feng Lan
- Beijing Lab for Cardiovascular Precision Medicine, Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
12
|
Wong AOT, Gurung B, Wong WS, Mak SY, Tse WW, Li CM, Lieu DK, Costa KD, Li RA, Hajjar RJ. Adverse effects of hydroxychloroquine and azithromycin on contractility and arrhythmogenicity revealed by human engineered cardiac tissues. J Mol Cell Cardiol 2020; 153:106-110. [PMID: 33373642 PMCID: PMC7765761 DOI: 10.1016/j.yjmcc.2020.12.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/10/2020] [Accepted: 12/22/2020] [Indexed: 12/18/2022]
Abstract
The coronavirus disease 2019 (COVID-19) outbreak caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a global pandemic as declared by World Health Organization (WHO). In the absence of an effective treatment, different drugs with unknown effectiveness, including antimalarial hydroxychloroquine (HCQ), with or without concurrent administration with azithromycin (AZM), have been tested for treating COVID-19 patients with developed pneumonia. However, the efficacy and safety of HCQ and/or AZM have been questioned by recent clinical reports. Direct effects of these drugs on the human heart remain very poorly defined. To better understand the mechanisms of action of HCQ +/− AZM, we employed bioengineered human ventricular cardiac tissue strip (hvCTS) and anisotropic sheet (hvCAS) assays, made with human pluripotent stem cell (hPSC)-derived ventricular cardiomyocytes (hvCMs), which have been designed for measuring cardiac contractility and electrophysiology, respectively. Our hvCTS experiments showed that AZM induced a dose-dependent negative inotropic effect which could be aggravated by HCQ; electrophysiologically, as revealed by the hvCAS platform, AZM prolonged action potentials and induced spiral wave formations. Collectively, our data were consistent with reported clinical risks of HCQ and AZM on QTc prolongation/ventricular arrhythmias and development of heart failure. In conclusion, our study exposed the risks of HCQ/AZM administration while providing mechanistic insights for their toxicity. Our bioengineered human cardiac tissue constructs therefore provide a useful platform for screening cardiac safety and efficacy when developing therapeutics against COVID-19.
Collapse
Affiliation(s)
| | - Bimal Gurung
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | | | | | - Wan Wai Tse
- Novoheart, Irvine, California, United States
| | - Chloe M Li
- German Swiss International School, The Peak, Hong Kong
| | - Deborah K Lieu
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, California, United States
| | - Kevin D Costa
- Novoheart, Irvine, California, United States; Icahn School of Medicine at Mount Sinai, Manhattan, New York, United States
| | - Ronald A Li
- Novoheart, Irvine, California, United States; Dr. Li Dak-Sum Research Centre, The University of Hong Kong - Karolinska Institutet Collaboration in Regenerative Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong; Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong; Sardocor, Boston, Massachusetts, United States.
| | | |
Collapse
|
13
|
Coyle RC, Barrs RW, Richards DJ, Ladd EP, Menick DR, Mei Y. Targeting HIF-α for robust prevascularization of human cardiac organoids. J Tissue Eng Regen Med 2020; 15:189-202. [PMID: 33868541 DOI: 10.1002/term.3165] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prevascularized 3D microtissues have been shown to be an effective cell delivery vehicle for cardiac repair. To this end, our lab has explored the development of self-organizing, prevascularized human cardiac organoids by co-seeding human cardiomyocytes with cardiac fibroblasts, endothelial cells, and stromal cells into agarose microwells. We hypothesized that this prevascularization process is facilitated by the endogenous upregulation of hypoxia-inducible factor (HIF) pathway in the avascular 3D microtissues. In this study, we used Molidustat, a selective PHD (prolyl hydroxylase domain enzymes) inhibitor that stabilizes HIF-α, to treat human cardiac organoids, which resulted in 150 ± 61% improvement in endothelial expression (CD31) and 220 ± 20% improvement in the number of lumens per organoids. We hypothesized that the improved endothelial expression seen in Molidustat treated human cardiac organoids was dependent upon upregulation of VEGF, a well-known downstream target of HIF pathway. Through the use of immunofluorescent staining and ELISA assays, we determined that Molidustat treatment improved VEGF expression of non-endothelial cells and resulted in improved co-localization of supporting cell types and endothelial structures. We further demonstrated that Molidustat treated human cardiac organoids maintain cardiac functionality. Lastly, we showed that Molidustat treatment improves survival of cardiac organoids when exposed to both hypoxic and ischemic conditions in vitro. For the first time, we demonstrate that targeted HIF-α stabilization provides a robust strategy to improve endothelial expression and lumen formation in cardiac microtissues, which will provide a powerful framework for prevascularization of various microtissues in developing successful cell transplantation therapies.
Collapse
Affiliation(s)
- Robert C Coyle
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Ryan W Barrs
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Dylan J Richards
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Emma P Ladd
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Donald R Menick
- Ralph H. Johnson Veterans Affairs Medical Center, Medical University of South Carolina, Charleston, SC 29425, USA.,Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston SC 29425, USA
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA.,Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
14
|
Lam YY, Keung W, Chan CH, Geng L, Wong N, Brenière-Letuffe D, Li RA, Cheung YF. Single-Cell Transcriptomics of Engineered Cardiac Tissues From Patient-Specific Induced Pluripotent Stem Cell-Derived Cardiomyocytes Reveals Abnormal Developmental Trajectory and Intrinsic Contractile Defects in Hypoplastic Right Heart Syndrome. J Am Heart Assoc 2020; 9:e016528. [PMID: 33059525 PMCID: PMC7763394 DOI: 10.1161/jaha.120.016528] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background To understand the intrinsic cardiac developmental and functional abnormalities in pulmonary atresia with intact ventricular septum (PAIVS) free from effects secondary to anatomic defects, we performed and compared single‐cell transcriptomic and phenotypic analyses of patient‐ and healthy subject–derived human‐induced pluripotent stem cell–derived cardiomyocytes (hiPSC‐CMs) and engineered tissue models. Methods and Results We derived hiPSC lines from 3 patients with PAIVS and 3 healthy subjects and differentiated them into hiPSC‐CMs, which were then bioengineered into the human cardiac anisotropic sheet and human cardiac tissue strip custom‐designed for electrophysiological and contractile assessments, respectively. Single‐cell RNA sequencing (scRNA‐seq) of hiPSC‐CMs, human cardiac anisotropic sheet, and human cardiac tissue strip was performed to examine the transcriptomic basis for any phenotypic abnormalities using pseudotime and differential expression analyses. Through pseudotime analysis, we demonstrated that bioengineered tissue constructs provide pro‐maturational cues to hiPSC‐CMs, although the maturation and development were attenuated in PAIVS hiPSC‐CMs. Furthermore, reduced contractility and prolonged contractile kinetics were observed with PAIVS human cardiac tissue strips. Consistently, single‐cell RNA sequencing of PAIVS human cardiac tissue strips and hiPSC‐CMs exhibited diminished expression of cardiac contractile apparatus genes. By contrast, electrophysiological aberrancies were absent in PAIVS human cardiac anisotropic sheets. Conclusions Our findings were the first to reveal intrinsic abnormalities of cardiomyocyte development and function in PAIVS free from secondary effects. We conclude that hiPSC‐derived engineered tissues offer a unique method for studying primary cardiac abnormalities and uncovering pathogenic mechanisms that underlie sporadic congenital heart diseases.
Collapse
Affiliation(s)
- Yin-Yu Lam
- Department of Paediatrics and Adolescent Medicine Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR
| | - Wendy Keung
- Dr. Li Dak-Sum Research Centre HKU - KI Collaboration in Regenerative Medicine The University of Hong Kong Hong Kong SAR.,Ming-Wai Lau Centre for Reparative Medicine Karolinska Insititutet Hong Kong
| | - Chun-Ho Chan
- Department of Paediatrics and Adolescent Medicine Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR
| | - Lin Geng
- Dr. Li Dak-Sum Research Centre HKU - KI Collaboration in Regenerative Medicine The University of Hong Kong Hong Kong SAR
| | - Nicodemus Wong
- Department of Paediatrics and Adolescent Medicine Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR
| | | | - Ronald A Li
- Department of Paediatrics and Adolescent Medicine Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR.,Dr. Li Dak-Sum Research Centre HKU - KI Collaboration in Regenerative Medicine The University of Hong Kong Hong Kong SAR.,Ming-Wai Lau Centre for Reparative Medicine Karolinska Insititutet Hong Kong
| | - Yiu-Fai Cheung
- Department of Paediatrics and Adolescent Medicine Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR.,Dr. Li Dak-Sum Research Centre HKU - KI Collaboration in Regenerative Medicine The University of Hong Kong Hong Kong SAR.,Ming-Wai Lau Centre for Reparative Medicine Karolinska Insititutet Hong Kong
| |
Collapse
|
15
|
Modeling the heart with Novoheart’s MyHeart™ platform. FUTURE DRUG DISCOVERY 2020. [DOI: 10.4155/fdd-2020-0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Reliable and predictive human-specific in vitro heart models can revolutionize drug discovery and development. With the advent of pluripotent stem cell technologies, human cardiomyocytes can now be readily produced in large quantities. Using tissue engineering techniques, they can be further assembled into cardiac tissues of specific 2D and 3D configurations, to create models that behave and function like the native human heart. Novoheart (BC, Canada) uniquely offers the MyHeartTM Platform of bioengineered human heart constructs, designed to provide researchers with effective models of either healthy or diseased human hearts. As in vitro, human-based assays become more widely accepted, the next decade could witness a shift away from animal testing towards more accurate and scalable human assays like the MyHeartTM Platform.
Collapse
|
16
|
Wong AOT, Wong N, Geng L, Chow MZY, Lee EK, Wu H, Khine M, Kong CW, Costa KD, Keung W, Cheung YF, Li RA. Combinatorial Treatment of Human Cardiac Engineered Tissues With Biomimetic Cues Induces Functional Maturation as Revealed by Optical Mapping of Action Potentials and Calcium Transients. Front Physiol 2020; 11:165. [PMID: 32226389 PMCID: PMC7080659 DOI: 10.3389/fphys.2020.00165] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 02/12/2020] [Indexed: 01/16/2023] Open
Abstract
Although biomimetic stimuli, such as microgroove-induced alignment (μ), triiodothyronine (T3) induction, and electrical conditioning (EC), have been reported to promote maturation of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs), a systematic examination of their combinatorial effects on engineered cardiac tissue constructs and the underlying molecular pathways has not been reported. Herein, human embryonic stem cell-derived ventricular cardiomyocytes (hESC-VCMs) were used to generate a micro-patterned human ventricular cardiac anisotropic sheets (hvCAS) for studying the physiological effects of combinatorial treatments by a range of functional, calcium (Ca2+)-handling, and molecular analyses. High-resolution optical mapping showed that combined μ-T3-EC treatment of hvCAS increased the conduction velocity, anisotropic ratio, and proportion of mature quiescent-yet-excitable preparations by 2. 3-, 1. 8-, and 5-fold (>70%), respectively. Such electrophysiological changes could be attributed to an increase in inward sodium current density and a decrease in funny current densities, which is consistent with the observed up- and downregulated SCN1B and HCN2/4 transcripts, respectively. Furthermore, Ca2+-handling transcripts encoding for phospholamban (PLN) and sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) were upregulated, and this led to faster upstroke and decay kinetics of Ca2+-transients. RNA-sequencing and pathway mapping of T3-EC-treated hvCAS revealed that the TGF-β signaling was downregulated; the TGF-β receptor agonist and antagonist TGF-β1 and SB431542 partially reversed T3-EC induced quiescence and reduced spontaneous contractions, respectively. Taken together, we concluded that topographical cues alone primed cardiac tissue constructs for augmented electrophysiological and calcium handling by T3-EC. Not only do these studies improve our understanding of hPSC-CM biology, but the orchestration of these pro-maturational factors also improves the use of engineered cardiac tissues for in vitro drug screening and disease modeling.
Collapse
Affiliation(s)
- Andy On-Tik Wong
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Nicodemus Wong
- Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Dr. Li Dak-Sum Research Centre, The University of Hong Kong - Karolinska Institutet Collaboration in Regenerative Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Lin Geng
- Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Dr. Li Dak-Sum Research Centre, The University of Hong Kong - Karolinska Institutet Collaboration in Regenerative Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Maggie Zi-Ying Chow
- Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Eugene K Lee
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| | - Hongkai Wu
- Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong
| | - Michelle Khine
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| | - Chi-Wing Kong
- Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Kevin D Costa
- Icahn School of Medicine at Mount Sinai, Manhattan, NY, United States
| | - Wendy Keung
- Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Dr. Li Dak-Sum Research Centre, The University of Hong Kong - Karolinska Institutet Collaboration in Regenerative Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Yiu-Fai Cheung
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Ronald A Li
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Dr. Li Dak-Sum Research Centre, The University of Hong Kong - Karolinska Institutet Collaboration in Regenerative Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Ming-Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
17
|
Guan X, Xu W, Zhang H, Wang Q, Yu J, Zhang R, Chen Y, Xia Y, Wang J, Wang D. Transplantation of human induced pluripotent stem cell-derived cardiomyocytes improves myocardial function and reverses ventricular remodeling in infarcted rat hearts. Stem Cell Res Ther 2020; 11:73. [PMID: 32085809 PMCID: PMC7033912 DOI: 10.1186/s13287-020-01602-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/21/2020] [Accepted: 02/12/2020] [Indexed: 12/19/2022] Open
Abstract
Background Human-induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) have shed great light on cardiac regenerative medicine and specifically myocardial repair in heart failure patients. However, the treatment efficacy and the survival of iPSC-CMs in vivo after transplantation have yielded inconsistent results. Objectives The objective of this study was to evaluate the ability of human iPSC-CMs to improve myocardial function in a rat postinfarction heart failure model. Methods Eight-week-old male Sprague-Dawley rats were randomly selected to receive an intramyocardial injection of 5% albumin solution with or without 1 × 107 human iPSC-CMs 10 days after undergoing left anterior descending (LAD) coronary artery ligation. Cyclosporine A and methylprednisolone were administered before iPSC-CM injection and until the rats were killed to prevent graft rejection. Cardiac function was evaluated by echocardiography. The survival of grafted cardiomyocytes was confirmed by observing the fluorescent cell tracer Vybrant™ CM-DiI or expression of the enhanced green fluorescent protein (eGFP) in transplanted cells, or survival was demonstrated by polymerase chain reaction (PCR)-based detection of human mitochondrial DNA. Sirius red stain was used to evaluate the fibrosis ratio. Hematoxylin-eosin staining was used to observe the formation of teratomas. Results Four weeks after intramyocardial injection of iPSC-CMs, animals undergoing iPSC-CM transplantation had lower mortality than the control group. Animals injected with cell-free solution (control group) demonstrated significant left ventricular (LV) functional deterioration, whereas grafting of iPSC-CMs attenuated this remodeling process. In the control group, the ejection fraction deteriorated by 10.11% (from 46.36 to 41.67%), and fractional shortening deteriorated by 9.23% (from 24.37 to 22.12%) by 4 weeks. In the iPSC-CM injection group, the ejection fraction improved by 18.86% (from 44.09 to 52.41%), and fractional shortening improved by 23.69% (from 23.08 to 28.54%). Cell labeling, tracking, and molecular biology techniques indicated that the grafted cardiomyocytes survived in the rat heart 1 month after iPSC-CM transplantation. Myocardial fibrosis was also attenuated in the iPSC-CM treatment group. Conclusions Human iPSC-CM grafts survived in infarcted rat hearts and restored myocardial function 4 weeks after transplantation. Cell replacement therapy also reversed ventricular remodeling, indicating the potential of iPSC-CMs for cardiac repair strategies. Electronic supplementary material The online version of this article (10.1186/s13287-020-01602-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xumin Guan
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Wanzi Xu
- Department of Thoracic and Cardiovascular Surgery, Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China
| | - He Zhang
- Department of Thoracic and Cardiovascular Surgery, Peking Union Medical College Nanjing Drum Tower Hospital, Nanjing, 210008, Jiangsu, China
| | - Qian Wang
- HELP Therapeutics, Nanjing, 211166, Jiangsu, China
| | - Jiuyang Yu
- HELP Therapeutics, Nanjing, 211166, Jiangsu, China
| | - Ruyi Zhang
- The Laboratory Animal Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yamin Chen
- HELP Therapeutics, Nanjing, 211166, Jiangsu, China
| | - Yunlong Xia
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Jiaxian Wang
- HELP Therapeutics, Nanjing, 211166, Jiangsu, China. .,Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Dongjin Wang
- Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
18
|
Schwach V, Passier R. Native cardiac environment and its impact on engineering cardiac tissue. Biomater Sci 2020; 7:3566-3580. [PMID: 31338495 DOI: 10.1039/c8bm01348a] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) generally have an immature fetal-like phenotype when directly compared to isolated CMs from human hearts, despite significant advance in differentiation of human pluripotent stem cells (hPSCs) to multiple cardiac lineages. Therefore, hPSC-CMs may not accurately mimic all facets of healthy and diseased human adult CMs. During embryonic development, the cardiac extracellular matrix (ECM) experiences a gradual assembly of matrix proteins that transits along the maturation of CMs. Mimicking these dynamic stages may contribute to hPSC-CMs maturation in vitro. Thus, in this review, we describe the progressive build-up of the cardiac ECM during embryonic development, the ECM of the adult human heart and the application of natural and synthetic biomaterials for cardiac tissue engineering with hPSC-CMs.
Collapse
Affiliation(s)
- Verena Schwach
- Dept of Applied Stem Cell Technologies, TechMed Centre, University of Twente, The Netherlands.
| | | |
Collapse
|
19
|
Sung TC, Liu CH, Huang WL, Lee YC, Kumar SS, Chang Y, Ling QD, Hsu ST, Higuchi A. Efficient differentiation of human ES and iPS cells into cardiomyocytes on biomaterials under xeno-free conditions. Biomater Sci 2019; 7:5467-5481. [PMID: 31656967 DOI: 10.1039/c9bm00817a] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Current xeno-free and chemically defined methods for the differentiation of hPSCs (human pluripotent stem cells) into cardiomyocytes are not efficient and are sometimes not reproducible. Therefore, it is necessary to develop reliable and efficient methods for the differentiation of hPSCs into cardiomyocytes for future use in cardiovascular research related to drug discovery, cardiotoxicity screening, and disease modeling. We evaluated two representative differentiation methods that were reported previously, and we further developed original, more efficient methods for the differentiation of hPSCs into cardiomyocytes under xeno-free, chemically defined conditions. The developed protocol successively differentiated hPSCs into cardiomyocytes, approximately 90-97% of which expressed the cardiac marker cTnT, with beating speeds and sarcomere lengths that were similar to those of a healthy adult human heart. The optimal cell culture biomaterials for the cardiac differentiation of hPSCs were also evaluated using extracellular matrix-mimetic material-coated dishes. Synthemax II-coated and Laminin-521-coated dishes were found to be the most effective and efficient biomaterials for the cardiac differentiation of hPSCs according to the observation of hPSC-derived cardiomyocytes with high survival ratios, high beating colony numbers, a similar beating frequency to that of a healthy adult human heart, high purity levels (high cTnT expression) and longer sarcomere lengths similar to those of a healthy adult human heart.
Collapse
Affiliation(s)
- Tzu-Cheng Sung
- The Eye Hospital of Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang 325027, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Ribeiro AJS, Guth BD, Engwall M, Eldridge S, Foley CM, Guo L, Gintant G, Koerner J, Parish ST, Pierson JB, Brock M, Chaudhary KW, Kanda Y, Berridge B. Considerations for an In Vitro, Cell-Based Testing Platform for Detection of Drug-Induced Inotropic Effects in Early Drug Development. Part 2: Designing and Fabricating Microsystems for Assaying Cardiac Contractility With Physiological Relevance Using Human iPSC-Cardiomyocytes. Front Pharmacol 2019; 10:934. [PMID: 31555128 PMCID: PMC6727630 DOI: 10.3389/fphar.2019.00934] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 07/22/2019] [Indexed: 12/14/2022] Open
Abstract
Contractility of the myocardium engines the pumping function of the heart and is enabled by the collective contractile activity of its muscle cells: cardiomyocytes. The effects of drugs on the contractility of human cardiomyocytes in vitro can provide mechanistic insight that can support the prediction of clinical cardiac drug effects early in drug development. Cardiomyocytes differentiated from human-induced pluripotent stem cells have high potential for overcoming the current limitations of contractility assays because they attach easily to extracellular materials and last long in culture, while having human- and patient-specific properties. Under these conditions, contractility measurements can be non-destructive and minimally invasive, which allow assaying sub-chronic effects of drugs. For this purpose, the function of cardiomyocytes in vitro must reflect physiological settings, which is not observed in cultured cardiomyocytes derived from induced pluripotent stem cells because of the fetal-like properties of their contractile machinery. Primary cardiomyocytes or tissues of human origin fully represent physiological cellular properties, but are not easily available, do not last long in culture, and do not attach easily to force sensors or mechanical actuators. Microengineered cellular systems with a more mature contractile function have been developed in the last 5 years to overcome this limitation of stem cell-derived cardiomyocytes, while simultaneously measuring contractile endpoints with integrated force sensors/actuators and image-based techniques. Known effects of engineered microenvironments on the maturity of cardiomyocyte contractility have also been discovered in the development of these systems. Based on these discoveries, we review here design criteria of microengineered platforms of cardiomyocytes derived from pluripotent stem cells for measuring contractility with higher physiological relevance. These criteria involve the use of electromechanical, chemical and morphological cues, co-culture of different cell types, and three-dimensional cellular microenvironments. We further discuss the use and the current challenges for developing and improving these novel technologies for predicting clinical effects of drugs based on contractility measurements with cardiomyocytes differentiated from induced pluripotent stem cells. Future research should establish contexts of use in drug development for novel contractility assays with stem cell-derived cardiomyocytes.
Collapse
Affiliation(s)
- Alexandre J S Ribeiro
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Brian D Guth
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany.,PreClinical Drug Development Platform (PCDDP), North-West University, Potchefstroom, South Africa
| | - Michael Engwall
- Safety Pharmacology and Animal Research Center, Amgen Research, Thousand Oaks, CA, United States
| | - Sandy Eldridge
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - C Michael Foley
- Department of Integrative Pharmacology, Integrated Sciences and Technology, AbbVie, North Chicago, IL, United States
| | - Liang Guo
- Laboratory of Investigative Toxicology, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Gary Gintant
- Department of Integrative Pharmacology, Integrated Sciences and Technology, AbbVie, North Chicago, IL, United States
| | - John Koerner
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Stanley T Parish
- Health and Environmental Sciences Institute, Washington, DC, United States
| | - Jennifer B Pierson
- Health and Environmental Sciences Institute, Washington, DC, United States
| | - Mathew Brock
- Department of Safety Assessment, Genentech, South San Francisco, CA, United States
| | - Khuram W Chaudhary
- Global Safety Pharmacology, GlaxoSmithKline plc, Collegeville, PA, United States
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, Kanagawa, Japan
| | - Brian Berridge
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| |
Collapse
|
21
|
Wong AOT, Wong G, Shen M, Chow MZY, Tse WW, Gurung B, Mak SY, Lieu DK, Costa KD, Chan CW, Martelli A, Nabhan JF, Li RA. Correlation between frataxin expression and contractility revealed by in vitro Friedreich's ataxia cardiac tissue models engineered from human pluripotent stem cells. Stem Cell Res Ther 2019; 10:203. [PMID: 31286988 PMCID: PMC6615274 DOI: 10.1186/s13287-019-1305-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/30/2019] [Accepted: 06/17/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Friedreich's ataxia (FRDA) is an autosomal recessive disease caused by a non-coding mutation in the first intron of the frataxin (FXN) gene that suppresses its expression. Compensatory hypertrophic cardiomyopathy, dilated cardiomyopathy, and conduction system abnormalities in FRDA lead to cardiomyocyte (CM) death and fibrosis, consequently resulting in heart failure and arrhythmias. Murine models have been developed to study disease pathology in the past two decades; however, differences between human and mouse physiology and metabolism have limited the relevance of animal studies in cardiac disease conditions. To bridge this gap, we aimed to generate species-specific, functional in vitro experimental models of FRDA using 2-dimensional (2D) and 3-dimensional (3D) engineered cardiac tissues from FXN-deficient human pluripotent stem cell-derived ventricular cardiomyocytes (hPSC-hvCMs) and to compare their contractile and electrophysiological properties with healthy tissue constructs. METHODS Healthy control and FRDA patient-specific hPSC-hvCMs were derived by directed differentiation using a small molecule-based protocol reported previously. We engineered the hvCMs into our established human ventricular cardiac tissue strip (hvCTS) and human ventricular cardiac anisotropic sheet (hvCAS) models, and functional assays were performed on days 7-17 post-tissue fabrication to assess the electrophysiology and contractility of FRDA patient-derived and FXN-knockdown engineered tissues, in comparison with healthy controls. To further validate the disease model, forced expression of FXN was induced in FXN-deficient tissues to test if disease phenotypes could be rescued. RESULTS Here, we report for the first time the generation of human engineered tissue models of FRDA cardiomyopathy from hPSCs: FXN-deficient hvCTS displayed attenuated developed forces (by 70-80%) compared to healthy controls. High-resolution optical mapping of hvCAS with reduced FXN expression also revealed electrophysiological defects consistent with clinical observations, including action potential duration prolongation and maximum capture frequency reduction. Interestingly, a clear positive correlation between FXN expression and contractility was observed (ρ > 0.9), and restoration of FXN protein levels by lentiviral transduction rescued contractility defects in FXN-deficient hvCTS. CONCLUSIONS We conclude that human-based in vitro cardiac tissue models of FRDA provide a translational, disease-relevant biomimetic platform for the evaluation of novel therapeutics and to provide insight into FRDA disease progression.
Collapse
Affiliation(s)
| | - Gabriel Wong
- Novoheart, Vancouver, British Columbia V6C 2V6 Canada
| | - Michael Shen
- Novoheart, Vancouver, British Columbia V6C 2V6 Canada
| | | | - Wan Wai Tse
- Novoheart, Vancouver, British Columbia V6C 2V6 Canada
| | - Bimal Gurung
- Novoheart, Vancouver, British Columbia V6C 2V6 Canada
| | - Suet Yee Mak
- Novoheart, Vancouver, British Columbia V6C 2V6 Canada
| | | | | | - Camie W. Chan
- Novoheart, Vancouver, British Columbia V6C 2V6 Canada
| | - Alain Martelli
- Current address: Astellas Innovation Management Astellas Pharma, 1030 Massachusetts Avenue, Cambridge, MA 02138 USA
| | - Joseph F. Nabhan
- Current address: Astellas Innovation Management Astellas Pharma, 1030 Massachusetts Avenue, Cambridge, MA 02138 USA
| | - Ronald A. Li
- Novoheart, Vancouver, British Columbia V6C 2V6 Canada
| |
Collapse
|
22
|
Blazeski A, Lowenthal J, Zhu R, Ewoldt J, Boheler KR, Tung L. Functional Properties of Engineered Heart Slices Incorporating Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Stem Cell Reports 2019; 12:982-995. [PMID: 31056480 PMCID: PMC6524004 DOI: 10.1016/j.stemcr.2019.04.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 04/02/2019] [Accepted: 04/02/2019] [Indexed: 12/18/2022] Open
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) hold great promise for cardiac studies, but their structural and functional immaturity precludes their use as faithful models of adult myocardium. Here we describe engineered heart slices (EHS), preparations of decellularized porcine myocardium repopulated with hiPSC-CMs that exhibit structural and functional improvements over standard culture. EHS exhibited multicellular, aligned bundles of elongated CMs with organized sarcomeres, positive inotropic responses to isoproterenol, anisotropic conduction of action potentials, and electrophysiological functionality for more than 200 days. We developed a new drug assay, GRIDS, that serves as a "fingerprint" of cardiac drug sensitivity for a range of pacing rates and drug concentrations. GRIDS maps characterized differences in drug sensitivity between EHS and monolayers more clearly than changes in action potential durations or conduction velocities. EHS represent a tissue-like model for long-term culture, structural, and functional improvement, and higher fidelity drug response of hiPSC-CMs.
Collapse
Affiliation(s)
- Adriana Blazeski
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21205, USA
| | - Justin Lowenthal
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21205, USA
| | - Renjun Zhu
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jourdan Ewoldt
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21205, USA
| | - Kenneth R Boheler
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21205, USA; Stem Cell and Regenerative Medicine Consortium, LKS Faculty of Medicine, Hong Kong University, Hong Kong, SAR; Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Leslie Tung
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
23
|
Keung W, Chan PKW, Backeris PC, Lee EK, Wong N, Wong AOT, Wong GKY, Chan CWY, Fermini B, Costa KD, Li RA. Human Cardiac Ventricular-Like Organoid Chambers and Tissue Strips From Pluripotent Stem Cells as a Two-Tiered Assay for Inotropic Responses. Clin Pharmacol Ther 2019; 106:402-414. [PMID: 30723889 DOI: 10.1002/cpt.1385] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 01/18/2019] [Indexed: 12/14/2022]
Abstract
Traditional drug discovery is an inefficient process. Human pluripotent stem cell-derived cardiomyocytes can potentially fill the gap between animal and clinical studies, but conventional two-dimensional cultures inadequately recapitulate the human cardiac phenotype. Here, we systematically examined the pharmacological responses of engineered human ventricular-like cardiac tissue strips (hvCTS) and organoid chambers (hvCOC) to 25 cardioactive compounds covering various drug classes. While hvCTS effectively detected negative and null inotropic effects, the sensitivity to positive inotropes was modest. We further quantified the predictive capacity of hvCTS in a blinded screening, with accuracies for negative, positive, and null inotropic effects at 100%, 86%, and 80%, respectively. Interestingly, hvCOC, with a pro-maturation milieu that yields physiologically complex parameters, displayed enhanced positive inotropy. Based on these results, we propose a two-tiered screening system for avoiding false positives and negatives. Such an approach would facilitate drug discovery by leading to better overall success.
Collapse
Affiliation(s)
- Wendy Keung
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Shatin, Hong Kong.,Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong
| | - Patrick K W Chan
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Shatin, Hong Kong
| | - Peter C Backeris
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York,, USA
| | | | - Nicodemus Wong
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Shatin, Hong Kong
| | | | | | | | - Bernard Fermini
- Global Safety Pharmacology, Pfizer Worldwide Research and Development, Groton, Connecticut, USA
| | - Kevin D Costa
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York,, USA.,Novoheart, Vancouver, British Columbia, Canada
| | - Ronald A Li
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Shatin, Hong Kong.,Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong.,Novoheart, Vancouver, British Columbia, Canada
| |
Collapse
|
24
|
Rodriguez ML, Beussman KM, Chun KS, Walzer MS, Yang X, Murry CE, Sniadecki NJ. Substrate Stiffness, Cell Anisotropy, and Cell-Cell Contact Contribute to Enhanced Structural and Calcium Handling Properties of Human Embryonic Stem Cell-Derived Cardiomyocytes. ACS Biomater Sci Eng 2019; 5:3876-3888. [PMID: 33438427 DOI: 10.1021/acsbiomaterials.8b01256] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) can be utilized to understand the mechanisms underlying the development and progression of heart disease, as well as to develop better interventions and treatments for this disease. However, these cells are structurally and functionally immature, which undermines some of their adequacy in modeling adult heart tissue. Previous studies with immature cardiomyocytes have shown that altering substrate stiffness, cell anisotropy, and/or cell-cell contact can enhance the contractile and structural maturation of hPSC-CMs. In this study, the structural and calcium handling properties of human embryonic stem cell-derived cardiomyocytes (hESC-CMs) were enhanced by exposure to a downselected combination of these three maturation stimuli. First, hESC-CMs were seeded onto substrates composed of two commercial formulations of polydimethylsiloxane (PDMS), Sylgard 184 and Sylgard 527, whose stiffness ranged from 5 kPa to 101 kPa. Upon analyzing the morphological and calcium transient properties of these cells, it was concluded that a 21 kPa substrate yielded cells with the highest degree of maturation. Next, these PDMS substrates were microcontact-printed with laminin to force the cultured cells into rod-shaped geometries using line patterns that were 12, 18, or 24 μm in width. We found that cells on the 18 and 24 μm pattern widths had structural and functional properties that were superior to those on the 12 μm pattern. The hESC-CMs were then seeded onto these line-stamped surfaces at a density of 500 000 cells per 25-mm-diameter substrate, to enable the formation of cell-cell contacts at their distal ends. We discovered that this combination of culture conditions resulted in cells that were more structurally and functionally mature than those that were only exposed to one or two stimuli. Our results suggest that downselecting a combination of mechanobiological stimuli could prove to be an effective means of maturing hPSC-CMs in vitro.
Collapse
Affiliation(s)
- Marita L Rodriguez
- Department of Mechanical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Kevin M Beussman
- Department of Mechanical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Katherine S Chun
- Department of Mechanical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Melissa S Walzer
- Department of Pathology, University of Washington, Seattle, Washington 98195, United States
| | - Xiulan Yang
- Department of Pathology, University of Washington, Seattle, Washington 98195, United States.,Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, United States.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, United States
| | - Charles E Murry
- Department of Pathology, University of Washington, Seattle, Washington 98195, United States.,Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, United States.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, United States.,Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States.,Department of Medicine/Cardiology, University of Washington, Seattle, Washington 98195, United States
| | - Nathan J Sniadecki
- Department of Mechanical Engineering, University of Washington, Seattle, Washington 98195, United States.,Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, United States.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, United States.,Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
25
|
Santoro R, Perrucci GL, Gowran A, Pompilio G. Unchain My Heart: Integrins at the Basis of iPSC Cardiomyocyte Differentiation. Stem Cells Int 2019; 2019:8203950. [PMID: 30906328 PMCID: PMC6393933 DOI: 10.1155/2019/8203950] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/20/2018] [Accepted: 01/10/2019] [Indexed: 02/06/2023] Open
Abstract
The cellular response to the extracellular matrix (ECM) microenvironment mediated by integrin adhesion is of fundamental importance, in both developmental and pathological processes. In particular, mechanotransduction is of growing importance in groundbreaking cellular models such as induced pluripotent stem cells (iPSC), since this process may strongly influence cell fate and, thus, augment the precision of differentiation into specific cell types, e.g., cardiomyocytes. The decryption of the cellular machinery starting from ECM sensing to iPSC differentiation calls for new in vitro methods. Conveniently, engineered biomaterials activating controlled integrin-mediated responses through chemical, physical, and geometrical designs are key to resolving this issue and could foster clinical translation of optimized iPSC-based technology. This review introduces the main integrin-dependent mechanisms and signalling pathways involved in mechanotransduction. Special consideration is given to the integrin-iPSC linkage signalling chain in the cardiovascular field, focusing on biomaterial-based in vitro models to evaluate the relevance of this process in iPSC differentiation into cardiomyocytes.
Collapse
Affiliation(s)
- Rosaria Santoro
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Gianluca Lorenzo Perrucci
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Aoife Gowran
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Giulio Pompilio
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
- Dipartimento di Scienze Cliniche e di Comunità, Università degli Studi di Milano, via Festa del Perdono 7, Milan, Italy
| |
Collapse
|
26
|
Xu B, Magli A, Anugrah Y, Koester SJ, Perlingeiro RCR, Shen W. Nanotopography-responsive myotube alignment and orientation as a sensitive phenotypic biomarker for Duchenne Muscular Dystrophy. Biomaterials 2018; 183:54-66. [PMID: 30149230 PMCID: PMC6239205 DOI: 10.1016/j.biomaterials.2018.08.047] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/09/2018] [Accepted: 08/20/2018] [Indexed: 01/08/2023]
Abstract
Duchenne Muscular Dystrophy (DMD) is a fatal genetic disorder currently having no cure. Here we report that culture substrates patterned with nanogrooves and functionalized with Matrigel (or laminin) present an engineered cell microenvironment to allow myotubes derived from non-diseased, less-affected DMD, and severely-affected DMD human induced pluripotent stem cells (hiPSCs) to exhibit prominent differences in alignment and orientation, providing a sensitive phenotypic biomarker to potentially facilitate DMD drug development and early diagnosis. We discovered that myotubes differentiated from myogenic progenitors derived from non-diseased hiPSCs align nearly perpendicular to nanogrooves, a phenomenon not reported previously. We further found that myotubes derived from hiPSCs of a dystrophin-null DMD patient orient randomly, and those from hiPSCs of a patient carrying partially functional dystrophin align approximately 14° off the alignment direction of non-diseased myotubes. Substrates engineered with micron-scale grooves and/or cell adhesion molecules only interacting with integrins all guide parallel myotube alignment to grooves and lose the ability to distinguish different cell types. Disruption of the interaction between the Dystrophin-Associated-Protein-Complex (DAPC) and laminin by heparin or anti-α-dystroglycan antibody IIH6 disenables myotubes to align perpendicular to nanogrooves, suggesting that this phenotype is controlled by the DAPC-mediated cytoskeleton-extracellular matrix linkage.
Collapse
Affiliation(s)
- Bin Xu
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Alessandro Magli
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Yoska Anugrah
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Steven J Koester
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455, USA; Institute for Engineering in Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Rita C R Perlingeiro
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA; Institute for Engineering in Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Wei Shen
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA; Institute for Engineering in Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
27
|
Geng L, Kong CW, Wong AOT, Shum AMY, Chow MZY, Che H, Zhang C, Yau KL, Chan CW, Keung W, Li RA. Probing flecainide block of I Na using human pluripotent stem cell-derived ventricular cardiomyocytes adapted to automated patch-clamping and 2D monolayers. Toxicol Lett 2018; 294:61-72. [PMID: 29758359 DOI: 10.1016/j.toxlet.2018.05.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 04/23/2018] [Accepted: 05/07/2018] [Indexed: 11/19/2022]
Abstract
Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) are emerging tools for applications such as drug discovery and screening for pro-arrhythmogenicity and cardiotoxicity as leading causes for drug attrition. Understanding the electrophysiology (EP) of hPSC-CMs is essential but conventional manual patch-clamping is highly laborious and low-throughput. Here we adapted hPSC-CMs derived from two human embryonic stem cell (hESC) lines, HES2 and H7, for a 16-channel automated planar-recording approach for single-cell EP characterization. Automated current- and voltage-clamping, with an overall success rate of 55.0 ± 11.3%, indicated that 90% of hPSC-CMs displayed ventricular-like action potential (AP) and the ventricular cardiomyocytes (VCMs) derived from the two hESC lines expressed similar levels of INa, ICaL, Ikr and If and similarly lacked Ito and IK1. These well-characterized hPSC-VCMs could also be readily adapted for automated assays of pro-arrhythmic drug screening. As an example, we showed that flecainide (FLE) induced INa blockade, leftward steady-state inactivation shift, slowed recovery from inactivation in our hPSC-VCMs. Since single-cell EP assay is insufficient to predict drug-induced reentrant arrhythmias, hPSC-VCMs were further reassembled into 2D human ventricular cardiac monolayers (hvCMLs) for multi-cellular electrophysiological assessments. Indeed, FLE significantly slowed the conduction velocity while causing AP prolongation. Our RNA-seq data suggested that cell-cell interaction enhanced the maturity of hPSC-VCMs. Taken collectively, a combinatorial approach using single-cell EP and hvCMLs is needed to comprehensively assess drug-induced arrhythmogenicity.
Collapse
Affiliation(s)
- Lin Geng
- Stem Cell & Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong
| | - Chi-Wing Kong
- Stem Cell & Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Andy O T Wong
- Stem Cell & Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Angie Man-Yee Shum
- Stem Cell & Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Maggie Z Y Chow
- Stem Cell & Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Hui Che
- Stem Cell & Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Chenzi Zhang
- Stem Cell & Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Ka-Long Yau
- Stem Cell & Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Camie W Chan
- Stem Cell & Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Wendy Keung
- Stem Cell & Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong
| | - Ronald A Li
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong; Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Hong Kong.
| |
Collapse
|
28
|
Li RA, Keung W, Cashman TJ, Backeris PC, Johnson BV, Bardot ES, Wong AOT, Chan PKW, Chan CWY, Costa KD. Bioengineering an electro-mechanically functional miniature ventricular heart chamber from human pluripotent stem cells. Biomaterials 2018; 163:116-127. [PMID: 29459321 DOI: 10.1016/j.biomaterials.2018.02.024] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 02/09/2018] [Indexed: 12/14/2022]
Abstract
Tissue engineers and stem cell biologists have made exciting progress toward creating simplified models of human heart muscles or aligned monolayers to help bridge a longstanding gap between experimental animals and clinical trials. However, no existing human in vitro systems provide the direct measures of cardiac performance as a pump. Here, we developed a next-generation in vitro biomimetic model of pumping human heart chamber, and demonstrated its capability for pharmaceutical testing. From human pluripotent stem cell (hPSC)-derived ventricular cardiomyocytes (hvCM) embedded in collagen-based extracellular matrix hydrogel, we engineered a three-dimensional (3D) electro-mechanically coupled, fluid-ejecting miniature human ventricle-like cardiac organoid chamber (hvCOC). Structural characterization showed organized sarcomeres with myofibrillar microstructures. Transcript and RNA-seq analyses revealed upregulation of key Ca2+-handling, ion channel, and cardiac-specific proteins in hvCOC compared to lower-order 2D and 3D cultures of the same constituent cells. Clinically-important, physiologically complex contractile parameters such as ejection fraction, developed pressure, and stroke work, as well as electrophysiological properties including action potential and conduction velocity were measured: hvCOC displayed key molecular and physiological characteristics of the native ventricle, and showed expected mechanical and electrophysiological responses to a range of pharmacological interventions (including positive and negative inotropes). We conclude that such "human-heart-in-a-jar" technology could facilitate the drug discovery process by providing human-specific preclinical data during early stage drug development.
Collapse
Affiliation(s)
- Ronald A Li
- Ming-Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Sweden; Dr. Li Dak-Sum Research Centre, The University of Hong Kong-Karolinska Institutet Collaboration on Regenerative Medicine, The University of Hong Kong, Pokfulam, Hong Kong; Stem Cell & Regenerative Medicine Consortium, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong; Novoheart Limited, Shatin, Hong Kong.
| | - Wendy Keung
- Ming-Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Sweden; Dr. Li Dak-Sum Research Centre, The University of Hong Kong-Karolinska Institutet Collaboration on Regenerative Medicine, The University of Hong Kong, Pokfulam, Hong Kong; Stem Cell & Regenerative Medicine Consortium, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Timothy J Cashman
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peter C Backeris
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bryce V Johnson
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Evan S Bardot
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andy O T Wong
- Dr. Li Dak-Sum Research Centre, The University of Hong Kong-Karolinska Institutet Collaboration on Regenerative Medicine, The University of Hong Kong, Pokfulam, Hong Kong; Stem Cell & Regenerative Medicine Consortium, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Patrick K W Chan
- Ming-Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Sweden; Stem Cell & Regenerative Medicine Consortium, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Camie W Y Chan
- Stem Cell & Regenerative Medicine Consortium, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong; Novoheart Limited, Shatin, Hong Kong
| | - Kevin D Costa
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Novoheart Limited, Shatin, Hong Kong.
| |
Collapse
|
29
|
Wang J, Cui C, Nan H, Yu Y, Xiao Y, Poon E, Yang G, Wang X, Wang C, Li L, Boheler KR, Ma X, Cheng X, Ni Z, Chen M. Graphene Sheet-Induced Global Maturation of Cardiomyocytes Derived from Human Induced Pluripotent Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2017; 9:25929-25940. [PMID: 28718622 DOI: 10.1021/acsami.7b08777] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Human induced pluripotent stem cells (hiPSCs) can proliferate infinitely. Their ability to differentiate into cardiomyocytes provides abundant sources for disease modeling, drug screening and regenerative medicine. However, hiPSC-derived cardiomyocytes (hiPSC-CMs) display a low degree of maturation and fetal-like properties. Current in vitro differentiation methods do not mimic the structural, mechanical, or physiological properties of the cardiogenesis niche. Recently, we present an efficient cardiac maturation platform that combines hiPSCs monolayer cardiac differentiation with graphene substrate, which is a biocompatible and superconductive material. The hiPSCs lines were successfully maintained on the graphene sheets and were able to differentiate into functional cardiomyocytes. This strategy markedly increased the myofibril ultrastructural organization, elevated the conduction velocity, and enhanced both the Ca2+ handling and electrophysiological properties in the absence of electrical stimulation. On the graphene substrate, the expression of connexin 43 increased along with the conduction velocity. Interestingly, the bone morphogenetic proteins signaling was also significantly activated during early cardiogenesis, confirmed by RNA sequencing analysis. Here, we reasoned that graphene substrate as a conductive biomimetic surface could facilitate the intrinsic electrical propagation, mimicking the microenvironment of the native heart, to further promote the global maturation of hiPSC-CMs. Our findings highlight the capability of electrically active substrates to influence cardiomyocyte development. We believe that application of graphene sheets will be useful for simple, fast, and scalable maturation of regenerated cardiomyocytes.
Collapse
Affiliation(s)
- Jiaxian Wang
- National Center for Human Genetics, National Research Institute for Family Planning , Beijing 100081, China
| | - Chang Cui
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University , Nanjing 210029, China
| | - Haiyan Nan
- Department of Physics, Southeast University , Nanjing 211189, China
| | - Yuanfang Yu
- Department of Physics, Southeast University , Nanjing 211189, China
| | - Yini Xiao
- State Key Laboratory of Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academic of Sciences , Shanghai 200031, China
| | - Ellen Poon
- Stem Cell and Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong , Pokfulam 999077, Hong Kong
| | - Gang Yang
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University , Nanjing 210029, China
| | - Xijie Wang
- National Shanghai Center for New Drug Safety Evaluation and Research , Shanghai 201210, China
| | - Chenchen Wang
- SARI Center for Stem Cell and Nanomedicine, Shanghai Advanced Research Institute, University of Chinese Academy of Sciences , Shanghai 201210, China
| | - Lingsong Li
- SARI Center for Stem Cell and Nanomedicine, Shanghai Advanced Research Institute, University of Chinese Academy of Sciences , Shanghai 201210, China
| | - Kenneth Richard Boheler
- Stem Cell and Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong , Pokfulam 999077, Hong Kong
| | - Xu Ma
- National Center for Human Genetics, National Research Institute for Family Planning , Beijing 100081, China
| | - Xin Cheng
- State Key Laboratory of Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academic of Sciences , Shanghai 200031, China
| | - Zhenhua Ni
- Department of Physics, Southeast University , Nanjing 211189, China
| | - Minglong Chen
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University , Nanjing 210029, China
| |
Collapse
|
30
|
Park SJ, Kim KH, Jeon WY, Seo J, Han JM, Kim JS, Chung HM, Lee JH, Moon SH, Kim HH. Enzyme catalyzed electrostimulation of human embryonic stem cell-derived cardiomyocytes influence contractility and synchronization. Biochem Eng J 2017. [DOI: 10.1016/j.bej.2017.03.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
31
|
Yan S, Li X, Dai J, Wang Y, Wang B, Lu Y, Shi J, Huang P, Gong J, Yao Y. Electrospinning of PVA/sericin nanofiber and the effect on epithelial-mesenchymal transition of A549 cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017. [PMID: 28629038 DOI: 10.1016/j.msec.2017.05.048] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This research aims to investigate the cell-nanomaterial interaction between epithelial-mesenchymal transition of A549 cell and electrospinning nanofibers composed of polyvinyl alcohol (PVA)/silk sericin (SS). The electrospinning of regenerated nanofiber was performed with water as a spinning solvent and glutaraldehyde as a chemical cross-linker. Solution concentration, applied voltage and spin distances as well as other parameters were optimized to generate fine nanofibers with smooth surface in good homogeneity. From the scanning electron microscopy (SEM) analysis, the nanofibers had an average diameter of 200nm. Epithelial-mesenchymal transition (EMT) is a process by which epithelial cells lose their cell polarity to become mesenchymal stem cells. This transition is affected by multiple biochemical and physical factors in cell metabolism cascade. Herein, we investigate the biophysical effect on A549 EMT by culturing cells on nanofibrous mats with different topography and composition. The cell viability was evaluated by biochemical assay and its morphology was observed with SEM. The results demonstrate that cells appropriately attached to the surface of the nanofibrous mats with extended morphology by their filopodia. Gene expression analysis was conducted by real-time PCR using multiple markers for detecting EMT: N-cadherin (NCad), Vimentin (Vim), Fibronectin (Fib) and Matrix metallopeptidase (MMP9). An increasing expression pattern was observed on NCad, Vim, Fib, with respect to a negative control as cell cultured on polystyrene dish. This result indicates the 200nm PVA/SS nanofibers may induce A549 cells to process epithelial-mesenchymal transition during the culturing.
Collapse
Affiliation(s)
- Shanshan Yan
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China; Shanghai Institute of Ceramics, Chinese Academy of Science, 1295 Dingxi Road, Changning, Shanghai 200050, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Shijingshan, Beijing 100049, China
| | - Xiuchun Li
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China
| | - Jing Dai
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China
| | - Yiqun Wang
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China
| | - Binbin Wang
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Shijingshan, Beijing 100049, China; Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Xuhui, Shanghai 200031, China
| | - Yi Lu
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China
| | - Jianlin Shi
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China; Shanghai Institute of Ceramics, Chinese Academy of Science, 1295 Dingxi Road, Changning, Shanghai 200050, China
| | - Pengyu Huang
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China
| | - Jinkang Gong
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China.
| | - Yuan Yao
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China.
| |
Collapse
|
32
|
Higuchi A, Suresh Kumar S, Ling QD, Alarfaj AA, Munusamy MA, Murugan K, Hsu ST, Benelli G, Umezawa A. Polymeric design of cell culture materials that guide the differentiation of human pluripotent stem cells. Prog Polym Sci 2017. [DOI: 10.1016/j.progpolymsci.2016.09.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
33
|
Shum AMY, Che H, Wong AOT, Zhang C, Wu H, Chan CWY, Costa K, Khine M, Kong CW, Li RA. A Micropatterned Human Pluripotent Stem Cell-Based Ventricular Cardiac Anisotropic Sheet for Visualizing Drug-Induced Arrhythmogenicity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1602448. [PMID: 27805726 DOI: 10.1002/adma.201602448] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 09/02/2016] [Indexed: 06/06/2023]
Abstract
A novel cardiomimetic biohybrid material, termed as the human ventricular cardiac anisotropic sheet (hvCAS) is reported. Well-characterized human pluripotent stem-cell-derived ventricular cardiomyocytes are strategically aligned to reproduce key electrophysiological features of native human ventricle, which, along with specific selection criteria, allows for a direct visualization of arrhythmic spiral re-entry and represents a revolutionary tool to assess preclinical drug-induced arrhythmogenicity.
Collapse
Affiliation(s)
- Angie M Y Shum
- Stem Cell & Regenerative Medicine Consortium, LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong
| | - Hui Che
- Stem Cell & Regenerative Medicine Consortium, LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong
| | - Andy On-Tik Wong
- Stem Cell & Regenerative Medicine Consortium, LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong
| | - Chenzi Zhang
- Stem Cell & Regenerative Medicine Consortium, LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong
| | - Hongkai Wu
- Biomedical Engineering, Hong Kong University of Science & Technology, Clear Water Bay, Hong Kong
| | - Camie W Y Chan
- Novoheart Ltd., Hong Kong Science Park, Shatin, Hong Kong
| | - Kevin Costa
- Icahn School of Medicine at Mount Sinai, Manhattan, NYC, 10029-5674, USA
| | - Michelle Khine
- Biomedical Engineering, University of California at Irvine, Irvine, CA, 92697-2715, USA
| | - Chi-Wing Kong
- Stem Cell & Regenerative Medicine Consortium, LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong
| | - Ronald A Li
- Dr. Li Dak-Sum Research Centre, University of Hong Kong, Pokfulam, Hong Kong
- Ming-Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Stockholm, 17177, Sweden
| |
Collapse
|
34
|
Calderon D, Bardot E, Dubois N. Probing early heart development to instruct stem cell differentiation strategies. Dev Dyn 2016; 245:1130-1144. [PMID: 27580352 DOI: 10.1002/dvdy.24441] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 08/20/2016] [Accepted: 08/20/2016] [Indexed: 12/19/2022] Open
Abstract
Scientists have studied organs and their development for centuries and, along that path, described models and mechanisms explaining the developmental principles of organogenesis. In particular, with respect to the heart, new fundamental discoveries are reported continuously that keep changing the way we think about early cardiac development. These discoveries are driven by the need to answer long-standing questions regarding the origin of the earliest cells specified to the cardiac lineage, the differentiation potential of distinct cardiac progenitor cells, and, very importantly, the molecular mechanisms underlying these specification events. As evidenced by numerous examples, the wealth of developmental knowledge collected over the years has had an invaluable impact on establishing efficient strategies to generate cardiovascular cell types ex vivo, from either pluripotent stem cells or via direct reprogramming approaches. The ability to generate functional cardiovascular cells in an efficient and reliable manner will contribute to therapeutic strategies aimed at alleviating the increasing burden of cardiovascular disease and morbidity. Here we will discuss the recent discoveries in the field of cardiac progenitor biology and their translation to the pluripotent stem cell model to illustrate how developmental concepts have instructed regenerative model systems in the past and promise to do so in the future. Developmental Dynamics 245:1130-1144, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Damelys Calderon
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, NY, USA.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Evan Bardot
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, NY, USA.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Nicole Dubois
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, NY, USA.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY, USA
| |
Collapse
|
35
|
Carson D, Hnilova M, Yang X, Nemeth CL, Tsui JH, Smith AS, Jiao A, Regnier M, Murry CE, Tamerler C, Kim DH. Nanotopography-Induced Structural Anisotropy and Sarcomere Development in Human Cardiomyocytes Derived from Induced Pluripotent Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2016; 8:21923-32. [PMID: 26866596 PMCID: PMC5681855 DOI: 10.1021/acsami.5b11671] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Understanding the phenotypic development of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) is a prerequisite to advancing regenerative cardiac therapy, disease modeling, and drug screening applications. Lack of consistent hiPSC-CM in vitro data can be largely attributed to the inability of conventional culture methods to mimic the structural, biochemical, and mechanical aspects of the myocardial niche accurately. Here, we present a nanogrid culture array comprised of nanogrooved topographies, with groove widths ranging from 350 to 2000 nm, to study the effect of different nanoscale structures on the structural development of hiPSC-CMs in vitro. Nanotopographies were designed to have a biomimetic interface, based on observations of the oriented myocardial extracellular matrix (ECM) fibers found in vivo. Nanotopographic substrates were integrated with a self-assembling chimeric peptide containing the Arg-Gly-Asp (RGD) cell adhesion motif. Using this platform, cell adhesion to peptide-coated substrates was found to be comparable to that of conventional fibronectin-coated surfaces. Cardiomyocyte organization and structural development were found to be dependent on the nanotopographical feature size in a biphasic manner, with improved development achieved on grooves in the 700-1000 nm range. These findings highlight the capability of surface-functionalized, bioinspired substrates to influence cardiomyocyte development, and the capacity for such platforms to serve as a versatile assay for investigating the role of topographical guidance cues on cell behavior. Such substrates could potentially create more physiologically relevant in vitro cardiac tissues for future drug screening and disease modeling studies.
Collapse
Affiliation(s)
- Daniel Carson
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Marketa Hnilova
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Xiulan Yang
- Department of Pathology, University of Washington, Seattle, Washington 98195, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, United States
| | - Cameron L. Nemeth
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Jonathan H. Tsui
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Alec S.T. Smith
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Alex Jiao
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
- Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, United States
| | - Charles E. Murry
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
- Department of Pathology, University of Washington, Seattle, Washington 98195, United States
- Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, United States
| | - Candan Tamerler
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, United States
- Department of Mechanical Engineering and Bioengineering Research Center, University of Kansas, Lawrence, Kansas 66045, United States
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
- Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98109, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98109, United States
- Corresponding Author: . Phone: 1-206-616-1133. Fax: 1-206-685-3300
| |
Collapse
|
36
|
Wang Z, Tonderys D, Leggett SE, Williams EK, Kiani MT, Steinberg RS, Qiu Y, Wong IY, Hurt RH. Wrinkled, wavelength-tunable graphene-based surface topographies for directing cell alignment and morphology. CARBON 2016; 97:14-24. [PMID: 25848137 PMCID: PMC4384125 DOI: 10.1016/j.carbon.2015.03.040] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Textured surfaces with periodic topographical features and long-range order are highly attractive for directing cell-material interactions. They mimic physiological environments more accurately than planar surfaces and can fundamentally alter cell alignment, shape, gene expression, and cellular assembly into superstructures or microtissues. Here we demonstrate for the first time that wrinkled graphene-based surfaces are suitable as textured cell attachment substrates, and that engineered wrinkling can dramatically alter cell alignment and morphology. The wrinkled surfaces are fabricated by graphene oxide wet deposition onto pre-stretched elastomers followed by relaxation and mild thermal treatment to stabilize the films in cell culture medium. Multilayer graphene oxide films form periodic, delaminated buckle textures whose wavelengths and amplitudes can be systematically tuned by variation in the wet deposition process. Human and murine fibroblasts attach to these textured films and remain viable, while developing pronounced alignment and elongation relative to those on planar graphene controls. Compared to lithographic patterning of nanogratings, this method has advantages in the simplicity and scalability of fabrication, as well as the opportunity to couple the use of topographic cues with the unique conductive, adsorptive, or barrier properties of graphene materials for functional biomedical devices.
Collapse
Affiliation(s)
- Zhongying Wang
- School of Engineering, Brown University, Providence, RI 02912
- Department of Chemistry, Brown University, Providence, RI 02912
| | - Daniel Tonderys
- School of Engineering, Brown University, Providence, RI 02912
- Center for Biomedical Engineering, Brown University, Providence, RI 02912
| | - Susan E. Leggett
- School of Engineering, Brown University, Providence, RI 02912
- Center for Biomedical Engineering, Brown University, Providence, RI 02912
- Pathobiology Graduate Program, Brown University, Providence, RI 02912
| | - Evelyn Kendall Williams
- School of Engineering, Brown University, Providence, RI 02912
- Center for Biomedical Engineering, Brown University, Providence, RI 02912
| | - Mehrdad T. Kiani
- School of Engineering, Brown University, Providence, RI 02912
- Center for Biomedical Engineering, Brown University, Providence, RI 02912
| | | | - Yang Qiu
- School of Engineering, Brown University, Providence, RI 02912
| | - Ian Y. Wong
- School of Engineering, Brown University, Providence, RI 02912
- Center for Biomedical Engineering, Brown University, Providence, RI 02912
- Pathobiology Graduate Program, Brown University, Providence, RI 02912
- Institute for Molecular and Nanoscale Innovation, Brown University, Providence, RI 02912
| | - Robert H. Hurt
- School of Engineering, Brown University, Providence, RI 02912
- Center for Biomedical Engineering, Brown University, Providence, RI 02912
- Institute for Molecular and Nanoscale Innovation, Brown University, Providence, RI 02912
| |
Collapse
|
37
|
Cashman TJ, Josowitz R, Johnson BV, Gelb BD, Costa KD. Human Engineered Cardiac Tissues Created Using Induced Pluripotent Stem Cells Reveal Functional Characteristics of BRAF-Mediated Hypertrophic Cardiomyopathy. PLoS One 2016; 11:e0146697. [PMID: 26784941 PMCID: PMC4718533 DOI: 10.1371/journal.pone.0146697] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 12/21/2015] [Indexed: 12/22/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a leading cause of sudden cardiac death that often goes undetected in the general population. HCM is also prevalent in patients with cardio-facio-cutaneous syndrome (CFCS), which is a genetic disorder characterized by aberrant signaling in the RAS/MAPK signaling cascade. Understanding the mechanisms of HCM development in such RASopathies may lead to novel therapeutic strategies, but relevant experimental models of the human condition are lacking. Therefore, the objective of this study was to develop the first 3D human engineered cardiac tissue (hECT) model of HCM. The hECTs were created using human cardiomyocytes obtained by directed differentiation of induced pluripotent stem cells derived from a patient with CFCS due to an activating BRAF mutation. The mutant myocytes were directly conjugated at a 3:1 ratio with a stromal cell population to create a tissue of defined composition. Compared to healthy patient control hECTs, BRAF-hECTs displayed a hypertrophic phenotype by culture day 6, with significantly increased tissue size, twitch force, and atrial natriuretic peptide (ANP) gene expression. Twitch characteristics reflected increased contraction and relaxation rates and shorter twitch duration in BRAF-hECTs, which also had a significantly higher maximum capture rate and lower excitation threshold during electrical pacing, consistent with a more arrhythmogenic substrate. By culture day 11, twitch force was no longer different between BRAF and wild-type hECTs, revealing a temporal aspect of disease modeling with tissue engineering. Principal component analysis identified diastolic force as a key factor that changed from day 6 to day 11, supported by a higher passive stiffness in day 11 BRAF-hECTs. In summary, human engineered cardiac tissues created from BRAF mutant cells recapitulated, for the first time, key aspects of the HCM phenotype, offering a new in vitro model for studying intrinsic mechanisms and screening new therapeutic approaches for this lethal form of heart disease.
Collapse
Affiliation(s)
- Timothy J. Cashman
- The Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York City, New York, United States of America
| | - Rebecca Josowitz
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York City, New York, United States of America
| | - Bryce V. Johnson
- The Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York City, New York, United States of America
| | - Bruce D. Gelb
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York City, New York, United States of America
| | - Kevin D. Costa
- The Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York City, New York, United States of America
- * E-mail:
| |
Collapse
|
38
|
Bae WG, Kim J, Choung YH, Chung Y, Suh KY, Pang C, Chung JH, Jeong HE. Bio-inspired configurable multiscale extracellular matrix-like structures for functional alignment and guided orientation of cells. Biomaterials 2015; 69:158-64. [DOI: 10.1016/j.biomaterials.2015.08.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 08/02/2015] [Accepted: 08/04/2015] [Indexed: 11/26/2022]
|
39
|
Qi Y, Li Z, Kong CW, Tang NL, Huang Y, Li RA, Yao X. Uniaxial cyclic stretch stimulates TRPV4 to induce realignment of human embryonic stem cell-derived cardiomyocytes. J Mol Cell Cardiol 2015; 87:65-73. [DOI: 10.1016/j.yjmcc.2015.08.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 08/01/2015] [Accepted: 08/06/2015] [Indexed: 12/28/2022]
|
40
|
Huyer LD, Montgomery M, Zhao Y, Xiao Y, Conant G, Korolj A, Radisic M. Biomaterial based cardiac tissue engineering and its applications. Biomed Mater 2015; 10:034004. [PMID: 25989939 PMCID: PMC4464787 DOI: 10.1088/1748-6041/10/3/034004] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cardiovascular disease is a leading cause of death worldwide, necessitating the development of effective treatment strategies. A myocardial infarction involves the blockage of a coronary artery leading to depletion of nutrient and oxygen supply to cardiomyocytes and massive cell death in a region of the myocardium. Cardiac tissue engineering is the growth of functional cardiac tissue in vitro on biomaterial scaffolds for regenerative medicine application. This strategy relies on the optimization of the complex relationship between cell networks and biomaterial properties. In this review, we discuss important biomaterial properties for cardiac tissue engineering applications, such as elasticity, degradation, and induced host response, and their relationship to engineered cardiac cell environments. With these properties in mind, we also emphasize in vitro use of cardiac tissues for high-throughput drug screening and disease modelling.
Collapse
Affiliation(s)
- Locke Davenport Huyer
- 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
| | - Miles Montgomery
- 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
| | - Yimu Zhao
- 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
| | - Yun Xiao
- 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
| | - Genevieve Conant
- 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
| | - Anastasia Korolj
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
| | - 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 and IBBME, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
41
|
Farouz Y, Chen Y, Terzic A, Menasché P. Concise Review: Growing Hearts in the Right Place: On the Design of Biomimetic Materials for Cardiac Stem Cell Differentiation. Stem Cells 2015; 33:1021-35. [DOI: 10.1002/stem.1929] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 11/10/2014] [Accepted: 12/01/2014] [Indexed: 12/25/2022]
Affiliation(s)
- Yohan Farouz
- Department of Chemistry, Paris Sciences et Lettres, Ecole Normale Supérieure de Paris; CNRS UMR; Paris France
- Sorbonne Paris Cité; Paris Descartes University; Paris France
- INSERM U970; Paris France
| | - Yong Chen
- Department of Chemistry, Paris Sciences et Lettres, Ecole Normale Supérieure de Paris; CNRS UMR; Paris France
| | | | - Philippe Menasché
- Sorbonne Paris Cité; Paris Descartes University; Paris France
- INSERM U970; Paris France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou; Department of Cardiovascular Surgery; Paris France
| |
Collapse
|
42
|
Higuchi A, Ling QD, Kumar SS, Chang Y, Alarfaj AA, Munusamy MA, Murugan K, Hsu ST, Umezawa A. Physical cues of cell culture materials lead the direction of differentiation lineages of pluripotent stem cells. J Mater Chem B 2015; 3:8032-8058. [DOI: 10.1039/c5tb01276g] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Differentiation methods of hPSCs into specific cell lineages. Differentiation of hPSCsviaEB formation (types AB, A–D) or without EB formation (types E–H).
Collapse
Affiliation(s)
- Akon Higuchi
- Department of Chemical and Materials Engineering, National Central University
- Taoyuan 32001
- Taiwan
- National Research Institute for Child Health and Development
- Center for Regenerative Medicine
| | - Qing-Dong Ling
- Cathay Medical Research Institute
- Cathay General Hospital
- Taipei
- Taiwan
- Graduate Institute of Systems Biology and Bioinformatics
| | - S. Suresh Kumar
- Department of Medical Microbiology and Parasitology
- Universiti Putra Malaysia
- Selangor
- Malaysia
| | - Yung Chang
- Department of Chemical Engineering
- R&D Center for Membrane Technology
- Chung Yuan Christian University
- Taoyuan
- Taiwan
| | - Abdullah A. Alarfaj
- Department of Botany and Microbiology
- College of Science
- King Saud University
- Riyadh
- Saudi Arabia
| | - Murugan A. Munusamy
- Department of Botany and Microbiology
- College of Science
- King Saud University
- Riyadh
- Saudi Arabia
| | - Kadarkarai Murugan
- Division of Entomology
- Department of Zoology
- School of Life Sciences
- Bharathiar University
- Coimbatore 641046
| | - Shih-Tien Hsu
- Department of Internal Medicine
- Taiwan Landseed Hospital
- Taoyuan
- Taiwan
| | - Akihiro Umezawa
- National Research Institute for Child Health and Development
- Center for Regenerative Medicine
- Tokyo 157-8535
- Japan
| |
Collapse
|
43
|
Chen G, Li S, Karakikes I, Ren L, Chow MZY, Chopra A, Keung W, Yan B, Chan CWY, Costa KD, Kong CW, Hajjar RJ, Chen CS, Li RA. Phospholamban as a crucial determinant of the inotropic response of human pluripotent stem cell-derived ventricular cardiomyocytes and engineered 3-dimensional tissue constructs. Circ Arrhythm Electrophysiol 2014; 8:193-202. [PMID: 25504561 DOI: 10.1161/circep.114.002049] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Human (h) embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) serve as a potential unlimited ex vivo source of cardiomyocytes (CMs). However, a well-accepted roadblock has been their immature phenotype. hESC/iPSC-derived ventricular (v) CMs and their engineered cardiac microtissues (hvCMTs) similarly displayed positive chronotropic but null inotropic responses to β-adrenergic stimulation. Given that phospholamban (PLB) is robustly present in adult but poorly expressed in hESC/iPSC-vCMs and its defined biological role in β-adrenergic signaling, we investigated the functional consequences of PLB expression in hESC/iPSC-vCMs and hvCMTs. METHODS AND RESULTS First, we confirmed that PLB protein was differentially expressed in hESC (HES2, H9)- and iPSC-derived and adult vCMs. We then transduced hES2-vCMs with the recombinant adenoviruses (Ad) Ad-PLB or Ad-S16E-PLB to overexpress wild-type PLB or the pseudophosphorylated point-mutated variant, respectively. As anticipated from the inhibitory effect of unphosphorylated PLB on sarco/endoplasmic reticulum Ca2+-ATPase, Ad-PLB transduction significantly attenuated electrically evoked Ca2+ transient amplitude and prolonged the 50% decay time. Importantly, Ad-PLB-transduced hES2-vCMs uniquely responded to isoproterenol. Ad-S16E-PLB-transduced hES2-vCMs displayed an intermediate phenotype. The same trends were observed with H9- and iPSC-vCMs. Directionally, similar results were also seen with Ad-PLB-transduced and Ad-S16E-transduced hvCMTs. However, Ad-PLB altered neither the global transcriptome nor ICa,L, implicating a PLB-specific effect. CONCLUSIONS Engineered upregulation of PLB expression in hESC/iPSC-vCMs restores a positive inotropic response to β-adrenergic stimulation. These results not only provide a better mechanistic understanding of the immaturity of hESC/iPSC-vCMs but will also lead to improved disease models and transplantable prototypes with adult-like physiological responses.
Collapse
Affiliation(s)
- Gaopeng Chen
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, Manhattan, NY (G.C., I.K., K.D.C., R.J.H., R.A.L.); Department of Physiology (G.C., S.L., L.R., M.Z.-Y.C., W.K., C.-W.K., R.A.L.), Stem Cell and Regenerative Medicine Consortium (G.C., S.L., L.R., M.Z.-Y.C., W.K., B.Y., C.W.Y.C., C.-W.K., R.A.L.), Department of Anatomy (C.W.Y.C.), LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong; Department of Bioengineering, Boston University, MA (A.C., C.S.C.); Harvard Wyss Institute for Biologically Inspired Engineering, Boston, MA (A.C., C.S.C.); and Department of Biology, Hong Kong Baptist University, Hong Kong (B.Y.)
| | - Sen Li
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, Manhattan, NY (G.C., I.K., K.D.C., R.J.H., R.A.L.); Department of Physiology (G.C., S.L., L.R., M.Z.-Y.C., W.K., C.-W.K., R.A.L.), Stem Cell and Regenerative Medicine Consortium (G.C., S.L., L.R., M.Z.-Y.C., W.K., B.Y., C.W.Y.C., C.-W.K., R.A.L.), Department of Anatomy (C.W.Y.C.), LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong; Department of Bioengineering, Boston University, MA (A.C., C.S.C.); Harvard Wyss Institute for Biologically Inspired Engineering, Boston, MA (A.C., C.S.C.); and Department of Biology, Hong Kong Baptist University, Hong Kong (B.Y.)
| | - Ioannis Karakikes
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, Manhattan, NY (G.C., I.K., K.D.C., R.J.H., R.A.L.); Department of Physiology (G.C., S.L., L.R., M.Z.-Y.C., W.K., C.-W.K., R.A.L.), Stem Cell and Regenerative Medicine Consortium (G.C., S.L., L.R., M.Z.-Y.C., W.K., B.Y., C.W.Y.C., C.-W.K., R.A.L.), Department of Anatomy (C.W.Y.C.), LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong; Department of Bioengineering, Boston University, MA (A.C., C.S.C.); Harvard Wyss Institute for Biologically Inspired Engineering, Boston, MA (A.C., C.S.C.); and Department of Biology, Hong Kong Baptist University, Hong Kong (B.Y.)
| | - Lihuan Ren
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, Manhattan, NY (G.C., I.K., K.D.C., R.J.H., R.A.L.); Department of Physiology (G.C., S.L., L.R., M.Z.-Y.C., W.K., C.-W.K., R.A.L.), Stem Cell and Regenerative Medicine Consortium (G.C., S.L., L.R., M.Z.-Y.C., W.K., B.Y., C.W.Y.C., C.-W.K., R.A.L.), Department of Anatomy (C.W.Y.C.), LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong; Department of Bioengineering, Boston University, MA (A.C., C.S.C.); Harvard Wyss Institute for Biologically Inspired Engineering, Boston, MA (A.C., C.S.C.); and Department of Biology, Hong Kong Baptist University, Hong Kong (B.Y.)
| | - Maggie Zi-Ying Chow
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, Manhattan, NY (G.C., I.K., K.D.C., R.J.H., R.A.L.); Department of Physiology (G.C., S.L., L.R., M.Z.-Y.C., W.K., C.-W.K., R.A.L.), Stem Cell and Regenerative Medicine Consortium (G.C., S.L., L.R., M.Z.-Y.C., W.K., B.Y., C.W.Y.C., C.-W.K., R.A.L.), Department of Anatomy (C.W.Y.C.), LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong; Department of Bioengineering, Boston University, MA (A.C., C.S.C.); Harvard Wyss Institute for Biologically Inspired Engineering, Boston, MA (A.C., C.S.C.); and Department of Biology, Hong Kong Baptist University, Hong Kong (B.Y.)
| | - Anant Chopra
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, Manhattan, NY (G.C., I.K., K.D.C., R.J.H., R.A.L.); Department of Physiology (G.C., S.L., L.R., M.Z.-Y.C., W.K., C.-W.K., R.A.L.), Stem Cell and Regenerative Medicine Consortium (G.C., S.L., L.R., M.Z.-Y.C., W.K., B.Y., C.W.Y.C., C.-W.K., R.A.L.), Department of Anatomy (C.W.Y.C.), LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong; Department of Bioengineering, Boston University, MA (A.C., C.S.C.); Harvard Wyss Institute for Biologically Inspired Engineering, Boston, MA (A.C., C.S.C.); and Department of Biology, Hong Kong Baptist University, Hong Kong (B.Y.)
| | - Wendy Keung
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, Manhattan, NY (G.C., I.K., K.D.C., R.J.H., R.A.L.); Department of Physiology (G.C., S.L., L.R., M.Z.-Y.C., W.K., C.-W.K., R.A.L.), Stem Cell and Regenerative Medicine Consortium (G.C., S.L., L.R., M.Z.-Y.C., W.K., B.Y., C.W.Y.C., C.-W.K., R.A.L.), Department of Anatomy (C.W.Y.C.), LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong; Department of Bioengineering, Boston University, MA (A.C., C.S.C.); Harvard Wyss Institute for Biologically Inspired Engineering, Boston, MA (A.C., C.S.C.); and Department of Biology, Hong Kong Baptist University, Hong Kong (B.Y.)
| | - Bin Yan
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, Manhattan, NY (G.C., I.K., K.D.C., R.J.H., R.A.L.); Department of Physiology (G.C., S.L., L.R., M.Z.-Y.C., W.K., C.-W.K., R.A.L.), Stem Cell and Regenerative Medicine Consortium (G.C., S.L., L.R., M.Z.-Y.C., W.K., B.Y., C.W.Y.C., C.-W.K., R.A.L.), Department of Anatomy (C.W.Y.C.), LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong; Department of Bioengineering, Boston University, MA (A.C., C.S.C.); Harvard Wyss Institute for Biologically Inspired Engineering, Boston, MA (A.C., C.S.C.); and Department of Biology, Hong Kong Baptist University, Hong Kong (B.Y.)
| | - Camie W Y Chan
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, Manhattan, NY (G.C., I.K., K.D.C., R.J.H., R.A.L.); Department of Physiology (G.C., S.L., L.R., M.Z.-Y.C., W.K., C.-W.K., R.A.L.), Stem Cell and Regenerative Medicine Consortium (G.C., S.L., L.R., M.Z.-Y.C., W.K., B.Y., C.W.Y.C., C.-W.K., R.A.L.), Department of Anatomy (C.W.Y.C.), LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong; Department of Bioengineering, Boston University, MA (A.C., C.S.C.); Harvard Wyss Institute for Biologically Inspired Engineering, Boston, MA (A.C., C.S.C.); and Department of Biology, Hong Kong Baptist University, Hong Kong (B.Y.)
| | - Kevin D Costa
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, Manhattan, NY (G.C., I.K., K.D.C., R.J.H., R.A.L.); Department of Physiology (G.C., S.L., L.R., M.Z.-Y.C., W.K., C.-W.K., R.A.L.), Stem Cell and Regenerative Medicine Consortium (G.C., S.L., L.R., M.Z.-Y.C., W.K., B.Y., C.W.Y.C., C.-W.K., R.A.L.), Department of Anatomy (C.W.Y.C.), LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong; Department of Bioengineering, Boston University, MA (A.C., C.S.C.); Harvard Wyss Institute for Biologically Inspired Engineering, Boston, MA (A.C., C.S.C.); and Department of Biology, Hong Kong Baptist University, Hong Kong (B.Y.)
| | - Chi-Wing Kong
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, Manhattan, NY (G.C., I.K., K.D.C., R.J.H., R.A.L.); Department of Physiology (G.C., S.L., L.R., M.Z.-Y.C., W.K., C.-W.K., R.A.L.), Stem Cell and Regenerative Medicine Consortium (G.C., S.L., L.R., M.Z.-Y.C., W.K., B.Y., C.W.Y.C., C.-W.K., R.A.L.), Department of Anatomy (C.W.Y.C.), LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong; Department of Bioengineering, Boston University, MA (A.C., C.S.C.); Harvard Wyss Institute for Biologically Inspired Engineering, Boston, MA (A.C., C.S.C.); and Department of Biology, Hong Kong Baptist University, Hong Kong (B.Y.)
| | - Roger J Hajjar
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, Manhattan, NY (G.C., I.K., K.D.C., R.J.H., R.A.L.); Department of Physiology (G.C., S.L., L.R., M.Z.-Y.C., W.K., C.-W.K., R.A.L.), Stem Cell and Regenerative Medicine Consortium (G.C., S.L., L.R., M.Z.-Y.C., W.K., B.Y., C.W.Y.C., C.-W.K., R.A.L.), Department of Anatomy (C.W.Y.C.), LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong; Department of Bioengineering, Boston University, MA (A.C., C.S.C.); Harvard Wyss Institute for Biologically Inspired Engineering, Boston, MA (A.C., C.S.C.); and Department of Biology, Hong Kong Baptist University, Hong Kong (B.Y.)
| | - Christopher S Chen
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, Manhattan, NY (G.C., I.K., K.D.C., R.J.H., R.A.L.); Department of Physiology (G.C., S.L., L.R., M.Z.-Y.C., W.K., C.-W.K., R.A.L.), Stem Cell and Regenerative Medicine Consortium (G.C., S.L., L.R., M.Z.-Y.C., W.K., B.Y., C.W.Y.C., C.-W.K., R.A.L.), Department of Anatomy (C.W.Y.C.), LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong; Department of Bioengineering, Boston University, MA (A.C., C.S.C.); Harvard Wyss Institute for Biologically Inspired Engineering, Boston, MA (A.C., C.S.C.); and Department of Biology, Hong Kong Baptist University, Hong Kong (B.Y.)
| | - Ronald A Li
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, Manhattan, NY (G.C., I.K., K.D.C., R.J.H., R.A.L.); Department of Physiology (G.C., S.L., L.R., M.Z.-Y.C., W.K., C.-W.K., R.A.L.), Stem Cell and Regenerative Medicine Consortium (G.C., S.L., L.R., M.Z.-Y.C., W.K., B.Y., C.W.Y.C., C.-W.K., R.A.L.), Department of Anatomy (C.W.Y.C.), LKS Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong; Department of Bioengineering, Boston University, MA (A.C., C.S.C.); Harvard Wyss Institute for Biologically Inspired Engineering, Boston, MA (A.C., C.S.C.); and Department of Biology, Hong Kong Baptist University, Hong Kong (B.Y.).
| |
Collapse
|
44
|
Chen Y, Wang J, Shen B, Chan CWY, Wang C, Zhao Y, Chan HN, Tian Q, Chen Y, Yao C, Hsing IM, Li RA, Wu H. Engineering a Freestanding Biomimetic Cardiac Patch Using Biodegradable Poly(lactic-co-glycolic acid) (PLGA) and Human Embryonic Stem Cell-derived Ventricular Cardiomyocytes (hESC-VCMs). Macromol Biosci 2014; 15:426-36. [DOI: 10.1002/mabi.201400448] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Revised: 11/15/2014] [Indexed: 01/06/2023]
Affiliation(s)
- Yin Chen
- Division of Biomedical Engineering; The Hong Kong University of Science and Technology; Hong Kong China
| | - Junping Wang
- Stem Cell & Regenerative Medicine Consortium, LKS Faculty of Medicine; The University of Hong Kong; Hong Kong China
- Department of Physiology, LKS Faculty of Medicine; The University of Hong Kong; Hong Kong China
| | - Bo Shen
- Department of Chemistry; The Hong Kong University of Science and Technology; Hong Kong China
| | - Camie W. Y. Chan
- Stem Cell & Regenerative Medicine Consortium, LKS Faculty of Medicine; The University of Hong Kong; Hong Kong China
- Department of Anatomy, LKS Faculty of Medicine; The University of Hong Kong; Hong Kong China
| | - Chaoyi Wang
- Department of Civil and Environmental Engineering; The Hong Kong University of Science and Technology; Hong Kong China
| | - Yihua Zhao
- Department of Chemistry; The Hong Kong University of Science and Technology; Hong Kong China
| | - Ho N. Chan
- Department of Chemistry; The Hong Kong University of Science and Technology; Hong Kong China
| | - Qian Tian
- Department of Chemistry; The Hong Kong University of Science and Technology; Hong Kong China
| | - Yangfan Chen
- Department of Chemistry; The Hong Kong University of Science and Technology; Hong Kong China
| | - Chunlei Yao
- Division of Biomedical Engineering; The Hong Kong University of Science and Technology; Hong Kong China
| | - I-Ming Hsing
- Division of Biomedical Engineering; The Hong Kong University of Science and Technology; Hong Kong China
- Department of Chemical and Biomolecular Engineering; The Hong Kong University of Science and Technology; Hong Kong China
| | - Ronald A. Li
- Stem Cell & Regenerative Medicine Consortium, LKS Faculty of Medicine; The University of Hong Kong; Hong Kong China
- Department of Physiology, LKS Faculty of Medicine; The University of Hong Kong; Hong Kong China
| | - Hongkai Wu
- Division of Biomedical Engineering; The Hong Kong University of Science and Technology; Hong Kong China
- Department of Chemistry; The Hong Kong University of Science and Technology; Hong Kong China
| |
Collapse
|
45
|
Lin S, Lee EK, Nguyen N, Khine M. Thermally-induced miniaturization for micro- and nanofabrication: progress and updates. LAB ON A CHIP 2014; 14:3475-88. [PMID: 25075652 PMCID: PMC9061274 DOI: 10.1039/c4lc00528g] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The field of micro- and nanofabrication has developed extensively in the past several decades with rising interest in alternative fabrication techniques. Growth of these areas has been driven by needs that remain unaddressed by traditional lithographical methods: inexpensive, upscalable, biocompatible, and easily integrated into complete lab-on-a-chip (LOC) systems. Shape memory polymers (SMPs) have been explored as an alternative substrate. This review first focuses on structure fabrication at the micron and nanoscale using specifically heat-shrinkable SMPs and highlights the innovative improvements to this technology in the past several years. The second part of the review illustrates demonstrated applications of these micro- and nanostructures fabricated from heat-shrinkable SMP films. The review concludes with a discussion about future prospects of heat-shrinkable SMP structures for integration into LOC systems.
Collapse
Affiliation(s)
- Sophia Lin
- Department of Chemical Engineering and Materials Science, University of California, Irvine, Irvine, CA 92627, USA
| | | | | | | |
Collapse
|
46
|
Lücker PB, Javaherian S, Soleas JP, Halverson D, Zandstra PW, McGuigan AP. A microgroove patterned multiwell cell culture plate for high-throughput studies of cell alignment. Biotechnol Bioeng 2014; 111:2537-48. [DOI: 10.1002/bit.25298] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Revised: 05/21/2014] [Accepted: 05/21/2014] [Indexed: 11/11/2022]
Affiliation(s)
- Petra B. Lücker
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; 200 College St. Toronto Ontario M5T 3J9 Canada
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto Ontario Canada
| | - Sahar Javaherian
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; 200 College St. Toronto Ontario M5T 3J9 Canada
| | - John P. Soleas
- Institute of Medical Science; University of Toronto; Toronto Ontario Canada
| | - Duncan Halverson
- Department of Chemistry; University of Toronto; Toronto Ontario Canada
| | - Peter W. Zandstra
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto Ontario Canada
| | - Alison P. McGuigan
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; 200 College St. Toronto Ontario M5T 3J9 Canada
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto Ontario Canada
| |
Collapse
|
47
|
Keung W, Boheler KR, Li RA. Developmental cues for the maturation of metabolic, electrophysiological and calcium handling properties of human pluripotent stem cell-derived cardiomyocytes. Stem Cell Res Ther 2014; 5:17. [PMID: 24467782 PMCID: PMC4055054 DOI: 10.1186/scrt406] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Human pluripotent stem cells (hPSCs), including embryonic and induced pluripotent stem cells, are abundant sources of cardiomyocytes (CMs) for cell replacement therapy and other applications such as disease modeling, drug discovery and cardiotoxicity screening. However, hPSC-derived CMs display immature structural, electrophysiological, calcium-handling and metabolic properties. Here, we review various biological as well as physical and topographical cues that are known to associate with the development of native CMs in vivo to gain insights into the development of strategies for facilitated maturation of hPSC-CMs.
Collapse
|
48
|
Abstract
The heart is a large organ containing many cell types, each of which is necessary for normal function. Because of this, cardiac regenerative medicine presents many unique challenges. Because each of the many types of cells within the heart has unique physiological and electrophysiological characteristics, donor cells must be well matched to the area of the heart into which they are grafted to avoid mechanical dysfunction or arrhythmia. In addition, grafted cells must be functionally integrated into host tissue to effectively repair cardiac function. Because of its size and physiological function, the metabolic needs of the heart are considerable. Therefore grafts must contain not only cardiomyocytes but also a functional vascular network to meet their needs for oxygen and nutrition. In this article we review progress in the use of pluripotent stem cells as a source of donor cardiomyocytes and highlight current unmet needs in the field. We also examine recent tissue engineering approaches integrating cells with various engineered materials that should address some of these unmet needs.
Collapse
Affiliation(s)
- Yunkai Dai
- Bioengineering Department, Clemson University, Clemson, South Carolina
| | - Ann C. Foley
- Bioengineering Department, Clemson University, Clemson, South Carolina
- Department of Cell and Regenerative Medicine, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
49
|
Mendoza N, Tu R, Chen A, Lee E, Khine M. Shrink-induced biomimetic wrinkled substrates for functional cardiac cell alignment and culture. Methods Mol Biol 2014; 1181:97-108. [PMID: 25070330 DOI: 10.1007/978-1-4939-1047-2_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The anisotropic alignment of cardiomyocytes in native myocardium tissue is a functional feature that is absent in traditional in vitro cardiac cell culture. Microenvironmental factors cue structural organization of the myocardium, which promotes the mechanical contractile properties and electrophysiological patterns seen in mature cardiomyocytes. Current nano- and microfabrication techniques, such as photolithography, generate simplified cell culture topographies that are not truly representative of the multifaceted and multi-scale fibrils of the cardiac extracellular matrix. In addition, such technologies are costly and require a clean room for fabrication. This chapter offers an easy, fast, robust, and inexpensive fabrication of biomimetic multi-scale wrinkled surfaces through the process of plasma treating and shrinking prestressed thermoplastic. Additionally, this chapter includes techniques for culturing stem cells and their cardiac derivatives on these substrates. Importantly, this wrinkled cell culture platform is compatible with both fluorescence and bright-field imaging; real-time physiological monitoring of CM action potential propagation and contraction properties can elucidate cardiotoxicity drug effects.
Collapse
Affiliation(s)
- Nicole Mendoza
- Department of Biomedical Engineering, University of California, 5200 Engineering Hall Irvine, Irvine, CA, USA
| | | | | | | | | |
Collapse
|
50
|
Chen A, Lee E, Tu R, Santiago K, Grosberg A, Fowlkes C, Khine M. Integrated platform for functional monitoring of biomimetic heart sheets derived from human pluripotent stem cells. Biomaterials 2014; 35:675-83. [PMID: 24144905 PMCID: PMC9300448 DOI: 10.1016/j.biomaterials.2013.10.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 10/01/2013] [Indexed: 10/26/2022]
Abstract
We present an integrated platform comprised of a biomimetic substrate and physiologically aligned human pluripotent stem cell-derived cardiomyocytes (CMs) with optical detection and algorithms to monitor subtle changes in cardiac properties under various conditions. In the native heart, anisotropic tissue structures facilitate important concerted mechanical contraction and electrical propagation. To recapitulate the architecture necessary for a physiologically accurate heart response, we have developed a simple way to create large areas of aligned CMs with improved functional properties using shrink-wrap film. Combined with simple bright field imaging, obviating the need for fluorescent labels or beads, we quantify and analyze key cardiac contractile parameters. To evaluate the performance capabilities of this platform, the effects of two drugs, E-4031 and isoprenaline, were examined. Cardiac cells supplemented with E-4031 exhibited an increase in contractile duration exclusively due to prolonged relaxation peak. Notably, cells aligned on the biomimetic platform responded detectably down to a dosage of 3 nM E-4031, which is lower than the IC50 in the hERG channel assay. Cells supplemented with isoprenaline exhibited increased contractile frequency and acceleration. Interestingly, cells grown on the biomimetic substrate were more responsive to isoprenaline than those grown on the two control surfaces, suggesting topography may help induce more mature ion channel development. This simple and low-cost platform could thus be a powerful tool for longitudinal assays as well as an effective tool for drug screening and basic cardiac research.
Collapse
Affiliation(s)
- Aaron Chen
- Department of Chemical Engineering and Materials Science, University of California, Irvine, CA, USA
| | - Eugene Lee
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
| | - Roger Tu
- Department of Biological Sciences, University of California, Irvine, CA, USA
| | - Kevin Santiago
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
| | - Anna Grosberg
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
| | - Charless Fowlkes
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
- Department of Computer Science, University of California, Irvine, CA, USA
| | - Michelle Khine
- Department of Chemical Engineering and Materials Science, University of California, Irvine, CA, USA
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
| |
Collapse
|