201
|
Zhu W, Zheng Y, Liu J, Zhao C, Sun N, Qu X, Yang H. Analysis of Fatty Acid Metabolism in Fetal and Failing Hearts by Single-Cell RNA Sequencing Revealed SLC27A6 as a Critical Gene in Heart Maturation. ACTA CARDIOLOGICA SINICA 2023; 39:580-598. [PMID: 37456940 PMCID: PMC10346055 DOI: 10.6515/acs.202307_39(4).20221219b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 12/19/2022] [Indexed: 07/18/2023]
Abstract
BACKGROUND Heart failure is associated with shifts in substrate preferences and energy insufficiency. Although cardiac metabolism has been explored at the organ level, the metabolic changes at the individual cell level remain unclear. This study employed single-cell ribonucleic acid (RNA) sequencing to investigate the cell-type-specific characteristics of gene expression related to fatty acid metabolism. METHODS Single-cell RNA sequencing data from fetal hearts were processed to analyze gene expression patterns related to fatty acid metabolism. Immunofluorescence staining and Western blotting techniques were employed to validate the expression of specific proteins. Additionally, calcium recording and contractility measurements were performed to assess the functional implications of fatty acid metabolism in cardiomyocytes. RESULTS Based on single-cell RNA sequencing data analysis, we found that a decrease in overall energy requirements underlies the downregulation of fatty acid oxidation-related genes in the later period of heart maturation and the compensatory increase of fatty acid metabolism in individual cardiomyocytes during heart failure. Furthermore, we found that solute carrier family 27 member 6 (SLC27A6), a fatty acid transport protein, is involved in cardiac maturation. SLC27A6 knockdown in human induced pluripotent stem cell-derived cardiomyocytes resulted in an immature cardiomyocyte transcriptional profile, abnormal morphology, impaired Ca2+ handling activity, and contractility. CONCLUSIONS Overall, our study offers a novel perspective for exploring cardiac fatty acid metabolism in fetal and failing hearts along with new insights into the cellular mechanism underlying fatty acid metabolic alterations in individual cardiac cells. It thus facilitates further exploration of cardiac physiology and pathology.
Collapse
Affiliation(s)
- Wenjia Zhu
- Department of Physiology and Pathophysiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai
| | - Yufan Zheng
- Department of Physiology and Pathophysiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai
| | - Jiaying Liu
- Department of Physiology and Pathophysiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai
| | - Chao Zhao
- Department of Physiology and Pathophysiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai
| | - Ning Sun
- Department of Physiology and Pathophysiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai
- Wuxi School of Medicine, Jiangnan University, Jiangsu, China
| | - Xiuxia Qu
- Wuxi School of Medicine, Jiangnan University, Jiangsu, China
| | - Hui Yang
- Department of Physiology and Pathophysiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai
- Wuxi School of Medicine, Jiangnan University, Jiangsu, China
| |
Collapse
|
202
|
Liu L, Xu F, Jin H, Qiu B, Yang J, Zhang W, Gao Q, Lin B, Chen S, Sun D. Integrated Manufacturing of Suspended and Aligned Nanofibrous Scaffold for Structural Maturation and Synchronous Contraction of HiPSC-Derived Cardiomyocytes. Bioengineering (Basel) 2023; 10:702. [PMID: 37370633 DOI: 10.3390/bioengineering10060702] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/30/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Electrospun nanofiber constructs represent a promising alternative for mimicking the natural extracellular matrix in vitro and have significant potential for cardiac patch applications. While the effect of fiber orientation on the morphological structure of cardiomyocytes has been investigated, fibers only provide contact guidance without accounting for substrate stiffness due to their deposition on rigid substrates (e.g., glass or polystyrene). This paper introduces an in situ fabrication method for suspended and well aligned nanofibrous scaffolds via roller electrospinning, providing an anisotropic microenvironment with reduced stiffness for cardiac tissue engineering. A fiber surface modification strategy, utilizing oxygen plasma treatment combined with sodium dodecyl sulfate solution, was proposed to maintain the hydrophilicity of polycaprolactone (PCL) fibers, promoting cellular adhesion. Human-induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs), cultured on aligned fibers, exhibited an elongated morphology with extension along the fiber axis. In comparison to Petri dishes and suspended random fiber scaffolds, hiPSC-CMs on suspended aligned fiber scaffolds demonstrated enhanced sarcomere organization, spontaneous synchronous contraction, and gene expression indicative of maturation. This work demonstrates the suspended and aligned nano-fibrous scaffold provides a more realistic biomimetic environment for hiPSC-CMs, which promoted further research on the inducing effect of fiber scaffolds on hiPSC-CMs microstructure and gene-level expression.
Collapse
Affiliation(s)
- Lingling Liu
- Sabondong Micron Nano Science and Technology Research Institute, Xiamen University, Xiamen 361102, China
| | - Feng Xu
- Sabondong Micron Nano Science and Technology Research Institute, Xiamen University, Xiamen 361102, China
| | - Hang Jin
- Sabondong Micron Nano Science and Technology Research Institute, Xiamen University, Xiamen 361102, China
| | - Bin Qiu
- Sabondong Micron Nano Science and Technology Research Institute, Xiamen University, Xiamen 361102, China
| | - Jianhui Yang
- Sabondong Micron Nano Science and Technology Research Institute, Xiamen University, Xiamen 361102, China
| | - Wangzihan Zhang
- Sabondong Micron Nano Science and Technology Research Institute, Xiamen University, Xiamen 361102, China
| | - Qiang Gao
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangzhou 510080, China
- Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou 510080, China
| | - Bin Lin
- Guangdong Beating Origin Regenerative Medicine Co., Ltd., Foshan 528231, China
| | - Songyue Chen
- Sabondong Micron Nano Science and Technology Research Institute, Xiamen University, Xiamen 361102, China
| | - Daoheng Sun
- Sabondong Micron Nano Science and Technology Research Institute, Xiamen University, Xiamen 361102, China
| |
Collapse
|
203
|
Zhang F, Cheng H, Qu K, Qian X, Lin Y, Zhang Y, Qian S, Huang N, Cui C, Chen M. Continuous contractile force and electrical signal recordings of 3D cardiac tissue utilizing conductive hydrogel pillars on a chip. Mater Today Bio 2023; 20:100626. [PMID: 37122834 PMCID: PMC10130626 DOI: 10.1016/j.mtbio.2023.100626] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/02/2023] [Accepted: 04/04/2023] [Indexed: 05/02/2023] Open
Abstract
Heart-on-chip emerged as a potential tool for cardiac tissue engineering, recapitulating key physiological cues in cardiac pathophysiology. Controlled electrical stimulation and the ability to provide directly analyzed functional readouts are essential to evaluate the physiology of cardiac tissues in the heart-on-chip platforms. In this scenario, a novel heart-on-chip platform integrating two soft conductive hydrogel pillar electrodes was presented here. Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and cardiac fibroblasts were seeded into the apparatus to create 3D human cardiac tissues. The application of electrical stimulation improved functional performance by altering the dynamics of tissue structure and contractile development. The contractile forces that cardiac tissues contract was accurately measured through optical tracking of hydrogel pillar displacement. Furthermore, the conductive properties of hydrogel pillars allowed direct and non-invasive electrophysiology studies, enabling continuous monitoring of signal changes in real-time while dynamically administering drugs to the cardiac tissues, as shown by a chronotropic reaction to isoprenaline and verapamil. Overall, the platform for acquiring contractile force and electrophysiological signals in situ allowed monitoring the tissue development trend without interrupting the culture process and could have diverse applications in preclinical drug testing, disease modeling, and therapeutic discovery.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Hongyi Cheng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
- Gusu School, Nanjing Medical University, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, 215002, China
| | - Kaiyun Qu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Xuetian Qian
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, China
| | - Yongping Lin
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Yike Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Sichong Qian
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Beijing, 100029, China
| | - Ningping Huang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Corresponding author.
| | - Chang Cui
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
- Corresponding author.
| | - Minglong Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Gusu School, Nanjing Medical University, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, 215002, China
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 210000, China
- Corresponding author. Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China.
| |
Collapse
|
204
|
In vitro cell stretching devices and their applications: From cardiomyogenic differentiation to tissue engineering. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2023. [DOI: 10.1016/j.medntd.2023.100220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
|
205
|
Murphy JF, Costa KD, Turnbull IC. Rianú: Multi-tissue tracking software for increased throughput of engineered cardiac tissue screening. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE UPDATE 2023; 3:100107. [PMID: 37476002 PMCID: PMC10359020 DOI: 10.1016/j.cmpbup.2023.100107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
Background The field of tissue engineering has provided valuable three-dimensional species-specific models of the human myocardium in the form of human Engineered Cardiac Tissues (hECTs) and similar constructs. However, hECT systems are often bottlenecked by a lack of openly available software that can collect data from multiple tissues at a time, even in multi-tissue bioreactors, which limits throughput in phenotypic and therapeutic screening applications. Methods We developed Rianú, an open-source web application capable of simultaneously tracking multiple hECTs on flexible end-posts. This software is operating system agnostic and deployable on a remote server, accessible via a web browser with no local hardware or software requirements. The software incorporates object-tracking capabilities for multiple objects simultaneously, an algorithm for twitch tracing analysis and contractile force calculation, and a data compilation system for comparative analysis within and amongst groups. Validation tests were performed using in-silico and in-vitro experiments for comparison with established methods and interventions. Results Rianú was able to detect the displacement of the flexible end-posts with a sub-pixel sensitivity of 0.555 px/post (minimum increment in post displacement) and a lower limit of 1.665 px/post (minimum post displacement). Compared to our established reference for contractility assessment, Rianú had a high correlation for all parameters analyzed (ranging from R 2 = 0.7514 to R 2 = 0.9695 ), demonstrating its high accuracy and reliability. Conclusions Rianú provides simultaneous tracking of multiple hECTs, expediting the recording and analysis processes, and simplifies time-based intervention studies. It also allows data collection from different formats and has scale-up capabilities proportional to the number of tissues per field of view. These capabilities will enhance throughput of hECTs and similar assays for in-vitro analysis in disease modeling and drug screening applications.
Collapse
Affiliation(s)
- Jack F. Murphy
- Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1014, New York City, 10029, NY, USA
| | - Kevin D. Costa
- Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1014, New York City, 10029, NY, USA
| | - Irene C. Turnbull
- Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1014, New York City, 10029, NY, USA
| |
Collapse
|
206
|
Simmons DW, Schuftan DR, Ramahdita G, Huebsch N. Hydrogel-Assisted Double Molding Enables Rapid Replication of Stereolithographic 3D Prints for Engineered Tissue Design. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37200617 DOI: 10.1021/acsami.3c02279] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Tissue-engineered in vitro models are an essential tool in biomedical research. Tissue geometry is a key determinant of function, but controlling the geometry of microscale tissues remains challenging. Additive manufacturing approaches have emerged as a promising means for rapid and iterative changes in the geometry of microdevices. However, it has been shown that poly(dimethylsiloxane) (PDMS) cross-linking is often inhibited at the interface of materials printed with stereolithography. While approaches to replica mold stereolithographic three-dimensional (3D) prints have been described, these methods are inconsistent and often lead to print destruction when unsuccessful. Additionally, 3D-printed materials often leach toxic chemicals into directly molded PDMS. Here, we developed a double molding approach that allows precise replication of high-resolution stereolithographic prints into poly(dimethylsiloxane) (PDMS) elastomer, facilitating rapid design iterations and highly parallelized sample production. Inspired by lost wax casting, we used hydrogels as intermediary molds to transfer high-resolution features from high-resolution 3D prints into PDMS, while previously published work focused on enabling direct molding of PDMS onto 3D prints through the use of coatings and post-cross-linking treatments of the 3D print itself. Hydrogel mechanical properties, including cross-link density, predict replication fidelity. We demonstrate the ability of this approach to replicate a variety of shapes that would be impossible to create using photolithography techniques traditionally used to create engineered tissue designs. This method also enabled the replication of 3D-printed features into PDMS that would not be possible with direct molding as the stiffness of these materials leads to material fracture when unmolding, while the increased toughness in the hydrogels can elastically deform around complex features and maintain replication fidelity. Finally, we highlight the ability of this method to minimize the potential for toxic materials to transfer from the original 3D print into the PDMS replica, enhancing its use for biological applications. This minimization of the transfer of toxic materials has not been reported in other previously reported methods describing replication of 3D prints into PDMS, and we demonstrate its use through the creation of stem cell-derived microheart muscles. This method can also be used in future studies to understand the effects of geometry on engineered tissues and their constitutive cells.
Collapse
Affiliation(s)
- Daniel W Simmons
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- NSF Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - David R Schuftan
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Ghiska Ramahdita
- NSF Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Department of Mechanical Engineering & Materials Science, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Nathaniel Huebsch
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- NSF Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| |
Collapse
|
207
|
Ma MS, Sundaram S, Lou L, Agarwal A, Chen CS, Bifano TG. High throughput screening system for engineered cardiac tissues. Front Bioeng Biotechnol 2023; 11:1177688. [PMID: 37251575 PMCID: PMC10210164 DOI: 10.3389/fbioe.2023.1177688] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
Introduction: Three dimensional engineered cardiac tissues (3D ECTs) have become indispensable as in vitro models to assess drug cardiotoxicity, a leading cause of failure in pharmaceutical development. A current bottleneck is the relatively low throughput of assays that measure spontaneous contractile forces exerted by millimeter-scale ECTs typically recorded through precise optical measurement of deflection of the polymer scaffolds that support them. The required resolution and speed limit the field of view to at most a few ECTs at a time using conventional imaging. Methods: To balance the inherent tradeoff among imaging resolution, field of view and speed, an innovative mosaic imaging system was designed, built, and validated to sense contractile force of 3D ECTs seeded on a 96-well plate. Results: The system performance was validated through real-time, parallel contractile force monitoring for up to 3 weeks. Pilot drug testing was conducted using isoproterenol. Discussion: The described tool increases contractile force sensing throughput to 96 samples per measurement; significantly reduces cost, time and labor needed for preclinical cardiotoxicity assay using 3D ECT. More broadly, our mosaicking approach is a general way to scale up image-based screening in multi-well formats.
Collapse
Affiliation(s)
- Marshall S. Ma
- Mechanical Engineering, Boston University, Boston, MA, United States
- Photonics Center, Boston University, Boston, MA, United States
| | | | - Lihua Lou
- Mechanical and Materials Engineering, Florida International University, Miami, FL, United States
| | - Arvind Agarwal
- Mechanical and Materials Engineering, Florida International University, Miami, FL, United States
| | | | - Thomas G. Bifano
- Mechanical Engineering, Boston University, Boston, MA, United States
- Photonics Center, Boston University, Boston, MA, United States
| |
Collapse
|
208
|
Banan Sadeghian R, Ueno R, Takata Y, Kawakami A, Ma C, Araoka T, Takasato M, Yokokawa R. Cells sorted off hiPSC-derived kidney organoids coupled with immortalized cells reliably model the proximal tubule. Commun Biol 2023; 6:483. [PMID: 37142732 PMCID: PMC10160057 DOI: 10.1038/s42003-023-04862-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 04/21/2023] [Indexed: 05/06/2023] Open
Abstract
Of late, numerous microphysiological systems have been employed to model the renal proximal tubule. Yet there is lack of research on refining the functions of the proximal tubule epithelial layer-selective filtration and reabsorption. In this report, pseudo proximal tubule cells extracted from human-induced pluripotent stem cell-derived kidney organoids are combined and cultured with immortalized proximal tubule cells. It is shown that the cocultured tissue is an impervious epithelium that offers improved levels of certain transporters, extracellular matrix proteins collagen and laminin, and superior glucose transport and P-glycoprotein activity. mRNA expression levels higher than those obtained from each cell type were detected, suggesting an anomalous synergistic crosstalk between the two. Alongside, the improvements in morphological characteristics and performance of the immortalized proximal tubule tissue layer exposed, upon maturation, to human umbilical vein endothelial cells are thoroughly quantified and compared. Glucose and albumin reabsorption, as well as xenobiotic efflux rates through P-glycoprotein were all improved. The data presented abreast highlight the advantages of the cocultured epithelial layer and the non-iPSC-based bilayer. The in vitro models presented herein can be helpful in personalized nephrotoxicity studies.
Collapse
Affiliation(s)
| | - Ryohei Ueno
- Department of Micro Engineering, Kyoto University, Kyoto, 615-8540, Japan
| | - Yuji Takata
- Department of Micro Engineering, Kyoto University, Kyoto, 615-8540, Japan
| | - Akihiko Kawakami
- Department of Micro Engineering, Kyoto University, Kyoto, 615-8540, Japan
| | - Cheng Ma
- Department of Micro Engineering, Kyoto University, Kyoto, 615-8540, Japan
| | - Toshikazu Araoka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Minoru Takasato
- RIKEN Center for Biosystems Dynamics Research (BDR), Kobe, 650-0047, Japan
- Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
- Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
| | - Ryuji Yokokawa
- Department of Micro Engineering, Kyoto University, Kyoto, 615-8540, Japan.
| |
Collapse
|
209
|
Abstract
The heart is the first functional organ established during embryogenesis. Investigating heart development and disease is a fascinating and crucial field of research because cardiovascular diseases remain the leading cause of morbidity and mortality worldwide. Therefore, there is great interest in establishing in vitro models for recapitulating both physiological and pathological aspects of human heart development, tissue function and malfunction. Derived from pluripotent stem cells, a large variety of three-dimensional cardiac in vitro models have been introduced in recent years. In this At a Glance article, we discuss the available methods to generate such models, grouped according to the following classification: cardiac organoids, cardiac microtissues and engineered cardiac tissues. For these models, we provide a systematic overview of their applications for disease modeling and therapeutic development, as well as their advantages and limitations to assist scientists in choosing the most suitable model for their research purpose.
Collapse
Affiliation(s)
- Lika Drakhlis
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover 30625, Germany
- Authors for correspondence (; )
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover 30625, Germany
- Authors for correspondence (; )
| |
Collapse
|
210
|
Xiang H, Xu H, Tan B, Yi Q, Zhang X, Wang R, Chen T, Xie Q, Tian J, Zhu J. AKAP1 Regulates Mitochondrial Dynamics during the Fatty-Acid-Promoted Maturation of Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes as Indicated by Proteomics Sequencing. Int J Mol Sci 2023; 24:ijms24098112. [PMID: 37175819 PMCID: PMC10178876 DOI: 10.3390/ijms24098112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are cells with promising applications. However, their immaturity has restricted their use in cell therapy, disease modeling, and other studies. Therefore, the current study focused on inducing the maturation of CMs. We supplemented hiPSC-CMs with fatty acids (FAs) to promote their phenotypic maturity. Proteomic sequencing was performed to identify regulators critical for promoting the maturation of hiPSC-CMs. AKAP1 was found to be significantly increased in FA-treated hiPSC-CMs, and the results were verified. Therefore, we inhibited AKAP1 expression in the FA-treated cells and analyzed the outcomes. FA supplementation promoted the morphological and functional maturation of the hiPSC-CMs, which was accompanied by the development of a mitochondrial network. Proteomic analysis results revealed that AKAP1 expression was significantly higher in FA-treated hiPSC-CMs than in control cells. In addition, increased phosphorylation of the mitochondrial dynamin Drp1 and an increased mitochondrial fusion rate were found in FA-treated hiPSC-CMs. After AKAP1 was knocked down, the level of DRP1 phosphorylation in the cell was decreased, and the mitochondrial fusion rate was reduced. FA supplementation effectively promoted the maturation of hiPSC-CMs, and in these cells, AKAP1 regulated mitochondrial dynamics, possibly playing a significant role.
Collapse
Affiliation(s)
- Han Xiang
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Hao Xu
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Department of Clinical Laboratory, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Bin Tan
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Qin Yi
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Xinyuan Zhang
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Department of Clinical Laboratory, Women and Children's Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Rui Wang
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Tangtian Chen
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Qiumin Xie
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Jie Tian
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Department of Cardiovascular (Internal Medicine), Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Jing Zhu
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| |
Collapse
|
211
|
Li Q, Lin Z, Liu R, Tang X, Huang J, He Y, Sui X, Tian W, Shen H, Zhou H, Sheng H, Shi H, Xiao L, Wang X, Liu J. Multimodal charting of molecular and functional cell states via in situ electro-sequencing. Cell 2023; 186:2002-2017.e21. [PMID: 37080201 PMCID: PMC11259179 DOI: 10.1016/j.cell.2023.03.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/21/2022] [Accepted: 03/16/2023] [Indexed: 04/22/2023]
Abstract
Paired mapping of single-cell gene expression and electrophysiology is essential to understand gene-to-function relationships in electrogenic tissues. Here, we developed in situ electro-sequencing (electro-seq) that combines flexible bioelectronics with in situ RNA sequencing to stably map millisecond-timescale electrical activity and profile single-cell gene expression from the same cells across intact biological networks, including cardiac and neural patches. When applied to human-induced pluripotent stem-cell-derived cardiomyocyte patches, in situ electro-seq enabled multimodal in situ analysis of cardiomyocyte electrophysiology and gene expression at the cellular level, jointly defining cell states and developmental trajectories. Using machine-learning-based cross-modal analysis, in situ electro-seq identified gene-to-electrophysiology relationships throughout cardiomyocyte development and accurately reconstructed the evolution of gene expression profiles based on long-term stable electrical measurements. In situ electro-seq could be applicable to create spatiotemporal multimodal maps in electrogenic tissues, potentiating the discovery of cell types and gene programs responsible for electrophysiological function and dysfunction.
Collapse
Affiliation(s)
- Qiang Li
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
| | - Zuwan Lin
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Ren Liu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
| | - Xin Tang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jiahao Huang
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yichun He
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Xin Sui
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Weiwen Tian
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
| | - Hao Shen
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
| | - Haowen Zhou
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Hao Sheng
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
| | - Hailing Shi
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ling Xiao
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Cardiovascular Disease Initiative and Precision Cardiology Laboratory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Xiao Wang
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Jia Liu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA.
| |
Collapse
|
212
|
Dark N, Cosson MV, Tsansizi LI, Owen TJ, Ferraro E, Francis AJ, Tsai S, Bouissou C, Weston A, Collinson L, Abi-Gerges N, Miller PE, MacLeod KT, Ehler E, Mitter R, Harding SE, Smith JC, Bernardo AS. Generation of left ventricle-like cardiomyocytes with improved structural, functional, and metabolic maturity from human pluripotent stem cells. CELL REPORTS METHODS 2023; 3:100456. [PMID: 37159667 PMCID: PMC10163040 DOI: 10.1016/j.crmeth.2023.100456] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 01/23/2023] [Accepted: 03/25/2023] [Indexed: 05/11/2023]
Abstract
Decreased left ventricle (LV) function caused by genetic mutations or injury often leads to debilitating and fatal cardiovascular disease. LV cardiomyocytes are, therefore, a potentially valuable therapeutical target. Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) are neither homogeneous nor functionally mature, which reduces their utility. Here, we exploit cardiac development knowledge to instruct differentiation of hPSCs specifically toward LV cardiomyocytes. Correct mesoderm patterning and retinoic acid pathway blocking are essential to generate near-homogenous LV-specific hPSC-CMs (hPSC-LV-CMs). These cells transit via first heart field progenitors and display typical ventricular action potentials. Importantly, hPSC-LV-CMs exhibit increased metabolism, reduced proliferation, and improved cytoarchitecture and functional maturity compared with age-matched cardiomyocytes generated using the standard WNT-ON/WNT-OFF protocol. Similarly, engineered heart tissues made from hPSC-LV-CMs are better organized, produce higher force, and beat more slowly but can be paced to physiological levels. Together, we show that functionally matured hPSC-LV-CMs can be obtained rapidly without exposure to current maturation regimes.
Collapse
Affiliation(s)
| | | | - Lorenza I. Tsansizi
- The Francis Crick Institute, London, UK
- NHLI, Imperial College London, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Andreia S. Bernardo
- The Francis Crick Institute, London, UK
- NHLI, Imperial College London, London, UK
| |
Collapse
|
213
|
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
|
214
|
Van de Sande D, Ghasemi M, Watters T, Burton F, Pham L, Altrocchi C, Gallacher DJ, Lu H, Smith G. Does Enhanced Structural Maturity of hiPSC-Cardiomyocytes Better for the Detection of Drug-Induced Cardiotoxicity? Biomolecules 2023; 13:676. [PMID: 37189424 PMCID: PMC10135569 DOI: 10.3390/biom13040676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/03/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) are currently used following the Comprehensive in vitro Proarrhythmic Assay (CiPA) initiative and subsequent recommendations in the International Council for Harmonization (ICH) guidelines S7B and E14 Q&A, to detect drug-induced cardiotoxicity. Monocultures of hiPSC-CMs are immature compared to adult ventricular cardiomyocytes and might lack the native heterogeneous nature. We investigated whether hiPSC-CMs, treated to enhance structural maturity, are superior in detecting drug-induced changes in electrophysiology and contraction. This was achieved by comparing hiPSC-CMs cultured in 2D monolayers on the current standard (fibronectin matrix, FM), to monolayers on a coating known to promote structural maturity (CELLvo™ Matrix Plus, MM). Functional assessment of electrophysiology and contractility was made using a high-throughput screening approach involving the use of both voltage-sensitive fluorescent dyes for electrophysiology and video technology for contractility. Using 11 reference drugs, the response of the monolayer of hiPSC-CMs was comparable in the two experimental settings (FM and MM). The data showed no functionally relevant differences in electrophysiology between hiPSC-CMs in standard FM and MM, while contractility read-outs indicated an altered amplitude of contraction but not changes in time course. RNA profiling for cardiac proteins shows similarity of the RNA expression across the two forms of 2D culture, suggesting that cell-to-matrix adhesion differences may explain account for differences in contraction amplitude. The results support the view that hiPSC-CMs in both 2D monolayer FM and MM that promote structural maturity are equally effective in detecting drug-induced electrophysiological effects in functional safety studies.
Collapse
Affiliation(s)
- Dieter Van de Sande
- Global Safety Pharmacology, Nonclinical Safety, Janssen Pharmaceutical NV, B-2340 Beerse, Belgium
| | - Mohammadreza Ghasemi
- School of Cardiovascular & Metabolic Health, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
- Clyde Biosciences Limited, BioCity Scotland, Lanarkshire ML1 5UH, Scotland, UK
| | - Taylor Watters
- School of Cardiovascular & Metabolic Health, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
- Clyde Biosciences Limited, BioCity Scotland, Lanarkshire ML1 5UH, Scotland, UK
| | - Francis Burton
- School of Cardiovascular & Metabolic Health, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
- Clyde Biosciences Limited, BioCity Scotland, Lanarkshire ML1 5UH, Scotland, UK
| | - Ly Pham
- Global Safety Pharmacology, Nonclinical Safety, Janssen Pharmaceutical NV, B-2340 Beerse, Belgium
| | - Cristina Altrocchi
- Global Safety Pharmacology, Nonclinical Safety, Janssen Pharmaceutical NV, B-2340 Beerse, Belgium
| | - David J. Gallacher
- Global Safety Pharmacology, Nonclinical Safety, Janssen Pharmaceutical NV, B-2340 Beerse, Belgium
| | - Huarong Lu
- Global Safety Pharmacology, Nonclinical Safety, Janssen Pharmaceutical NV, B-2340 Beerse, Belgium
| | - Godfrey Smith
- School of Cardiovascular & Metabolic Health, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
- Clyde Biosciences Limited, BioCity Scotland, Lanarkshire ML1 5UH, Scotland, UK
| |
Collapse
|
215
|
Mao M, Qu X, Zhang Y, Gu B, Li C, Liu R, Li X, Zhu H, He J, Li D. Leaf-venation-directed cellular alignment for macroscale cardiac constructs with tissue-like functionalities. Nat Commun 2023; 14:2077. [PMID: 37045852 PMCID: PMC10097867 DOI: 10.1038/s41467-023-37716-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
Recapitulating the complex structural, mechanical, and electrophysiological properties of native myocardium is crucial to engineering functional cardiac tissues. Here, we report a leaf-venation-directed strategy that enables the compaction and remodeling of cell-hydrogel hybrids into highly aligned and densely packed organizations in predetermined patterns. This strategy contributes to interconnected tubular structures with cell alignment along the hierarchical channels. Compared to randomly-distributed cells, the engineered leaf-venation-directed-cardiac tissues from neonatal rat cardiomyocytes manifest advanced maturation and functionality as evidenced by detectable electrophysiological activity, macroscopically synchronous contractions, and upregulated maturation genes. As a demonstration, human induced pluripotent stem cell-derived leaf-venation-directed-cardiac tissues are engineered with evident structural and functional improvement over time. With the elastic scaffolds, leaf-venation-directed tissues are assembled into 3D centimeter-scale cardiac constructs with programmed mechanical properties, which can be delivered through tubing without affecting cell viability. The present strategy may generate cardiac constructs with multifaceted functionalities to meet clinical demands.
Collapse
Affiliation(s)
- Mao Mao
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, PR China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Xiaoli Qu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, PR China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Yabo Zhang
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, PR China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Bingsong Gu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, PR China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Chen Li
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, PR China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Rongzhi Liu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, PR China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Xiao Li
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, PR China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Hui Zhu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, PR China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Jiankang He
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, PR China.
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, PR China.
| | - Dichen Li
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, PR China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, PR China
| |
Collapse
|
216
|
Mu X, Gerhard-Herman MD, Zhang YS. Building Blood Vessel Chips with Enhanced Physiological Relevance. ADVANCED MATERIALS TECHNOLOGIES 2023; 8:2201778. [PMID: 37693798 PMCID: PMC10489284 DOI: 10.1002/admt.202201778] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Indexed: 09/12/2023]
Abstract
Blood vessel chips are bioengineered microdevices, consisting of biomaterials, human cells, and microstructures, which recapitulate essential vascular structure and physiology and allow a well-controlled microenvironment and spatial-temporal readouts. Blood vessel chips afford promising opportunities to understand molecular and cellular mechanisms underlying a range of vascular diseases. The physiological relevance is key to these blood vessel chips that rely on bioinspired strategies and bioengineering approaches to translate vascular physiology into artificial units. Here, we discuss several critical aspects of vascular physiology, including morphology, material composition, mechanical properties, flow dynamics, and mass transport, which provide essential guidelines and a valuable source of bioinspiration for the rational design of blood vessel chips. We also review state-of-art blood vessel chips that exhibit important physiological features of the vessel and reveal crucial insights into the biological processes and disease pathogenesis, including rare diseases, with notable implications for drug screening and clinical trials. We envision that the advances in biomaterials, biofabrication, and stem cells improve the physiological relevance of blood vessel chips, which, along with the close collaborations between clinicians and bioengineers, enable their widespread utility.
Collapse
Affiliation(s)
- Xuan Mu
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Marie Denise Gerhard-Herman
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| |
Collapse
|
217
|
Zhang G, Li W, Yu M, Huang H, Wang Y, Han Z, Shi K, Ma L, Yu Z, Zhu X, Peng Z, Xu Y, Li X, Hu S, He J, Li D, Xi Y, Lan H, Xu L, Tang M, Xiao M. Electric-Field-Driven Printed 3D Highly Ordered Microstructure with Cell Feature Size Promotes the Maturation of Engineered Cardiac Tissues. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206264. [PMID: 36782337 PMCID: PMC10104649 DOI: 10.1002/advs.202206264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/31/2023] [Indexed: 06/18/2023]
Abstract
Engineered cardiac tissues (ECTs) derived from human induced pluripotent stem cells (hiPSCs) are viable alternatives for cardiac repair, patient-specific disease modeling, and drug discovery. However, the immature state of ECTs limits their clinical utility. The microenvironment fabricated using 3D scaffolds can affect cell fate, and is crucial for the maturation of ECTs. Herein, the authors demonstrate an electric-field-driven (EFD) printed 3D highly ordered microstructure with cell feature size to promote the maturation of ECTs. The simulation and experimental results demonstrate that the EFD jet microscale 3D printing overcomes the jet repulsion without any prior requirements for both conductive and insulating substrates. Furthermore, the 3D highly ordered microstructures with a fiber diameter of 10-20 µm and spacing of 60-80 µm have been fabricated by maintaining a vertical jet, achieving the largest ratio of fiber diameter/spacing of 0.29. The hiPSCs-derived cardiomyocytes formed ordered ECTs with their sarcomere growth along the fiber and developed synchronous functional ECTs inside the 3D-printed scaffold with matured calcium handling compared to the 2D coverslip. Therefore, the EFD jet 3D microscale printing process facilitates the fabrication of scaffolds providing a suitable microenvironment to promote the maturation of ECTs, thereby showing great potential for cardiac tissue engineering.
Collapse
Affiliation(s)
- Guangming Zhang
- Shandong Engineering Research Center for Additive ManufacturingQingdao University of TechnologyQingdao266520P. R. China
| | - Wenhai Li
- Shandong Engineering Research Center for Additive ManufacturingQingdao University of TechnologyQingdao266520P. R. China
| | - Miao Yu
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated HospitalMedical CollegeSoochow UniversitySuzhou215000P. R. China
| | - Hui Huang
- Shandong Engineering Research Center for Additive ManufacturingQingdao University of TechnologyQingdao266520P. R. China
| | - Yaning Wang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated HospitalMedical CollegeSoochow UniversitySuzhou215000P. R. China
| | - Zhifeng Han
- Shandong Engineering Research Center for Additive ManufacturingQingdao University of TechnologyQingdao266520P. R. China
| | - Kai Shi
- Shandong Engineering Research Center for Additive ManufacturingQingdao University of TechnologyQingdao266520P. R. China
| | - Lingxuan Ma
- Shandong Engineering Research Center for Additive ManufacturingQingdao University of TechnologyQingdao266520P. R. China
| | - Zhihao Yu
- Shandong Engineering Research Center for Additive ManufacturingQingdao University of TechnologyQingdao266520P. R. China
| | - Xiaoyang Zhu
- Shandong Engineering Research Center for Additive ManufacturingQingdao University of TechnologyQingdao266520P. R. China
| | - Zilong Peng
- Shandong Engineering Research Center for Additive ManufacturingQingdao University of TechnologyQingdao266520P. R. China
| | - Yue Xu
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated HospitalMedical CollegeSoochow UniversitySuzhou215000P. R. China
| | - Xiaoyun Li
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated HospitalMedical CollegeSoochow UniversitySuzhou215000P. R. China
| | - Shijun Hu
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated HospitalMedical CollegeSoochow UniversitySuzhou215000P. R. China
| | - Jiankang He
- State Key Laboratory for Manufacturing System EngineeringXi'an Jiaotong UniversityXi'an710049P. R. China
| | - Dichen Li
- State Key Laboratory for Manufacturing System EngineeringXi'an Jiaotong UniversityXi'an710049P. R. China
| | - Yongming Xi
- Department of Spinal SurgeryThe Affilliated Hosepital of Qingdao UniversityQingdao266003P. R. China
| | - Hongbo Lan
- Shandong Engineering Research Center for Additive ManufacturingQingdao University of TechnologyQingdao266520P. R. China
| | - Lin Xu
- Yantai Affiliated HospitalBinzhou Medical UniversityYantai264100P. R. China
- Institute of Rehabilitation EngineeringBinzhou Medical UniversityYantai264100P. R. China
| | - Mingliang Tang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated HospitalMedical CollegeSoochow UniversitySuzhou215000P. R. China
- Co‐innovation Center of NeuroregenerationNantong UniversityNantong226001P. R. China
| | - Miao Xiao
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated HospitalMedical CollegeSoochow UniversitySuzhou215000P. R. China
| |
Collapse
|
218
|
Dou W, Daoud A, Chen X, Wang T, Malhi M, Gong Z, Mirshafiei F, Zhu M, Shan G, Huang X, Maynes JT, Sun Y. Ultrathin and Flexible Bioelectronic Arrays for Functional Measurement of iPSC-Cardiomyocytes under Cardiotropic Drug Administration and Controlled Microenvironments. NANO LETTERS 2023; 23:2321-2331. [PMID: 36893018 DOI: 10.1021/acs.nanolett.3c00017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Emerging heart-on-a-chip technology is a promising tool to establish in vitro cardiac models for therapeutic testing and disease modeling. However, due to the technical complexity of integrating cell culture chambers, biosensors, and bioreactors into a single entity, a microphysiological system capable of reproducing controlled microenvironmental cues to regulate cell phenotypes, promote iPS-cardiomyocyte maturity, and simultaneously measure the dynamic changes of cardiomyocyte function in situ is not available. This paper reports an ultrathin and flexible bioelectronic array platform in 24-well format for higher-throughput contractility measurement under candidate drug administration or defined microenvironmental conditions. In the array, carbon black (CB)-PDMS flexible strain sensors were embedded for detecting iPSC-CM contractility signals. Carbon fiber electrodes and pneumatic air channels were integrated to provide electrical and mechanical stimulation to improve iPSC-CM maturation. Performed experiments validate that the bioelectronic array accurately reveals the effects of cardiotropic drugs and identifies mechanical/electrical stimulation strategies for promoting iPSC-CM maturation.
Collapse
Affiliation(s)
- Wenkun Dou
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Abdelkader Daoud
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Xin Chen
- Program in Developmental and Stem Cell Biology and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Tiancong Wang
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Manpreet Malhi
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Zheyuan Gong
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Fatemeh Mirshafiei
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Min Zhu
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Guanqiao Shan
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Xi Huang
- Program in Developmental and Stem Cell Biology and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Jason T Maynes
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, Ontario M5S 3G4, Canada
- Department of Computer Science, University of Toronto, Toronto, Ontario M5T 3A1, Canada
| |
Collapse
|
219
|
Wang BZ, Nash TR, Zhang X, Rao J, Abriola L, Kim Y, Zakharov S, Kim M, Luo LJ, Morsink M, Liu B, Lock RI, Fleischer S, Tamargo MA, Bohnen M, Welch CL, Chung WK, Marx SO, Surovtseva YV, Vunjak-Novakovic G, Fine BM. Engineered cardiac tissue model of restrictive cardiomyopathy for drug discovery. Cell Rep Med 2023; 4:100976. [PMID: 36921598 PMCID: PMC10040415 DOI: 10.1016/j.xcrm.2023.100976] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 12/19/2022] [Accepted: 02/21/2023] [Indexed: 03/16/2023]
Abstract
Restrictive cardiomyopathy (RCM) is defined as increased myocardial stiffness and impaired diastolic relaxation leading to elevated ventricular filling pressures. Human variants in filamin C (FLNC) are linked to a variety of cardiomyopathies, and in this study, we investigate an in-frame deletion (c.7416_7418delGAA, p.Glu2472_Asn2473delinAsp) in a patient with RCM. Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) with this variant display impaired relaxation and reduced calcium kinetics in 2D culture when compared with a CRISPR-Cas9-corrected isogenic control line. Similarly, mutant engineered cardiac tissues (ECTs) demonstrate increased passive tension and impaired relaxation velocity compared with isogenic controls. High-throughput small-molecule screening identifies phosphodiesterase 3 (PDE3) inhibition by trequinsin as a potential therapy to improve cardiomyocyte relaxation in this genotype. Together, these data demonstrate an engineered cardiac tissue model of RCM and establish the translational potential of this precision medicine approach to identify therapeutics targeting myocardial relaxation.
Collapse
Affiliation(s)
- Bryan Z Wang
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Trevor R Nash
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Xiaokan Zhang
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Jenny Rao
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Laura Abriola
- Yale Center for Molecular Discovery, Yale University, New Haven, CT 06520, USA
| | - Youngbin Kim
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Sergey Zakharov
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Michael Kim
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Lori J Luo
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Margaretha Morsink
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Bohao Liu
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Roberta I Lock
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Sharon Fleischer
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Manuel A Tamargo
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Michael Bohnen
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Carrie L Welch
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Wendy K Chung
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Steven O Marx
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Yulia V Surovtseva
- Yale Center for Molecular Discovery, Yale University, New Haven, CT 06520, USA
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA; Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA; College of Dental Medicine, Columbia University, New York, NY 10032, USA
| | - Barry M Fine
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
220
|
Sesena-Rubfiaro A, Prajapati NJ, Paolino L, Lou L, Cotayo D, Pandey P, Shaver M, Hutcheson J, Agarwal A, He J. Membrane Remodeling of Human-Engineered Cardiac Tissue by Chronic Electric Stimulation. ACS Biomater Sci Eng 2023; 9:1644-1655. [PMID: 36765460 PMCID: PMC10542861 DOI: 10.1021/acsbiomaterials.2c01370] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) show immature features, but these are improved by integration into 3D cardiac constructs. In addition, it has been demonstrated that physical manipulations such as electrical stimulation (ES) are highly effective in improving the maturation of human-engineered cardiac tissue (hECT) derived from hiPSC-CMs. Here, we continuously applied an ES in capacitive coupling configuration, which is below the pacing threshold, to millimeter-sized hECTs for 1-2 weeks. Meanwhile, the structural and functional developments of the hECTs were monitored and measured using an array of assays. Of particular note, a nanoscale imaging technique, scanning ion conductance microscopy (SICM), has been used to directly image membrane remodeling of CMs at different locations on the tissue surface. Periodic crest/valley patterns with a distance close to the sarcomere length appeared on the membrane of CMs near the edge of the tissue after ES, suggesting the enhanced transverse tubulation network. The SICM observation is also supported by the fluorescence images of the transverse tubulation network and α-actinin. Correspondingly, essential cardiac functions such as calcium handling and contraction force generation were improved. Our study provides evidence that chronic subthreshold ES can still improve the structural and functional developments of hECTs.
Collapse
Affiliation(s)
| | - Navin J. Prajapati
- Department of Physics, Florida International University, Miami, FL 33199, USA
| | - Lia Paolino
- Department of Biomedical Engineering, Florida International University, Miami, FL 33199, USA
| | - Lihua Lou
- Department of Mechanical and Materials Engineering, Florida International University, Miami, FL 33174, USA
| | - Daniel Cotayo
- Department of Physics, Florida International University, Miami, FL 33199, USA
| | - Popular Pandey
- Department of Physics, Florida International University, Miami, FL 33199, USA
| | - Mohammed Shaver
- Department of Biomedical Engineering, Florida International University, Miami, FL 33199, USA
| | - Joshua Hutcheson
- Department of Biomedical Engineering, Florida International University, Miami, FL 33199, USA
- Biomolecular Science Institute, Florida International University, Miami FL 33199, USA
| | - Arvind Agarwal
- Department of Mechanical and Materials Engineering, Florida International University, Miami, FL 33174, USA
| | - Jin He
- Department of Physics, Florida International University, Miami, FL 33199, USA
- Biomolecular Science Institute, Florida International University, Miami FL 33199, USA
| |
Collapse
|
221
|
Abstract
Cardiovascular diseases are a group of heart and blood vessel disorders which remain a leading cause of morbidity and mortality worldwide. Currently, cardiovascular disease research commonly depends on in vivo rodent models and in vitro human cell culture models. Despite their widespread use in cardiovascular disease research, there are some long-standing limitations: animal models often fail to faithfully mimic human response, while traditional cell models ignore the in vivo microenvironment, intercellular communications, and tissue-tissue interactions. The convergence of microfabrication and tissue engineering has given rise to organ-on-a-chip technologies. The organ-on-a-chip is a microdevice containing microfluidic chips, cells, and extracellular matrix to reproduce the physiological processes of a certain part of the human body, and is nowadays considered a promising bridge between in vivo models and in vitro 2D or 3D cell culture models. Considering the difficulty in obtaining human vessel and heart samples, the development of vessel-on-a-chip and heart-on-a-chip systems can guide cardiovascular disease research in the future. In this review, we elaborate methods and materials to fabricate organ-on-a-chip systems and summarize the construction of vessel and heart chips. The construction of vessels-on-a-chip must consider the cyclic mechanical stretch and fluid shear stress, while hemodynamic forces and cardiomyocyte maturation are key factors in building hearts-on-a-chip. We also introduce the application of organs-on-a-chip in cardiovascular disease study.
Collapse
|
222
|
Loiben AM, Chien WM, Friedman CE, Chao LSL, Weber G, Goldstein A, Sniadecki NJ, Murry CE, Yang KC. Cardiomyocyte Apoptosis Is Associated with Contractile Dysfunction in Stem Cell Model of MYH7 E848G Hypertrophic Cardiomyopathy. Int J Mol Sci 2023; 24:4909. [PMID: 36902340 PMCID: PMC10003263 DOI: 10.3390/ijms24054909] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/16/2023] [Accepted: 02/21/2023] [Indexed: 03/08/2023] Open
Abstract
Missense mutations in myosin heavy chain 7 (MYH7) are a common cause of hypertrophic cardiomyopathy (HCM), but the molecular mechanisms underlying MYH7-based HCM remain unclear. In this work, we generated cardiomyocytes derived from isogenic human induced pluripotent stem cells to model the heterozygous pathogenic MYH7 missense variant, E848G, which is associated with left ventricular hypertrophy and adult-onset systolic dysfunction. MYH7E848G/+ increased cardiomyocyte size and reduced the maximum twitch forces of engineered heart tissue, consistent with the systolic dysfunction in MYH7E848G/+ HCM patients. Interestingly, MYH7E848G/+ cardiomyocytes more frequently underwent apoptosis that was associated with increased p53 activity relative to controls. However, genetic ablation of TP53 did not rescue cardiomyocyte survival or restore engineered heart tissue twitch force, indicating MYH7E848G/+ cardiomyocyte apoptosis and contractile dysfunction are p53-independent. Overall, our findings suggest that cardiomyocyte apoptosis is associated with the MYH7E848G/+ HCM phenotype in vitro and that future efforts to target p53-independent cell death pathways may be beneficial for the treatment of HCM patients with systolic dysfunction.
Collapse
Affiliation(s)
- Alexander M. Loiben
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98109, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA
- Department of Medicine/Cardiology, University of Washington, Seattle, WA 98109, USA
| | - Wei-Ming Chien
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98109, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA
- Department of Medicine/Cardiology, University of Washington, Seattle, WA 98109, USA
- Cardiology/Hospital Specialty Medicine, VA Puget Sound HCS, Seattle, WA 98108, USA
| | - Clayton E. Friedman
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98109, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA
- Department of Medicine/Cardiology, University of Washington, Seattle, WA 98109, USA
| | - Leslie S.-L. Chao
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98109, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA
- Department of Medicine/Cardiology, University of Washington, Seattle, WA 98109, USA
| | - Gerhard Weber
- Department of Medicine/Cardiology, University of Washington, Seattle, WA 98109, USA
| | - Alex Goldstein
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98109, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA
- Department of Medicine/Cardiology, University of Washington, Seattle, WA 98109, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98195, USA
- Department of Lab Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Nathan J. Sniadecki
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98109, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA
- Department of Medicine/Cardiology, University of Washington, Seattle, WA 98109, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98195, USA
- Department of Lab Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Charles E. Murry
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98109, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA
- Department of Medicine/Cardiology, University of Washington, Seattle, WA 98109, USA
- Department of Lab Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Kai-Chun Yang
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98109, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA
- Department of Medicine/Cardiology, University of Washington, Seattle, WA 98109, USA
- Cardiology/Hospital Specialty Medicine, VA Puget Sound HCS, Seattle, WA 98108, USA
| |
Collapse
|
223
|
Qiu J, Liu XJ, You BA, Ren N, Liu H. Application of Nanomaterials in Stem Cell-Based Therapeutics for Cardiac Repair and Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206487. [PMID: 36642861 DOI: 10.1002/smll.202206487] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/16/2022] [Indexed: 06/17/2023]
Abstract
Cardiovascular disease is a leading cause of disability and death worldwide. Although the survival rate of patients with heart diseases can be improved with contemporary pharmacological treatments and surgical procedures, none of these therapies provide a significant improvement in cardiac repair and regeneration. Stem cell-based therapies are a promising approach for functional recovery of damaged myocardium. However, the available stem cells are difficult to differentiate into cardiomyocytes, which result in the extremely low transplantation efficiency. Nanomaterials are widely used to regulate the myocardial differentiation of stem cells, and play a very important role in cardiac tissue engineering. This study discusses the current status and limitations of stem cells and cell-derived exosomes/micro RNAs based cardiac therapy, describes the cardiac repair mechanism of nanomaterials, summarizes the recent advances in nanomaterials used in cardiac repair and regeneration, and evaluates the advantages and disadvantages of the relevant nanomaterials. Besides discussing the potential clinical applications of nanomaterials in cardiac therapy, the perspectives and challenges of nanomaterials used in stem cell-based cardiac repair and regeneration are also considered. Finally, new research directions in this field are proposed, and future research trends are highlighted.
Collapse
Affiliation(s)
- Jie Qiu
- Medical Research Institute, Jinan Nanjiao Hospital, Jinan, 250002, P. R. China
| | - Xiang-Ju Liu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, 250012, P. R. China
| | - Bei-An You
- Department of Cardiovascular Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Jinan, 266035, P. R. China
| | - Na Ren
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, P. R. China
| | - Hong Liu
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, P. R. China
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, P. R. China
| |
Collapse
|
224
|
Yong U, Kim D, Kim H, Hwang DG, Cho S, Nam H, Kim S, Kim T, Jeong U, Kim K, Chung WK, Yeo WH, Jang J. Biohybrid 3D Printing of a Tissue-Sensor Platform for Wireless, Real-Time, and Continuous Monitoring of Drug-Induced Cardiotoxicity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2208983. [PMID: 36528341 DOI: 10.1002/adma.202208983] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/05/2022] [Indexed: 06/17/2023]
Abstract
Drug-induced cardiotoxicity is regarded as a major hurdle in the early stages of drug development. Although there are various methods for preclinical cardiotoxicity tests, they cannot completely predict the cardiotoxic potential of a compound due to the lack of physiological relevance. Recently, 3D engineered heart tissue (EHT) has been used to investigate cardiac muscle functions as well as pharmacological effects by exhibiting physiological auxotonic contractions. However, there is still no adequate platform for continuous monitoring to test acute and chronic pharmacological effects in vitro. Here, a biohybrid 3D printing method for fabricating a tissue-sensor platform, composed of a bipillar-grafted strain gauge sensor and EHT, is first introduced. Two pillars are three-dimensionally printed as grafts onto a strain gauge-embedded substrate to promote the EHT contractility and guide the self-assembly of the EHTs along with the strain gauge. In addition, the integration of a wireless multi-channel electronic system allows for continuous monitoring of the EHT contractile force by the tissue-sensor platform and, ultimately, for the observation of the acute and chronic drug effects of cardiotoxicants. In summary, biohybrid 3D printing technology is expected to be a potential fabrication method to provide a next-generation tissue-sensor platform for an effective drug development process.
Collapse
Affiliation(s)
- Uijung Yong
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37666, Republic of Korea
| | - Donghwan Kim
- School of Interdisciplinary Bioscience and Bioengineering, POSTECH, Pohang, 37666, Republic of Korea
| | - Hojoong Kim
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Dong Gyu Hwang
- School of Interdisciplinary Bioscience and Bioengineering, POSTECH, Pohang, 37666, Republic of Korea
| | - Sungkeon Cho
- Department of Mechanical Engineering, POSTECH, Pohang, 37673, Republic of Korea
| | - Hyoryung Nam
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37666, Republic of Korea
| | - Sejin Kim
- Department of Mechanical Engineering, POSTECH, Pohang, 37673, Republic of Korea
| | - Taeyeong Kim
- Department of Materials Science and Engineering, POSTECH, Pohang, 37673, Republic of Korea
| | - Unyong Jeong
- Department of Materials Science and Engineering, POSTECH, Pohang, 37673, Republic of Korea
| | - Keehoon Kim
- Department of Mechanical Engineering, POSTECH, Pohang, 37673, Republic of Korea
| | - Wan Kyun Chung
- Department of Mechanical Engineering, POSTECH, Pohang, 37673, Republic of Korea
| | - Woon-Hong Yeo
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, Atlanta, GA, 30332, USA
- IEN Center for Human-Centric Interfaces and Engineering, Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Jinah Jang
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37666, Republic of Korea
- School of Interdisciplinary Bioscience and Bioengineering, POSTECH, Pohang, 37666, Republic of Korea
- Department of Mechanical Engineering, POSTECH, Pohang, 37673, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, 03722, Republic of Korea
| |
Collapse
|
225
|
Jiang Y, Li Y, Zhang Y, Hu D, Zhang S, Wang C, Huang S, Zhang A, Jia Z, You R. NSC228155 alleviates septic cardiomyopathy via protecting mitochondria and inhibiting inflammation. Int Immunopharmacol 2023; 116:109847. [PMID: 36774857 DOI: 10.1016/j.intimp.2023.109847] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/12/2023]
Abstract
Septic cardiomyopathy is a lethal symptom of sepsis. Discovery of effective therapy that prevents cardiac injury in sepsis is critical in the clinical management of sepsis. NSC228155 is a novel compound with therapeutic potential on acute kidney injury by preventing apoptosis and protecting mitochondria. Whether NSC228155 protects against septic cardiomyopathy is unclear. In the present study, adult C57BL/6J mice were i.p injected with 5 mg/kg/day NSC228155 for 2 days before 10 mg/kg lipopolysaccharide (LPS) injection. Cardiac functional testing and sampling for serum and tissue were performed 12 and 24 h post LPS injection, respectively. NSC228155 significantly improved cardiac function examined by echocardiography, decreased the serum lactate dehydrogenase (LDH) and creatine kinase-MB, and pathologically alleviated cardiac injury in LPS mice. Accordingly, NSC228155 attenuated cardiomyocytes' mitochondrial damage as shown by decreased damaged mitochondrial ratio and activated signals for mitochondrial biogenesis, dynamics and mitophagy in LPS mice model. Metabolomics analysis demonstrated that NSC228155 corrected the metabolic disturbance involved in oxidative stress and energy metabolism, and decreased tissue injury metabolites in LPS-stimulated cardiac tissue. In the LPS-stimulated cardiac cell culture derived from human induced pluripotent stem cells, NSC228155 effectively restored the beating frequency, decreased LDH release, and protected mitochondria. NSC228155 also inhibited inflammation shown by decreased pro-inflammatory mediators in both serum and cardiac tissue in LPS model. Taken together, NSC228155 significantly improved cardiac function by directly preventing against cardiac cell injury and inhibiting inflammation in LPS model, hence may be a potential novel therapy against septic cardiomyopathy.
Collapse
Affiliation(s)
- Yuteng Jiang
- School of Medicine, Southeast University, Nanjing, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Yanwei Li
- School of Medicine, Southeast University, Nanjing, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Yiyuan Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Huai'an Hospital Affiliated to Xuzhou Medical University, Huai'an, Jiangsu, China
| | - Dandan Hu
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Shengnan Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Chunli Wang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Songming Huang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- School of Medicine, Southeast University, Nanjing, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.
| | - Zhanjun Jia
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.
| | - Ran You
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
226
|
Rupert C, López JE, Cortez-Toledo E, De la Cruz Cabrera O, Chesler NC, Simpson PC, Campbell SG, Baker AJ. Increased length-dependent activation of human engineered heart tissue after chronic α 1A-adrenergic agonist treatment: testing a novel heart failure therapy. Am J Physiol Heart Circ Physiol 2023; 324:H293-H304. [PMID: 36637971 PMCID: PMC9886349 DOI: 10.1152/ajpheart.00279.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 12/06/2022] [Accepted: 12/23/2022] [Indexed: 01/14/2023]
Abstract
Chronic stimulation of cardiac α1A-adrenergic receptors (α1A-ARs) improves symptoms in multiple preclinical models of heart failure. However, the translational significance remains unclear. Human engineered heart tissues (EHTs) provide a means of quantifying the effects of chronic α1A-AR stimulation on human cardiomyocyte physiology. EHTs were created from thin slices of decellularized pig myocardium seeded with human induced pluripotent stem cell (iPSC)-derived cardiomyocytes and fibroblasts. With a paired experimental design, EHTs were cultured for 3 wk, mechanically tested, cultured again for 2 wk with α1A-AR agonist A61603 (10 nM) or vehicle control, and retested after drug washout for 24 h. Separate control experiments determined the effects of EHT age (3-5 wk) or repeat mechanical testing. We found that chronic A61603 treatment caused a 25% increase of length-dependent activation (LDA) of contraction compared with vehicle treatment (n = 7/group, P = 0.035). EHT force was not increased after chronic A61603 treatment. However, after vehicle treatment, EHT force was increased by 35% relative to baseline testing (n = 7/group, P = 0.022), suggesting EHT maturation. Control experiments suggested that increased EHT force resulted from repeat mechanical testing, not from EHT aging. RNA-seq analysis confirmed that the α1A-AR is expressed in human EHTs and found chronic A61603 treatment affected gene expression in biological pathways known to be activated by α1A-ARs, including the MAP kinase signaling pathway. In conclusion, increased LDA in human EHT after chronic A61603 treatment raises the possibility that chronic stimulation of the α1A-AR might be beneficial for increasing LDA in human myocardium and might be beneficial for treating human heart failure by restoring LDA.NEW & NOTEWORTHY Chronic stimulation of α1A-adrenergic receptors (α1A-ARs) is known to mediate therapeutic effects in animal heart failure models. To investigate the effects of chronic α1A-AR stimulation in human cardiomyocytes, we tested engineered heart tissue (EHT) created with iPSC-derived cardiomyocytes. RNA-seq analysis confirmed human EHT expressed α1A-ARs. Chronic (2 wk) α1A-AR stimulation with A61603 (10 nM) increased length-dependent activation (LDA) of contraction. Chronic α1A-AR stimulation might be beneficial for treating human heart failure by restoring LDA.
Collapse
Affiliation(s)
- C. Rupert
- Propria LLC, Branford, Connecticut, United States
| | - J. E. López
- Division of Cardiovascular Medicine, Department of Internal Medicine,
University of California Davis, Davis, California, United States
| | - E. Cortez-Toledo
- Division of Cardiovascular Medicine, Department of Internal Medicine,
University of California Davis, Davis, California, United States
| | | | - N. C. Chesler
- Edwards Lifesciences Foundation Cardiovascular Innovation Research Center, Irvine, California, United States
- Department of Biomedical Engineering, University of California, Irvine, California, United States
| | - P. C. Simpson
- Cardiology Division, Veterans Affairs Medical Center, San Francisco, California, United States
- Department of Medicine, University of California, San Francisco, California, United States
| | - S. G. Campbell
- Departments of Biomedical Engineering and Cellular and Molecular Physiology, Yale University, New Haven, Connecticut, United States
| | - A. J. Baker
- Cardiology Division, Veterans Affairs Medical Center, San Francisco, California, United States
- Department of Medicine, University of California, San Francisco, California, United States
| |
Collapse
|
227
|
Wang C, Ramahdita G, Genin G, Huebsch N, Ma Z. Dynamic mechanobiology of cardiac cells and tissues: Current status and future perspective. BIOPHYSICS REVIEWS 2023; 4:011314. [PMID: 37008887 PMCID: PMC10062054 DOI: 10.1063/5.0141269] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/08/2023] [Indexed: 03/31/2023]
Abstract
Mechanical forces impact cardiac cells and tissues over their entire lifespan, from development to growth and eventually to pathophysiology. However, the mechanobiological pathways that drive cell and tissue responses to mechanical forces are only now beginning to be understood, due in part to the challenges in replicating the evolving dynamic microenvironments of cardiac cells and tissues in a laboratory setting. Although many in vitro cardiac models have been established to provide specific stiffness, topography, or viscoelasticity to cardiac cells and tissues via biomaterial scaffolds or external stimuli, technologies for presenting time-evolving mechanical microenvironments have only recently been developed. In this review, we summarize the range of in vitro platforms that have been used for cardiac mechanobiological studies. We provide a comprehensive review on phenotypic and molecular changes of cardiomyocytes in response to these environments, with a focus on how dynamic mechanical cues are transduced and deciphered. We conclude with our vision of how these findings will help to define the baseline of heart pathology and of how these in vitro systems will potentially serve to improve the development of therapies for heart diseases.
Collapse
Affiliation(s)
| | - Ghiska Ramahdita
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, USA
| | | | | | - Zhen Ma
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
228
|
Zhao Y, Wang EY, Lai FBL, Cheung K, Radisic M. Organs-on-a-chip: a union of tissue engineering and microfabrication. Trends Biotechnol 2023; 41:410-424. [PMID: 36725464 PMCID: PMC9985977 DOI: 10.1016/j.tibtech.2022.12.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 02/03/2023]
Abstract
We review the emergence of the new field of organ-on-a-chip (OOAC) engineering, from the parent fields of tissue engineering and microfluidics. We place into perspective the tools and capabilities brought into the OOAC field by early tissue engineers and microfluidics experts. Liver-on-a-chip and heart-on-a-chip are used as two case studies of systems that heavily relied on tissue engineering techniques and that were amongst the first OOAC models to be implemented, motivated by the need to better assess toxicity to human tissues in preclinical drug development. We review current challenges in OOAC that often stem from the same challenges in the parent fields, such as stable vascularization and drug absorption.
Collapse
Affiliation(s)
- Yimu Zhao
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada; Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Erika Yan Wang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Fook B L Lai
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Krisco Cheung
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada; Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada.
| |
Collapse
|
229
|
Brown AL, Gerosa FM, Wang J, Hsiai T, Marsden AL. Recent advances in quantifying the mechanobiology of cardiac development via computational modeling. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2023; 25:100428. [PMID: 36583220 PMCID: PMC9794182 DOI: 10.1016/j.cobme.2022.100428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Mechanical forces are essential for coordinating cardiac morphogenesis, but much remains to be discovered about the interactions between mechanical forces and the mechanotransduction pathways they activate. Due to the elaborate and fundamentally multi-physics and multi-scale nature of cardiac mechanobiology, a complete understanding requires multiple experimental and analytical techniques. We identify three fundamental tools used in the field to probe these interactions: high resolution imaging, genetic and molecular analysis, and computational modeling. In this review, we focus on computational modeling and present recent studies employing this tool to investigate the mechanobiological pathways involved with cardiac development. These works demonstrate that understanding the detailed spatial and temporal patterns of biomechanical forces is crucial to building a comprehensive understanding of mechanobiology during cardiac development, and that computational modeling is an effective and efficient tool for obtaining such detail. In this context, multidisciplinary studies combining all three tools present the most compelling results.
Collapse
Affiliation(s)
- Aaron L. Brown
- Stanford University, Department of Mechanical Engineering, Stanford, USA, CA, 94305
| | - Fannie M. Gerosa
- Stanford University, Department of Pediatrics, Stanford, USA, CA 94305
- Stanford University, Institute for Computational & Mathematical Engineering, Stanford, USA, CA 94305
| | - Jing Wang
- University of California Los Angeles, Department of Bioengineering, Los Angeles, CA 90095
| | - Tzung Hsiai
- University of California Los Angeles, Department of Bioengineering, Los Angeles, CA 90095
- University of California Los Angeles, Division of Cardiology, Los Angeles, CA 90095
| | - Alison L. Marsden
- Stanford University, Department of Mechanical Engineering, Stanford, USA, CA, 94305
- Stanford University, Department of Pediatrics, Stanford, USA, CA 94305
- Stanford University, Institute for Computational & Mathematical Engineering, Stanford, USA, CA 94305
- Stanford University, Department of Bioengineering, Stanford, USA, CA 94305
| |
Collapse
|
230
|
Small Extracellular Vesicles Derived from Induced Pluripotent Stem Cells in the Treatment of Myocardial Injury. Int J Mol Sci 2023; 24:ijms24054577. [PMID: 36902008 PMCID: PMC10003569 DOI: 10.3390/ijms24054577] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 03/02/2023] Open
Abstract
Induced pluripotent stem cell (iPSC) therapy brings great hope to the treatment of myocardial injuries, while extracellular vesicles may be one of the main mechanisms of its action. iPSC-derived small extracellular vesicles (iPSCs-sEVs) can carry genetic and proteinaceous substances and mediate the interaction between iPSCs and target cells. In recent years, more and more studies have focused on the therapeutic effect of iPSCs-sEVs in myocardial injury. IPSCs-sEVs may be a new cell-free-based treatment for myocardial injury, including myocardial infarction, myocardial ischemia-reperfusion injury, coronary heart disease, and heart failure. In the current research on myocardial injury, the extraction of sEVs from mesenchymal stem cells induced by iPSCs was widely used. Isolation methods of iPSCs-sEVs for the treatment of myocardial injury include ultracentrifugation, isodensity gradient centrifugation, and size exclusion chromatography. Tail vein injection and intraductal administration are the most widely used routes of iPSCs-sEV administration. The characteristics of sEVs derived from iPSCs which were induced from different species and organs, including fibroblasts and bone marrow, were further compared. In addition, the beneficial genes of iPSC can be regulated through CRISPR/Cas9 to change the composition of sEVs and improve the abundance and expression diversity of them. This review focused on the strategies and mechanisms of iPSCs-sEVs in the treatment of myocardial injury, which provides a reference for future research and the application of iPSCs-sEVs.
Collapse
|
231
|
Abstract
Heart disease is a significant burden on global health care systems and is a leading cause of death each year. To improve our understanding of heart disease, high quality disease models are needed. These will facilitate the discovery and development of new treatments for heart disease. Traditionally, researchers have relied on 2D monolayer systems or animal models of heart disease to elucidate pathophysiology and drug responses. Heart-on-a-chip (HOC) technology is an emerging field where cardiomyocytes among other cell types in the heart can be used to generate functional, beating cardiac microtissues that recapitulate many features of the human heart. HOC models are showing great promise as disease modeling platforms and are poised to serve as important tools in the drug development pipeline. By leveraging advances in human pluripotent stem cell-derived cardiomyocyte biology and microfabrication technology, diseased HOCs are highly tuneable and can be generated via different approaches such as: using cells with defined genetic backgrounds (patient-derived cells), adding small molecules, modifying the cells' environment, altering cell ratio/composition of microtissues, among others. HOCs have been used to faithfully model aspects of arrhythmia, fibrosis, infection, cardiomyopathies, and ischemia, to name a few. In this review, we highlight recent advances in disease modeling using HOC systems, describing instances where these models outperformed other models in terms of reproducing disease phenotypes and/or led to drug development.
Collapse
Affiliation(s)
- Omar Mourad
- Toronto General Hospital Research Institute (O.M., R.Y., M.L., S.S.N.), University Health Network, Toronto, Canada
- Institute of Biomedical Engineering (O.M., R.Y., M.L., S.S.N.), University of Toronto, Canada
| | - Ryan Yee
- Toronto General Hospital Research Institute (O.M., R.Y., M.L., S.S.N.), University Health Network, Toronto, Canada
- Institute of Biomedical Engineering (O.M., R.Y., M.L., S.S.N.), University of Toronto, Canada
| | - Mengyuan Li
- Toronto General Hospital Research Institute (O.M., R.Y., M.L., S.S.N.), University Health Network, Toronto, Canada
- Institute of Biomedical Engineering (O.M., R.Y., M.L., S.S.N.), University of Toronto, Canada
| | - Sara S Nunes
- Toronto General Hospital Research Institute (O.M., R.Y., M.L., S.S.N.), University Health Network, Toronto, Canada
- Ajmera Transplant Center (S.S.N.), University Health Network, Toronto, Canada
- Institute of Biomedical Engineering (O.M., R.Y., M.L., S.S.N.), University of Toronto, Canada
- Department of Laboratory Medicine and Pathobiology (S.S.N.), University of Toronto, Canada
- Heart and Stroke/Richard Lewar Centre of Excellence (S.S.N.), University of Toronto, Canada
| |
Collapse
|
232
|
Singh BN, Yucel D, Garay BI, Tolkacheva EG, Kyba M, Perlingeiro RCR, van Berlo JH, Ogle BM. Proliferation and Maturation: Janus and the Art of Cardiac Tissue Engineering. Circ Res 2023; 132:519-540. [PMID: 36795845 PMCID: PMC9943541 DOI: 10.1161/circresaha.122.321770] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
During cardiac development and morphogenesis, cardiac progenitor cells differentiate into cardiomyocytes that expand in number and size to generate the fully formed heart. Much is known about the factors that regulate initial differentiation of cardiomyocytes, and there is ongoing research to identify how these fetal and immature cardiomyocytes develop into fully functioning, mature cells. Accumulating evidence indicates that maturation limits proliferation and conversely proliferation occurs rarely in cardiomyocytes of the adult myocardium. We term this oppositional interplay the proliferation-maturation dichotomy. Here we review the factors that are involved in this interplay and discuss how a better understanding of the proliferation-maturation dichotomy could advance the utility of human induced pluripotent stem cell-derived cardiomyocytes for modeling in 3-dimensional engineered cardiac tissues to obtain truly adult-level function.
Collapse
Affiliation(s)
- Bhairab N. Singh
- Department of Pediatrics, University of Minnesota, MN, USA
- Department of Biomedical Engineering, University of Minnesota, MN, USA
- Stem Cell Institute, University of Minnesota, MN, USA
| | - Dogacan Yucel
- Stem Cell Institute, University of Minnesota, MN, USA
- Department of Medicine, Cardiovascular Division, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
| | - Bayardo I. Garay
- Stem Cell Institute, University of Minnesota, MN, USA
- Department of Medicine, Cardiovascular Division, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
- Medical Scientist Training Program, University of Minnesota Medical School, MN, USA
| | - Elena G. Tolkacheva
- Department of Biomedical Engineering, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
- Institute for Engineering in Medicine, University of Minnesota, MN, USA
| | - Michael Kyba
- Department of Pediatrics, University of Minnesota, MN, USA
- Stem Cell Institute, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
| | - Rita C. R. Perlingeiro
- Stem Cell Institute, University of Minnesota, MN, USA
- Department of Medicine, Cardiovascular Division, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
| | - Jop H. van Berlo
- Stem Cell Institute, University of Minnesota, MN, USA
- Department of Medicine, Cardiovascular Division, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
| | - Brenda M. Ogle
- Department of Pediatrics, University of Minnesota, MN, USA
- Department of Biomedical Engineering, University of Minnesota, MN, USA
- Stem Cell Institute, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
- Institute for Engineering in Medicine, University of Minnesota, MN, USA
- Masonic Cancer Center, University of Minnesota, MN, USA
| |
Collapse
|
233
|
Tan Y, Chen Y, Lu T, Witman N, Yan B, Gong Y, Ai X, Yang L, Liu M, Luo R, Wang H, Ministrini S, Dong W, Wang W, Fu W. Engineering a conduction-consistent cardiac patch with rGO/PLCL electrospun nanofibrous membranes and human iPSC-derived cardiomyocytes. Front Bioeng Biotechnol 2023; 11:1094397. [PMID: 36845196 PMCID: PMC9944832 DOI: 10.3389/fbioe.2023.1094397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/25/2023] [Indexed: 02/10/2023] Open
Abstract
The healthy human heart has special directional arrangement of cardiomyocytes and a unique electrical conduction system, which is critical for the maintenance of effective contractions. The precise arrangement of cardiomyocytes (CMs) along with conduction consistency between CMs is essential for enhancing the physiological accuracy of in vitro cardiac model systems. Here, we prepared aligned electrospun rGO/PLCL membranes using electrospinning technology to mimic the natural heart structure. The physical, chemical and biocompatible properties of the membranes were rigorously tested. We next assembled human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) on electrospun rGO/PLCL membranes in order to construct a myocardial muscle patch. The conduction consistency of cardiomyocytes on the patches were carefully recorded. We found that cells cultivated on the electrospun rGO/PLCL fibers presented with an ordered and arranged structure, excellent mechanical properties, oxidation resistance and effective guidance. The addition of rGO was found to be beneficial for the maturation and synchronous electrical conductivity of hiPSC-CMs within the cardiac patch. This study verified the possibility of using conduction-consistent cardiac patches to enhance drug screening and disease modeling applications. Implementation of such a system could one day lead to in vivo cardiac repair applications.
Collapse
Affiliation(s)
- Yao Tan
- Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Chen
- Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tingting Lu
- Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Nevin Witman
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Bingqian Yan
- Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yiqi Gong
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xuefeng Ai
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Yang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Minglu Liu
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Runjiao Luo
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huijing Wang
- Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland,Department of Medicine and Surgery, Internal Medicine, Angiology and Atherosclerosis, University of Perugia, Perugia, Italy
| | - Wei Dong
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,*Correspondence: Wei Dong, ; Wei Wang, ; Wei Fu,
| | - Wei Wang
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,*Correspondence: Wei Dong, ; Wei Wang, ; Wei Fu,
| | - Wei Fu
- Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,*Correspondence: Wei Dong, ; Wei Wang, ; Wei Fu,
| |
Collapse
|
234
|
Paz-Artigas L, Montero-Calle P, Iglesias-García O, Mazo MM, Ochoa I, Ciriza J. Current approaches for the recreation of cardiac ischaemic environment in vitro. Int J Pharm 2023; 632:122589. [PMID: 36623742 DOI: 10.1016/j.ijpharm.2023.122589] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/14/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023]
Abstract
Myocardial ischaemia is one of the leading dead causes worldwide. Although animal experiments have historically provided a wealth of information, animal models are time and money consuming, and they usually miss typical human patient's characteristics associated with ischemia prevalence, including aging and comorbidities. Generating reliable in vitro models that recapitulate the human cardiac microenvironment during an ischaemic event can boost the development of new drugs and therapeutic strategies, as well as our understanding of the underlying cellular and molecular events, helping the optimization of therapeutic approaches prior to animal and clinical testing. Although several culture systems have emerged for the recreation of cardiac physiology, mimicking the features of an ischaemic heart tissue in vitro is challenging and certain aspects of the disease process remain poorly addressed. Here, current in vitro cardiac culture systems used for modelling cardiac ischaemia, from self-aggregated organoids to scaffold-based constructs and heart-on-chip platforms are described. The advantages of these models to recreate ischaemic hallmarks such as oxygen gradients, pathological alterations of mechanical strength or fibrotic responses are highlighted. The new models represent a step forward to be considered, but unfortunately, we are far away from recapitulating all complexity of the clinical situations.
Collapse
Affiliation(s)
- Laura Paz-Artigas
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Pilar Montero-Calle
- Regenerative Medicine Program, Cima Universidad de Navarra, and Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Olalla Iglesias-García
- Regenerative Medicine Program, Cima Universidad de Navarra, and Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Manuel M Mazo
- Regenerative Medicine Program, Cima Universidad de Navarra, and Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Hematology and Cell Therapy, Clínica Universidad de Navarra, and Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Ignacio Ochoa
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain; CIBER-BBN, ISCIII, Zaragoza, Spain.
| | - Jesús Ciriza
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain; CIBER-BBN, ISCIII, Zaragoza, Spain.
| |
Collapse
|
235
|
Wu P, Sai X, Li Z, Ye X, Jin L, Liu G, Li G, Yang P, Zhao M, Zhu S, Liu N, Zhu P. Maturation of induced pluripotent stem cell-derived cardiomyocytes and its therapeutic effect on myocardial infarction in mouse. Bioact Mater 2023; 20:286-305. [PMID: 35702609 PMCID: PMC9167678 DOI: 10.1016/j.bioactmat.2022.05.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 11/30/2022] Open
Abstract
Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) have an irreplaceable role in the treatment of myocardial infarction (MI), which can be injected into the transplanted area with new cardiomyocytes (Cardiomyocytes, CMs), and improve myocardial function. However, the immaturity of the structure and function of iPSC-CMs is the main bottleneck at present. Since collagen participates in the formation of extracellular matrix (ECM), we synthesized nano colloidal gelatin (Gel) with collagen as the main component, and confirmed that the biomaterial has good biocompatibility and is suitable for cellular in vitro growth. Subsequently, we combined the PI3K/AKT/mTOR pathway inhibitor BEZ-235 with Gel and found that the two combined increased the sarcomere length and action potential amplitude (APA) of iPSC-CMs, and improved the Ca2+ processing ability, the maturation of mitochondrial morphological structure and metabolic function. Not only that, Gel can also prolong the retention rate of iPSC-CMs in the myocardium and increase the expression of Cx43 and angiogenesis in the transplanted area of mature iPSC-CMs, which also provides a reliable basis for the subsequent treatment of mature iPSC-CMs. BEZ-235 + Gel promotes the maturation of sarcomere structure in iPSC-CMs. BEZ-235 + Gel promotes electrophysiological maturation of iPSC-CMs. BEZ-235 + Gel increases mitochondrial respiration in iPSC-CMs. Gel loaded with mature iPSC-CMs enhanced angiogenesis and gap junction formation at the injection site.
Collapse
|
236
|
Abstract
PURPOSE OF REVIEW To summarize and critically assess the contribution of genetics to the Long QT Syndrome (LQTS), with specific reference to the unraveling of its underlying mechanisms and to its impact on clinical practice. RECENT FINDINGS The evolution towards our current approach to therapy for LQTS patients is examined in terms of risk stratification, gene-specific management, and assessment of the clinical impact that genetic modifiers may have in modulating the natural history of the patients. Glimpses are provided on the newest multidisciplinary approaches to study disease mechanisms, test new candidate drugs and identify precision treatments. SUMMARY It is undeniable that genetics has revolutionized our mechanistic understanding of cardiac channelopathies. Its impact has been enormous but, curiously, the way LQTS patients are being treated today is largely the same that was used in the pregenetic era, even though management has been refined and gene-specific differences allow a more individually tailored antiarrhythmic protection. The synergy of genetic findings with modern in vitro and in silico tools may expand precision treatments; however, they will need to prove more effective than the current therapeutic approaches and equally safe.
Collapse
Affiliation(s)
- Peter J Schwartz
- Istituto Auxologico Italiano IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics
| | - Luca Sala
- Istituto Auxologico Italiano IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics.,Department of Biotechnology and Biosciences, University of Milano - Bicocca, Milan, Italy
| |
Collapse
|
237
|
Loiben AM, Chien WM, Friedman CE, Chao LSL, Weber G, Goldstein A, Sniadecki N, Murry CE, Yang KC. Cardiomyocyte apoptosis contributes to contractile dysfunction in stem cell model of MYH7 E848G hypertrophic cardiomyopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.24.525458. [PMID: 36747800 PMCID: PMC9900838 DOI: 10.1101/2023.01.24.525458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Missense mutations in myosin heavy chain 7 ( MYH7 ) are a common cause of hyper-trophic cardiomyopathy (HCM), but the molecular mechanisms underlying MYH7 -based HCM remain unclear. In this work, we generated cardiomyocytes derived from isogenic human induced pluripotent stem cells to model the heterozygous pathogenic MYH7 missense variant, E848G, which is associated with left ventricular hypertrophy and adultonset systolic dysfunction. MYH7 E848G/+ increased cardiomyocyte size and reduced the maximum twitch forces of engineered heart tissue, consistent with the systolic dysfunction in MYH7 E848G HCM patients. Interestingly, MYH7 E848G/+ cardiomyocytes more frequently underwent apoptosis that was associated with increased p53 activity relative to controls. However, genetic ablation of TP53 did not rescue cardiomyocyte survival or restore engineered heart tissue twitch force, indicating MYH7 E848G/+ cardiomyocyte apoptosis and contractile dysfunction are p53-independent. Overall, our findings suggest that cardiomyocyte apoptosis plays an important role in the MYH7 E848G/+ HCM phenotype in vitro and that future efforts to target p53-independent cell death pathways may be beneficial for the treatment of HCM patients with systolic dysfunction.
Collapse
Affiliation(s)
- Alexander M. Loiben
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
- Department of Medicine/Cardiology, University of Washington, Seattle, WA, USA
| | - Wei-Ming Chien
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
- Department of Medicine/Cardiology, University of Washington, Seattle, WA, USA
- Cardiology/Hospital Specialty Medicine, VA Puget Sound HCS, Seattle, WA, USA
| | - Clayton E. Friedman
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
- Department of Medicine/Cardiology, University of Washington, Seattle, WA, USA
| | - Leslie S-L. Chao
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
- Department of Medicine/Cardiology, University of Washington, Seattle, WA, USA
| | - Gerhard Weber
- Department of Medicine/Cardiology, University of Washington, Seattle, WA, USA
| | - Alex Goldstein
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
- Department of Medicine/Cardiology, University of Washington, Seattle, WA, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
- Department of Lab Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Nathan Sniadecki
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
- Department of Medicine/Cardiology, University of Washington, Seattle, WA, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Lab Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Charles E. Murry
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
- Department of Medicine/Cardiology, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Lab Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Kai-Chun Yang
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
- Department of Medicine/Cardiology, University of Washington, Seattle, WA, USA
- Cardiology/Hospital Specialty Medicine, VA Puget Sound HCS, Seattle, WA, USA
| |
Collapse
|
238
|
iPSC-Derived Cardiomyocytes in Inherited Cardiac Arrhythmias: Pathomechanistic Discovery and Drug Development. Biomedicines 2023; 11:biomedicines11020334. [PMID: 36830871 PMCID: PMC9953535 DOI: 10.3390/biomedicines11020334] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 01/20/2023] [Accepted: 01/22/2023] [Indexed: 01/26/2023] Open
Abstract
With the discovery of induced pluripotent stem cell (iPSCs) a wide range of cell types, including iPSC-derived cardiomyocytes (iPSC-CM), can now be generated from an unlimited source of somatic cells. These iPSC-CM are used for different purposes such as disease modelling, drug discovery, cardiotoxicity testing and personalised medicine. The 2D iPSC-CM models have shown promising results, but they are known to be more immature compared to in vivo adult cardiomyocytes. Novel approaches to create 3D models with the possible addition of other (cardiac) cell types are being developed. This will not only improve the maturity of the cells, but also leads to more physiologically relevant models that more closely resemble the human heart. In this review, we focus on the progress in the modelling of inherited cardiac arrhythmias in both 2D and 3D and on the use of these models in therapy development and drug testing.
Collapse
|
239
|
Brimmer S, Ji P, Birla AK, Keswani SG, Caldarone CA, Birla RK. Recent advances in biological pumps as a building block for bioartificial hearts. Front Bioeng Biotechnol 2023; 11:1061622. [PMID: 36741765 PMCID: PMC9895798 DOI: 10.3389/fbioe.2023.1061622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 01/04/2023] [Indexed: 01/22/2023] Open
Abstract
The field of biological pumps is a subset of cardiac tissue engineering and focused on the development of tubular grafts that are designed generate intraluminal pressure. In the simplest embodiment, biological pumps are tubular grafts with contractile cardiomyocytes on the external surface. The rationale for biological pumps is a transition from planar 3D cardiac patches to functional biological pumps, on the way to complete bioartificial hearts. Biological pumps also have applications as a standalone device, for example, to support the Fontan circulation in pediatric patients. In recent years, there has been a lot of progress in the field of biological pumps, with innovative fabrication technologies. Examples include the use of cell sheet engineering, self-organized heart muscle, bioprinting and in vivo bio chambers for vascularization. Several materials have been tested for biological pumps and included resected aortic segments from rodents, type I collagen, and fibrin hydrogel, to name a few. Multiple bioreactors have been tested to condition biological pumps and replicate the complex in vivo environment during controlled in vitro culture. The purpose of this article is to provide an overview of the field of the biological pumps, outlining progress in the field over the past several years. In particular, different fabrication methods, biomaterial platforms for tubular grafts and examples of bioreactors will be presented. In addition, we present an overview of some of the challenges that need to be overcome for the field of biological pumps to move forward.
Collapse
Affiliation(s)
- Sunita Brimmer
- Laboratory for Regenerative Tissue Repair, Texas Children’s Hospital, Houston, TX, United States,Center for Congenital Cardiac Research, Texas Children’s Hospital, Houston, TX, United States,Division of Congenital Heart Surgery, Texas Children’s Hospital, Houston, TX, United States
| | - Pengfei Ji
- Laboratory for Regenerative Tissue Repair, Texas Children’s Hospital, Houston, TX, United States,Center for Congenital Cardiac Research, Texas Children’s Hospital, Houston, TX, United States,Division of Congenital Heart Surgery, Texas Children’s Hospital, Houston, TX, United States
| | - Aditya K. Birla
- Laboratory for Regenerative Tissue Repair, Texas Children’s Hospital, Houston, TX, United States,Center for Congenital Cardiac Research, Texas Children’s Hospital, Houston, TX, United States
| | - Sundeep G. Keswani
- Laboratory for Regenerative Tissue Repair, Texas Children’s Hospital, Houston, TX, United States,Center for Congenital Cardiac Research, Texas Children’s Hospital, Houston, TX, United States,Department of Surgery, Baylor College of Medicine, Houston, TX, United States,Division of Pediatric Surgery, Department of Surgery, Texas Children’s Hospital, Houston, TX, United States
| | - Christopher A. Caldarone
- Center for Congenital Cardiac Research, Texas Children’s Hospital, Houston, TX, United States,Division of Congenital Heart Surgery, Texas Children’s Hospital, Houston, TX, United States,Department of Surgery, Baylor College of Medicine, Houston, TX, United States,Division of Pediatric Surgery, Department of Surgery, Texas Children’s Hospital, Houston, TX, United States
| | - Ravi K. Birla
- Laboratory for Regenerative Tissue Repair, Texas Children’s Hospital, Houston, TX, United States,Center for Congenital Cardiac Research, Texas Children’s Hospital, Houston, TX, United States,Division of Congenital Heart Surgery, Texas Children’s Hospital, Houston, TX, United States,Department of Surgery, Baylor College of Medicine, Houston, TX, United States,Division of Pediatric Surgery, Department of Surgery, Texas Children’s Hospital, Houston, TX, United States,*Correspondence: Ravi K. Birla,
| |
Collapse
|
240
|
Ernst P, Bidwell PA, Dora M, Thomas DD, Kamdar F. Cardiac calcium regulation in human induced pluripotent stem cell cardiomyocytes: Implications for disease modeling and maturation. Front Cell Dev Biol 2023; 10:986107. [PMID: 36742199 PMCID: PMC9889838 DOI: 10.3389/fcell.2022.986107] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 12/23/2022] [Indexed: 01/19/2023] Open
Abstract
Human induced pluripotent stem cell cardiomyocytes (hiPSC-CMs) are based on ground-breaking technology that has significantly impacted cardiovascular research. They provide a renewable source of human cardiomyocytes for a variety of applications including in vitro disease modeling and drug toxicity testing. Cardiac calcium regulation plays a critical role in the cardiomyocyte and is often dysregulated in cardiovascular disease. Due to the limited availability of human cardiac tissue, calcium handling and its regulation have most commonly been studied in the context of animal models. hiPSC-CMs can provide unique insights into human physiology and pathophysiology, although a remaining limitation is the relative immaturity of these cells compared to adult cardiomyocytes Therefore, this field is rapidly developing techniques to improve the maturity of hiPSC-CMs, further establishing their place in cardiovascular research. This review briefly covers the basics of cardiomyocyte calcium cycling and hiPSC technology, and will provide a detailed description of our current understanding of calcium in hiPSC-CMs.
Collapse
Affiliation(s)
- Patrick Ernst
- Cardiovascular Division, University of Minnesota, Minneapolis, MN, United States
| | - Philip A. Bidwell
- Cardiovascular Division, University of Minnesota, Minneapolis, MN, United States
| | - Michaela Dora
- College of Biological Sciences, University of Minnesota, Minneapolis, MN, United States
| | - David D. Thomas
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - Forum Kamdar
- Cardiovascular Division, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
241
|
Whole-Heart Tissue Engineering and Cardiac Patches: Challenges and Promises. BIOENGINEERING (BASEL, SWITZERLAND) 2023; 10:bioengineering10010106. [PMID: 36671678 PMCID: PMC9855348 DOI: 10.3390/bioengineering10010106] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/02/2023] [Accepted: 01/05/2023] [Indexed: 01/15/2023]
Abstract
Despite all the advances in preventing, diagnosing, and treating cardiovascular disorders, they still account for a significant part of mortality and morbidity worldwide. The advent of tissue engineering and regenerative medicine has provided novel therapeutic approaches for the treatment of various diseases. Tissue engineering relies on three pillars: scaffolds, stem cells, and growth factors. Gene and cell therapy methods have been introduced as primary approaches to cardiac tissue engineering. Although the application of gene and cell therapy has resulted in improved regeneration of damaged cardiac tissue, further studies are needed to resolve their limitations, enhance their effectiveness, and translate them into the clinical setting. Scaffolds from synthetic, natural, or decellularized sources have provided desirable characteristics for the repair of cardiac tissue. Decellularized scaffolds are widely studied in heart regeneration, either as cell-free constructs or cell-seeded platforms. The application of human- or animal-derived decellularized heart patches has promoted the regeneration of heart tissue through in vivo and in vitro studies. Due to the complexity of cardiac tissue engineering, there is still a long way to go before cardiac patches or decellularized whole-heart scaffolds can be routinely used in clinical practice. This paper aims to review the decellularized whole-heart scaffolds and cardiac patches utilized in the regeneration of damaged cardiac tissue. Moreover, various decellularization methods related to these scaffolds will be discussed.
Collapse
|
242
|
Wang J, An M, Haubner BJ, Penninger JM. Cardiac regeneration: Options for repairing the injured heart. Front Cardiovasc Med 2023; 9:981982. [PMID: 36712238 PMCID: PMC9877631 DOI: 10.3389/fcvm.2022.981982] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 12/29/2022] [Indexed: 01/13/2023] Open
Abstract
Cardiac regeneration is one of the grand challenges in repairing injured human hearts. Numerous studies of signaling pathways and metabolism on cardiac development and disease pave the way for endogenous cardiomyocyte regeneration. New drug delivery approaches, high-throughput screening, as well as novel therapeutic compounds combined with gene editing will facilitate the development of potential cell-free therapeutics. In parallel, progress has been made in the field of cell-based therapies. Transplantation of human pluripotent stem cell (hPSC)-derived cardiomyocytes (hPSC-CMs) can partially rescue the myocardial defects caused by cardiomyocyte loss in large animals. In this review, we summarize current cell-based and cell-free regenerative therapies, discuss the importance of cardiomyocyte maturation in cardiac regenerative medicine, and envision new ways of regeneration for the injured heart.
Collapse
Affiliation(s)
- Jun Wang
- Department of Medical Genetics, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Meilin An
- Department of Medical Genetics, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Bernhard Johannes Haubner
- Department of Internal Medicine III (Cardiology and Angiology), Innsbruck Medical University, Innsbruck, Austria
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Josef M. Penninger
- Department of Medical Genetics, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, VBC – Vienna BioCenter, Vienna, Austria
| |
Collapse
|
243
|
Gabetti S, Sileo A, Montrone F, Putame G, Audenino AL, Marsano A, Massai D. Versatile electrical stimulator for cardiac tissue engineering-Investigation of charge-balanced monophasic and biphasic electrical stimulations. Front Bioeng Biotechnol 2023; 10:1031183. [PMID: 36686253 PMCID: PMC9846083 DOI: 10.3389/fbioe.2022.1031183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 12/16/2022] [Indexed: 01/05/2023] Open
Abstract
The application of biomimetic physical stimuli replicating the in vivo dynamic microenvironment is crucial for the in vitro development of functional cardiac tissues. In particular, pulsed electrical stimulation (ES) has been shown to improve the functional properties of in vitro cultured cardiomyocytes. However, commercially available electrical stimulators are expensive and cumbersome devices while customized solutions often allow limited parameter tunability, constraining the investigation of different ES protocols. The goal of this study was to develop a versatile compact electrical stimulator (ELETTRA) for biomimetic cardiac tissue engineering approaches, designed for delivering controlled parallelizable ES at a competitive cost. ELETTRA is based on an open-source micro-controller running custom software and is combinable with different cell/tissue culture set-ups, allowing simultaneously testing different ES patterns on multiple samples. In particular, customized culture chambers were appositely designed and manufactured for investigating the influence of monophasic and biphasic pulsed ES on cardiac cell monolayers. Finite element analysis was performed for characterizing the spatial distributions of the electrical field and the current density within the culture chamber. Performance tests confirmed the accuracy, compliance, and reliability of the ES parameters delivered by ELETTRA. Biological tests were performed on neonatal rat cardiac cells, electrically stimulated for 4 days, by comparing, for the first time, the monophasic waveform (electric field = 5 V/cm) to biphasic waveforms by matching either the absolute value of the electric field variation (biphasic ES at ±2.5 V/cm) or the total delivered charge (biphasic ES at ±5 V/cm). Findings suggested that monophasic ES at 5 V/cm and, particularly, charge-balanced biphasic ES at ±5 V/cm were effective in enhancing electrical functionality of stimulated cardiac cells and in promoting synchronous contraction.
Collapse
Affiliation(s)
- Stefano Gabetti
- Department of Mechanical and Aerospace Engineering and PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy
| | - Antonio Sileo
- Department of Surgery and Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Federica Montrone
- Department of Mechanical and Aerospace Engineering and PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy
| | - Giovanni Putame
- Department of Mechanical and Aerospace Engineering and PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy
| | - Alberto L. Audenino
- Department of Mechanical and Aerospace Engineering and PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy
| | - Anna Marsano
- Department of Surgery and Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Diana Massai
- Department of Mechanical and Aerospace Engineering and PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy,*Correspondence: Diana Massai,
| |
Collapse
|
244
|
Zhang Y, Yu T, Ding J, Li Z. Bone-on-a-chip platforms and integrated biosensors: Towards advanced in vitro bone models with real-time biosensing. Biosens Bioelectron 2023; 219:114798. [PMID: 36257118 DOI: 10.1016/j.bios.2022.114798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 08/25/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
Abstract
Bone diseases, such as osteoporosis and bone defects, often lead to structural and functional deformities of the patient's body. Understanding the complicated pathophysiology and finding new drugs for bone diseases are in dire need but challenging with the conventional cell and animal models. Bone-on-a-chip (BoC) models recapitulate key features of bone at an unprecedented level and can potentially shift the paradigm of future bone research and therapeutic development. Nevertheless, current BoC models predominantly rely on off-chip analysis which provides only endpoint measurements. To this end, integrating biosensors within the BoC can provide non-invasive, continuous monitoring of the experiment progression, significantly facilitating bone research. This review aims to summarize research progress in BoC and biosensor integrations and share perspectives on this exciting but rudimentary research area. We first introduce the research progress of BoC models in the study of bone remodeling and bone diseases, respectively. We then summarize the need for BoC characterization and reported works on biosensor integration in organ chips. Finally, we discuss the limitations and future directions of BoC models and biosensor integrations as next-generation technologies for bone research.
Collapse
Affiliation(s)
- Yang Zhang
- School of Dentistry, Health Science Center, Shenzhen University, Shenzhen, 518060, China; School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Taozhao Yu
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Jingyi Ding
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Zida Li
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China.
| |
Collapse
|
245
|
Min S, Cho SW. Engineered human cardiac tissues for modeling heart diseases. BMB Rep 2023; 56:32-42. [PMID: 36443005 PMCID: PMC9887099 DOI: 10.5483/bmbrep.2022-0185] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 07/30/2023] Open
Abstract
Heart disease is one of the major life-threatening diseases with high mortality and incidence worldwide. Several model systems, such as primary cells and animals, have been used to understand heart diseases and establish appropriate treatments. However, they have limitations in accuracy and reproducibility in recapitulating disease pathophysiology and evaluating drug responses. In recent years, three-dimensional (3D) cardiac tissue models produced using tissue engineering technology and human cells have outperformed conventional models. In particular, the integration of cell reprogramming techniques with bioengineering platforms (e.g., microfluidics, scaffolds, bioprinting, and biophysical stimuli) has facilitated the development of heart-ona- chip, cardiac spheroid/organoid, and engineered heart tissue (EHT) to recapitulate the structural and functional features of the native human heart. These cardiac models have improved heart disease modeling and toxicological evaluation. In this review, we summarize the cell types for the fabrication of cardiac tissue models, introduce diverse 3D human cardiac tissue models, and discuss the strategies to enhance their complexity and maturity. Finally, recent studies in the modeling of various heart diseases are reviewed. [BMB Reports 2023; 56(1): 32-42].
Collapse
Affiliation(s)
- Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Korea
| |
Collapse
|
246
|
Martyniak A, Jeż M, Dulak J, Stępniewski J. Adaptation of cardiomyogenesis to the generation and maturation of cardiomyocytes from human pluripotent stem cells. IUBMB Life 2023; 75:8-29. [PMID: 36263833 DOI: 10.1002/iub.2685] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 10/05/2022] [Indexed: 12/29/2022]
Abstract
The advent of methods for efficient generation and cardiac differentiation of pluripotent stem cells opened new avenues for disease modelling, drug testing, and cell therapies of the heart. However, cardiomyocytes (CM) obtained from such cells demonstrate an immature, foetal-like phenotype that involves spontaneous contractions, irregular morphology, expression of embryonic isoforms of sarcomere components, and low level of ion channels. These and other features may affect cellular response to putative therapeutic compounds and the efficient integration into the host myocardium after in vivo delivery. Therefore, novel strategies to increase the maturity of pluripotent stem cell-derived CM are of utmost importance. Several approaches have already been developed relying on molecular changes that occur during foetal and postnatal maturation of the heart, its electromechanical activity, and the cellular composition. As a better understanding of these determinants may facilitate the generation of efficient protocols for in vitro acquisition of an adult-like phenotype by immature CM, this review summarizes the most important molecular factors that govern CM during embryonic development, postnatal changes that trigger heart maturation, as well as protocols that are currently used to generate mature pluripotent stem cell-derived cardiomyocytes.
Collapse
Affiliation(s)
- Alicja Martyniak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Mateusz Jeż
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Jacek Stępniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
247
|
Min S, Cho SW. Engineered human cardiac tissues for modeling heart diseases. BMB Rep 2023; 56:32-42. [PMID: 36443005 PMCID: PMC9887099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 01/28/2023] Open
Abstract
Heart disease is one of the major life-threatening diseases with high mortality and incidence worldwide. Several model systems, such as primary cells and animals, have been used to understand heart diseases and establish appropriate treatments. However, they have limitations in accuracy and reproducibility in recapitulating disease pathophysiology and evaluating drug responses. In recent years, three-dimensional (3D) cardiac tissue models produced using tissue engineering technology and human cells have outperformed conventional models. In particular, the integration of cell reprogramming techniques with bioengineering platforms (e.g., microfluidics, scaffolds, bioprinting, and biophysical stimuli) has facilitated the development of heart-ona- chip, cardiac spheroid/organoid, and engineered heart tissue (EHT) to recapitulate the structural and functional features of the native human heart. These cardiac models have improved heart disease modeling and toxicological evaluation. In this review, we summarize the cell types for the fabrication of cardiac tissue models, introduce diverse 3D human cardiac tissue models, and discuss the strategies to enhance their complexity and maturity. Finally, recent studies in the modeling of various heart diseases are reviewed. [BMB Reports 2023; 56(1): 32-42].
Collapse
Affiliation(s)
- Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Korea
| |
Collapse
|
248
|
Cai L, Wang R, Zhang D. Cardiac Disease Modeling with Engineered Heart Tissue. Handb Exp Pharmacol 2023; 281:235-255. [PMID: 37563250 DOI: 10.1007/164_2023_681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
The rhythmically beating heart is the foundation of life-sustaining blood flow. There are four chambers and many different types of cell in the heart, but the twisted myofibrillar structures formed by cardiomyocytes are particularly important for cardiac contraction and electrical impulse transmission properties. The ability to generate cardiomyocytes using human-induced pluripotent stem cells has essentially solved the cell supply shortage for in vitro simulation of cardiac tissue function; however, modeling heart at the tissue level needs mature myocardial structure, electrophysiology, and contractile characteristics. Here, the current research on human functionalized cardiac microtissue in modeling cardiac diseases is reviewed and the design criteria and practical applications of different human engineered heart tissues, including cardiac organoids, cardiac thin films, and cardiac microbundles are analyzed. Table summarizing the ability of several in vitro myocardial models to assess heart structure and function for cardiac disease modeling.
Collapse
Affiliation(s)
- Lin Cai
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, Hubei, China
| | - Ruxiang Wang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, Hubei, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, Hubei, China.
| |
Collapse
|
249
|
Ji S, Tu W, Huang C, Chen Z, Ren X, He B, Ding X, Chen Y, Xie X. The Aurora Kinase Inhibitor CYC116 Promotes the Maturation of Cardiomyocytes Derived from Human Pluripotent Stem Cells. Mol Cells 2022; 45:923-934. [PMID: 36572561 PMCID: PMC9794550 DOI: 10.14348/molcells.2022.0077] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/18/2022] [Accepted: 09/20/2022] [Indexed: 12/28/2022] Open
Abstract
Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have great potential in applications such as regenerative medicine, cardiac disease modeling, and in vitro drug evaluation. However, hPSC-CMs are immature, which limits their applications. During development, the maturation of CMs is accompanied by a decline in their proliferative capacity. This phenomenon suggests that regulating the cell cycle may facilitate the maturation of hPSC-CMs. Aurora kinases are essential kinases that regulate the cell cycle, the role of which is not well studied in hPSC-CM maturation. Here, we demonstrate that CYC116, an inhibitor of Aurora kinases, significantly promotes the maturation of CMs derived from both human embryonic stem cells (H1 and H9) and iPSCs (induced PSCs) (UC013), resulting in increased expression of genes related to cardiomyocyte function, better organization of the sarcomere, increased sarcomere length, increased number of mitochondria, and enhanced physiological function of the cells. In addition, a number of other Aurora kinase inhibitors have also been found to promote the maturation of hPSC-CMs. Our data suggest that blocking aurora kinase activity and regulating cell cycle progression may promote the maturation of hPSC-CMs.
Collapse
Affiliation(s)
- Sijia Ji
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- State Key Laboratory of Drug Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wanzhi Tu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- State Key Laboratory of Drug Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chenwen Huang
- State Key Laboratory of Drug Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ziyang Chen
- State Key Laboratory of Drug Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinyue Ren
- State Key Laboratory of Drug Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bingqing He
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- State Key Laboratory of Drug Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyan Ding
- Stem Cell Bank/Stem Cell Core Facility, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuelei Chen
- Stem Cell Bank/Stem Cell Core Facility, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xin Xie
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- State Key Laboratory of Drug Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
250
|
Barndt RJ, Liu Q, Tang Y, Haugh MP, Cui J, Chan SY, Wu H. Metabolic Maturation Exaggerates Abnormal Calcium Handling in a Lamp2 Knockout Human Pluripotent Stem Cell-Derived Cardiomyocyte Model of Danon Disease. Biomolecules 2022; 13:69. [PMID: 36671453 PMCID: PMC9855424 DOI: 10.3390/biom13010069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 12/27/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023] Open
Abstract
Danon disease (DD) is caused by mutations of the gene encoding lysosomal-associated membrane protein type 2 (LAMP2), which lead to impaired autophagy, glycogen accumulation, and cardiac hypertrophy. However, it is not well understood why a large portion of DD patients develop arrhythmia and sudden cardiac death. In the current study, we generated LAMP2 knockout (KO) human iPSC-derived cardiomyocytes (CM), which mimic the LAMP2 dysfunction in DD heart. Morphologic analysis demonstrated the sarcomere disarrangement in LAMP2 KO CMs. In functional studies, LAMP2 KO CMs showed near-normal calcium handling at base level. However, treatment of pro-maturation medium (MM) exaggerated the disease phenotype in the KO cells as they exhibited impaired calcium recycling and increased irregular beating events, which recapitulates the pro-arrhythmia phenotypes of DD patients. Further mechanistic study confirmed that MM treatment significantly enhanced the autophagic stress in the LAMP2 KO CMs, which was accompanied by an increase of both cellular and mitochondrial reactive oxygen species (ROS) levels. Excess ROS accumulation in LAMP2 KO CMs resulted in the over-activation of calcium/calmodulin dependent protein kinase IIδ (CaMKIIδ) and arrhythmogenesis, which was partially rescued by the treatment of ROS scavenger. In summary, our study has revealed ROS induced CaMKIIδ overactivation as a key mechanism that promotes cardiac arrhythmia in DD patients.
Collapse
Affiliation(s)
- Robert J. Barndt
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Qing Liu
- Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA
- Center for Human Genetics, Clemson University, Greenwood, SC 29646, USA
| | - Ying Tang
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Michael P. Haugh
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15213, USA
| | - Jeffery Cui
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Stephen Y. Chan
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Haodi Wu
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15213, USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|