1
|
Onozuka J, Taira R, Kadota S, Ichimura H, Shiba Y, Patra C, Ohnuma K. Synchronous beating between xenografted human cardiomyocytes and host zebrafish embryonic hearts. Biochem Biophys Res Commun 2025; 769:151933. [PMID: 40347622 DOI: 10.1016/j.bbrc.2025.151933] [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: 04/23/2025] [Revised: 04/30/2025] [Accepted: 05/01/2025] [Indexed: 05/14/2025]
Abstract
Injured human hearts are fibrotic, whereas zebrafish hearts functionally regenerate following myocardial injury. The unique regeneration niche microenvironment has been extensively studied in zebrafish hearts. However whether this can be extrapolated to humans remains unclear owing to significant species differences. We xenografted human induced pluripotent stem cell-derived cardiomyocytes (hiCMs) into the cardiac region of one-day post-fertilized zebrafish embryos and established a zebrafish xenograft model of hiCMs. This model can be used to explore the behavior of hiCMs transplanted into zebrafish hearts. Fluctuations in the fluorescence intensity of the genetically encoded calcium indicator protein GCaMP indicated that the donor hiCMs were beating. We analyzed the synchronization of the GCaMP + hiCMs transplanted into the zebrafish heart. We found synchronous beating between the host and 40 % of the zebrafish hearts with beating GCaMP-hiPSCs. Our chimeric heart model has the potential to bridge the regeneration capacity gap between zebrafish and humans and has proming future applications.
Collapse
Affiliation(s)
- Jo Onozuka
- Department of Science of Technology Innovation, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan
| | - Riko Taira
- Department of Materials Science and Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan
| | - Shin Kadota
- Department of Regenerative Science and Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan; Institute for Biomedical Sciences, Shinshu University, Matsumoto, 390-8621, Japan
| | - Hajime Ichimura
- Department of Regenerative Science and Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan; Division of Cardiovascular Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | - Yuji Shiba
- Department of Regenerative Science and Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan; Institute for Biomedical Sciences, Shinshu University, Matsumoto, 390-8621, Japan
| | - Chinmoy Patra
- Department of Developmental Biology, Agharkar Research Institute, Pune, 411004, India
| | - Kiyoshi Ohnuma
- Department of Science of Technology Innovation, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan; Department of Materials Science and Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan.
| |
Collapse
|
2
|
Montague EC, Ozcan B, Sefton E, Wulkan F, Alibhai FJ, Laflamme MA. Human pluripotent stem cell-based cardiac repair: Lessons learned and challenges ahead. Adv Drug Deliv Rev 2025; 222:115594. [PMID: 40334814 DOI: 10.1016/j.addr.2025.115594] [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: 11/04/2024] [Revised: 05/01/2025] [Accepted: 05/03/2025] [Indexed: 05/09/2025]
Abstract
The transplantation of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) and hPSC-derived cardiac progenitors (hPSC-CPs) represents a promising strategy for regenerating hearts damaged by myocardial infarction (MI). After nearly two decades of experience testing these cell populations in various small- and large-animal MI models, multiple clinical trials have recently been initiated. In this review, we consider the principal lessons learned from preclinical experience with hPSC-CMs and -CPs, focusing on three conclusions that have been supported by the majority of reported transplantation studies. First, hPSC-CMs and -CPs stably engraft in injured hearts and partially remuscularize the infarct scar, but more progress is needed to improve graft cell retention and survival. Second, the transplantation of hPSC-CMs and -CPs has been found to improve contractile function in infarcted hearts, but the mechanistic basis for these effects remains incompletely elucidated. Third, the graft tissue formed by these cells can integrate and activate synchronously with host myocardium, but this capacity for electromechanical integration has been associated with an elevated risk of graft-related arrhythmias. Here, we summarize the preclinical evidence supporting these three observations, identify the relevant gaps and barriers to translation, and summarize ongoing efforts to improve the safety and efficacy of hPSC-CM- and -CP-based regenerative therapies.
Collapse
Affiliation(s)
- E Coulter Montague
- Department of Biomedical Engineering, University of Toronto, ON, Canada; McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Bilgehan Ozcan
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Elana Sefton
- Department of Biomedical Engineering, University of Toronto, ON, Canada; McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Fanny Wulkan
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Faisal J Alibhai
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Michael A Laflamme
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada; Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada; Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
3
|
Forghani P, Liu W, Wang Z, Ling Z, Takaesu F, Yang E, Tharp GK, Nielsen S, Doraisingam S, Countryman S, Davis ME, Wu R, Jia S, Xu C. Spaceflight alters protein levels and gene expression associated with stress response and metabolic characteristics in human cardiac spheroids. Biomaterials 2025; 317:123080. [PMID: 39809079 PMCID: PMC11788069 DOI: 10.1016/j.biomaterials.2024.123080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025]
Abstract
Cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) possess tremendous advantage for cardiac regeneration. However, cell survival is challenging upon cell transplantation. Since microgravity can profoundly affect cellular properties, we investigated the effect of spaceflight on hiPSC-CMs. Cardiac spheroids derived from hiPSCs were transported to the International Space Station (ISS) via the SpaceX Crew-8 mission and cultured under space microgravity for 8 days. Beating cardiac spheroids were observed on the ISS and upon successful experimentation by the astronauts in space, the live cultures were returned to Earth. These cells had normal displacement (an indicator of contraction) and Ca2+ transient parameters in 3D live cell imaging. Proteomic analysis revealed that spaceflight upregulated many proteins involved in metabolism (n = 90), cellular component of mitochondrion (n = 62) and regulation of proliferation (n = 10). Specific metabolic pathways enriched by spaceflight included glutathione metabolism, biosynthesis of amino acids, and pyruvate metabolism. In addition, the top upregulated proteins in spaceflight samples included those involved in cellular stress response, cell survival, and metabolism. Transcriptomic profiles indicated that spaceflight upregulated genes associated with cardiomyocyte development, and cellular components of cardiac structure and mitochondrion. Furthermore, spaceflight upregulated genes in metabolic pathways associated with cell survival such as glycerophospholipid metabolism and glycerolipid metabolism. These findings indicate that short-term exposure of 3D hiPSC-CMs to the space environment led to significant changes in protein levels and gene expression involved in cell survival and metabolism.
Collapse
Affiliation(s)
- Parvin Forghani
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Wenhao Liu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Zeyu Wang
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
| | - Zhi Ling
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Felipe Takaesu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Evan Yang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Gregory K Tharp
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | | | | | | | - Michael E Davis
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Ronghu Wu
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
| | - Shu Jia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Chunhui Xu
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
| |
Collapse
|
4
|
Hong Y, Liu J, Wang W, Li H, Kong W, Li X, Zhang W, Pahlavan S, Tang YD, Wang X, Wang K. Pluripotent stem cell-derived cardiomyocyte transplantation: marching from bench to bedside. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2801-x. [PMID: 40418524 DOI: 10.1007/s11427-024-2801-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/12/2024] [Indexed: 05/27/2025]
Abstract
Cardiovascular diseases such as myocardial infarction, heart failure, and cardiomyopathy, persist as a leading global cause of death. Current treatment options have inherent limitations, particularly in terms of cardiac regeneration due to the limited regenerative capacity of adult human hearts. The transplantation of pluripotent stem cell-derived cardiomyocytes (PSC-CMs) has emerged as a promising and potential solution to address this challenge. This review aims to summarize the latest advancements and prospects of PSC-CM transplantation (PCT), along with the existing constraints, such as immune rejection and engraftment arrhythmias, and corresponding solutions. Encompassing a comprehensive range from fundamental research findings and preclinical experiments to ongoing clinical trials, we hope to offer insights into PCT from bench to bedside.
Collapse
Affiliation(s)
- Yi Hong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Center for Non-coding RNA Medicine, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing, 100191, China
- Department of Education, Peking University First Hospital, Peking University, Beijing, 100035, China
| | - Jiarui Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Center for Non-coding RNA Medicine, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing, 100191, China
| | - Weixuan Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Center for Non-coding RNA Medicine, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing, 100191, China
| | - Hao Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Center for Non-coding RNA Medicine, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing, 100191, China
| | - Weijing Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Center for Non-coding RNA Medicine, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing, 100191, China
| | - Xiaoxia Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Center for Non-coding RNA Medicine, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing, 100191, China
| | - Wei Zhang
- TianXinFu (Beijing) Medical Appliance Co., Ltd., Beijing, 102200, China
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, The Academic Center for Education, Culture and Research, Tehran, 14155-4364, Iran
| | - Yi-da Tang
- Department of Cardiology and Institute of Vascular Medicine, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Third Hospital, Beijing, 100191, China.
| | - Xi Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Center for Non-coding RNA Medicine, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing, 100191, China.
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China.
- Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China.
| | - Kai Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Center for Non-coding RNA Medicine, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing, 100191, China.
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China.
| |
Collapse
|
5
|
Gibbs CE, Boyle PM. Population-based computational simulations elucidate mechanisms of focal arrhythmia following stem cell injection. J Mol Cell Cardiol 2025; 204:5-16. [PMID: 40280466 DOI: 10.1016/j.yjmcc.2025.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 03/02/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025]
Abstract
Following a myocardial infarction (MI), a large portion of ventricular cells are replaced by scar, leading to adverse structural remodeling and heart failure. The use of stem cell-derived cardiomyocytes has shown promise in restoring cardiac function in animal models following an MI but leads to rapid focal ventricular tachycardia (VT). The VT in these animals can be variable, and its underlying mechanisms remain unknown. In this study, we used three distinct computational models derived from histological images of post-MI non-human primate ventricles to understand how human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) grafts can affect focal VT individually and synergistically. Specifically, we explored whether grafts could work cooperatively to create new arrhythmia and if geometric features such as graft tortuosity, area, host isolation, and amount of surrounding scar inhibited or enhanced the effect. We observed at least one instance of graft-host excitation (GHE) for eleven of the twenty-five individual grafts examined. Since we used a stochastic population-of-models-based approach to generate graft boundaries, we found that the number of configurations with GHE varied from graft to graft. We also examined grafts in aggregate and found that the high prevalence of GHE when all grafts were included arose from combinations of individually arrhythmogenic grafts (i.e., the overall increase in arrhythmogenicity resulted from graft complementarity rather than graft cooperativity). Further analysis of graft spatial features showed that arrhythmogenic grafts tend to be in areas with high host isolation (i.e., spatially confined regions of surviving myocardium interdigitated with engrafted cells) and when graft area and tortuosity were also high. These insights can aid in the design of novel injection schemes that could result in safer therapy for patients.
Collapse
Affiliation(s)
- Chelsea E Gibbs
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Patrick M Boyle
- Department of Bioengineering, University of Washington, Seattle, WA, USA; Division of Cardiology, University of Washington School of Medicine, Seattle, WA, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA; eScience Institute, University of Washington, Seattle, WA, USA.
| |
Collapse
|
6
|
Generali M, Kehl D, Meier D, Zorndt D, Atrott K, Saito H, Emmert MY, Hoerstrup SP. Generation and purification of iPSC-derived cardiomyocytes for clinical applications. Stem Cell Res Ther 2025; 16:189. [PMID: 40251664 PMCID: PMC12008852 DOI: 10.1186/s13287-025-04319-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/07/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Over the past decade, the field of cell therapy has rapidly expanded with the aim to replace and repair damaged cells and/or tissue. Depending on the disease many different cell types can be used as part of such a therapy. Here we focused on the potential treatment of myocardial infarction, where currently available treatment options are not able to regenerate the loss of healthy heart tissue. METHOD We generated good manufacturing practice (GMP)-compatible cardiomyocytes (iCMs) from transgene- and xenofree induced pluripotent stem cells (iPSCs) that can be seamless adapted for clinical applications. Further protocols were established for replating and freezing/thawing iCMs under xenofree conditions. RESULTS iCMs showed a cardiac phenotype, with the expression of specific cardiac markers and absence of pluripotency markers at RNA and protein level. To ensure a pure iCMs population for in vivo applications, we minimized risks of iPSC contamination using RNA-switch technology to ensure safety. CONCLUSION We describe the generation and further processing of xeno- and transgene-free iCMs. The use of GMP-compliant differentiation protocols ab initio facilitates the clinical translation of this project in later stages.
Collapse
Affiliation(s)
- M Generali
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.
| | - D Kehl
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - D Meier
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - D Zorndt
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - K Atrott
- Center for Surgical Research, University of Zurich, University Hospital Zurich, Zurich, Switzerland
| | - H Saito
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, 113-0032, Japan
| | - M Y Emmert
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité Universitätsmedizin Berlin, Berlin, Germany
| | - S P Hoerstrup
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Wyss Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
7
|
Mallya AS, Burrows T, Hsieh J, Louwagie T, Dutton J, Ogle B, Hubel A. DMSO-free cryopreservation of hiPSC-derived cardiomyocytes: Low temperature characterization and protocol development. RESEARCH SQUARE 2025:rs.3.rs-5183739. [PMID: 40321769 PMCID: PMC12047977 DOI: 10.21203/rs.3.rs-5183739/v1] [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: 05/11/2025]
Abstract
Background Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have attracted significant interest for use in disease modeling, drug discovery and potential therapeutic applications. However, conventional hiPSC-CM cryopreservation protocols largely use dimethyl sulfoxide (DMSO) as the cryoprotectant (CPA), which is linked with a loss of post-thaw recovery and function for various cell types and is not ideal for therapeutic protocols. Additionally, the effect of freezing parameters such as cooling rate and nucleation temperature on post-thaw recovery of hiPSC-CMs has not been explored. Methods hiPSC-CMs were generated by Wnt pathway inhibition, followed by sodium I-lactate purification. Subsequently, biophysical characterization of the cells was performed. A differential evolution (DE) algorithm was utilized to determine the optimal composition of a mixture of a sugar, sugar alcohol and amino acid to replace DMSO as the CPA. The hiPSC-CMs were subjected to controlled-rate freezing at different cooling rates and nucleation temperatures. The optimum freezing parameters were identified by post-thaw recoveries and the partitioning ratio obtained from low temperature Raman spectroscopy studies. The post-thaw osmotic behavior of hiPSC-CMs was studied by measuring diameter of cells resuspended in the isotonic culture medium over time. Immunocytochemistry and calcium transient studies were performed to evaluate post-thaw function. Results hiPSC-CMs were found to be slightly larger than hiPSCs and exhibited a large osmotically inactive volume. The best-performing DMSO-free solutions enabled post-thaw recoveries over 90%, which was significantly greater than DMSO (69.4 ± 6.4%). A rapid cooling rate of 5°C/min and a low nucleation temperature of -8°C was found to be optimal for hiPSC-CMs. hiPSC-CMs displayed anomalous osmotic behavior post-thaw, dropping sharply in volume after resuspension. Post-thaw function was preserved when hiPSC-CMs were frozen with the best-performing DMSO-free CPA or DMSO and the cells displayed similar cardiac markers pre-freeze and post-thaw. Conclusions It was shown that a CPA cocktail of naturally-occurring osmolytes could effectively replace DMSO for preserving hiPSC-CMs while preserving morphology and function. Understanding the anomalous osmotic behavior and managing the excessive dehydration of hiPSC-CMs could be crucial to improve post-thaw outcomes. Effective DMSO-free cryopreservation would accelerate the development of drug discovery and therapeutic applications of hiPSC-CMs.
Collapse
|
8
|
Silver SE, Howells AR, Arhontoulis DC, Randolph LN, Hyams NA, Barrs RW, Li M, Kerr CM, Robino RA, Morningstar JE, Bain JD, Floy ME, Norris RA, Bao X, Ruddy JM, Palecek SP, Ferreira LMR, Lian XL, Mei Y. Hypoimmunogenic hPSC-derived cardiac organoids for immune evasion and heart repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.09.648007. [PMID: 40291708 PMCID: PMC12027337 DOI: 10.1101/2025.04.09.648007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Human pluripotent stem cell (hPSC)-derived cardiac therapies hold great promise for heart regeneration but face major translational barriers due to allogeneic immune rejection. Here, we engineered hypoimmunogenic hPSCs using a two-step CRISPR-Cas9 strategy: (1) B2M knockout, eliminating HLA class I surface expression, and (2) knock-in of HLA-E or HLA-G trimer constructs in the AAVS1 safe harbor locus to confer robust immune evasion. Hypoimmunogenic hPSCs maintained pluripotency, efficiently differentiated into cardiac cell types that resisted both T and NK cell-mediated cytotoxicity in vitro , and self-assembled into engineered cardiac organoids. Comprehensive analyses of the hypoimmunogenic cells and organoids revealed preservation of transcriptomic, structural, and functional properties with minimal off-target effects from gene editing. In vivo , hypoimmunogenic cardiac organoids restored contractile function in infarcted rat hearts and demonstrated superior graft retention and immune evasion in humanized mice compared to wild-type counterparts. These findings establish the therapeutic potential of hypoimmunogenic hPSC-CMs in the cardiac organoid platform, laying the foundation for off-the-shelf cardiac cell therapies to treat cardiovascular disease, the leading cause of death worldwide.
Collapse
|
9
|
Xia X, Hu M, Zhou W, Jin Y, Yao X. Engineering cardiology with miniature hearts. Mater Today Bio 2025; 31:101505. [PMID: 39911371 PMCID: PMC11795835 DOI: 10.1016/j.mtbio.2025.101505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/28/2024] [Accepted: 01/18/2025] [Indexed: 02/07/2025] Open
Abstract
Cardiac organoids offer sophisticated 3D structures that emulate key aspects of human heart development and function. This review traces the evolution of cardiac organoid technology, from early stem cell differentiation protocols to advanced bioengineering approaches. We discuss the methodologies for creating cardiac organoids, including self-organization techniques, biomaterial-based scaffolds, 3D bioprinting, and organ-on-chip platforms, which have significantly enhanced the structural complexity and physiological relevance of in vitro cardiac models. We examine their applications in fundamental research and medical innovations, highlighting their potential to transform our understanding of cardiac biology and pathology. The integration of multiple cell types, vascularization strategies, and maturation protocols has led to more faithful representations of the adult human heart. However, challenges remain in achieving full functional maturity and scalability. We critically assess the current limitations and outline future directions for advancing cardiac organoid technology. By providing a comprehensive analysis of the field, this review aims to catalyze further innovation in cardiac tissue engineering and facilitate its translation to clinical applications.
Collapse
Affiliation(s)
- Xiaojun Xia
- Department of Cardiology, Center of Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Miner Hu
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310000, China
| | - Wenyan Zhou
- School of Medicine, Taizhou University, Taizhou, Zhejiang, 318000, China
| | - Yunpeng Jin
- Department of Cardiology, Center of Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Xudong Yao
- Department of Cardiology, Center of Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| |
Collapse
|
10
|
Cao C, Yang L, Song J, Liu Z, Li H, Li L, Fu J, Liu J. Cardiomyocyte regeneration after infarction: changes, opportunities and challenges. Mol Cell Biochem 2025:10.1007/s11010-025-05251-w. [PMID: 40097887 DOI: 10.1007/s11010-025-05251-w] [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: 11/14/2024] [Accepted: 03/08/2025] [Indexed: 03/19/2025]
Abstract
Myocardial infarction is a cardiovascular disease that poses a serious threat to human health. The traditional view is that adult mammalian cardiomyocytes have almost no regenerative ability, but recent studies have shown that they have regenerative potential under specific conditions. This article comprehensively describes the research progress of post-infarction cardiomyocyte regeneration, including the characteristics of cardiomyocytes and post-infarction changes, regeneration mechanisms, influencing factors, potential therapeutic strategies, challenges and future development directions, and deeply discusses the specific pathways and targets included in the regeneration mechanism, aiming to provide new ideas and methods for the treatment of myocardial infarction.
Collapse
Affiliation(s)
- Ce Cao
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Lili Yang
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Jianshu Song
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Zixin Liu
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Haoran Li
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Lei Li
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Jianhua Fu
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Jianxun Liu
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China.
| |
Collapse
|
11
|
Jebran AF, Seidler T, Tiburcy M, Daskalaki M, Kutschka I, Fujita B, Ensminger S, Bremmer F, Moussavi A, Yang H, Qin X, Mißbach S, Drummer C, Baraki H, Boretius S, Hasenauer C, Nette T, Kowallick J, Ritter CO, Lotz J, Didié M, Mietsch M, Meyer T, Kensah G, Krüger D, Sakib MS, Kaurani L, Fischer A, Dressel R, Rodriguez-Polo I, Stauske M, Diecke S, Maetz-Rensing K, Gruber-Dujardin E, Bleyer M, Petersen B, Roos C, Zhang L, Walter L, Kaulfuß S, Yigit G, Wollnik B, Levent E, Roshani B, Stahl-Henning C, Ströbel P, Legler T, Riggert J, Hellenkamp K, Voigt JU, Hasenfuß G, Hinkel R, Wu JC, Behr R, Zimmermann WH. Engineered heart muscle allografts for heart repair in primates and humans. Nature 2025; 639:503-511. [PMID: 39880949 PMCID: PMC11903342 DOI: 10.1038/s41586-024-08463-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 11/27/2024] [Indexed: 01/31/2025]
Abstract
Cardiomyocytes can be implanted to remuscularize the failing heart1-7. Challenges include sufficient cardiomyocyte retention for a sustainable therapeutic impact without intolerable side effects, such as arrhythmia and tumour growth. We investigated the hypothesis that epicardial engineered heart muscle (EHM) allografts from induced pluripotent stem cell-derived cardiomyocytes and stromal cells structurally and functionally remuscularize the chronically failing heart without limiting side effects in rhesus macaques. After confirmation of in vitro and in vivo (nude rat model) equivalence of the newly developed rhesus macaque EHM model with a previously established Good Manufacturing Practice-compatible human EHM formulation8, long-term retention (up to 6 months) and dose-dependent enhancement of the target heart wall by EHM grafts constructed from 40 to 200 million cardiomyocytes/stromal cells were demonstrated in macaques with and without myocardial infarction-induced heart failure. In the heart failure model, evidence for EHM allograft-enhanced target heart wall contractility and ejection fraction, which are measures for local and global heart support, was obtained. Histopathological and gadolinium-based perfusion magnetic resonance imaging analyses confirmed cell retention and functional vascularization. Arrhythmia and tumour growth were not observed. The obtained feasibility, safety and efficacy data provided the pivotal underpinnings for the approval of a first-in-human clinical trial on tissue-engineered heart repair. Our clinical data confirmed remuscularization by EHM implantation in a patient with advanced heart failure.
Collapse
Affiliation(s)
- Ahmad-Fawad Jebran
- Department of Cardiothoracic and Vascular Surgery, University Medical Center Göttingen, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
| | - Tim Seidler
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- Department of Cardiology, Campus Kerckhoff of the Justus-Liebig-Universität Gießen, Kerckhoff-Clinic, Bad Nauheim, Germany
| | - Malte Tiburcy
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
| | - Maria Daskalaki
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Platform Degenerative Diseases, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Ingo Kutschka
- Department of Cardiothoracic and Vascular Surgery, University Medical Center Göttingen, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
| | - Buntaro Fujita
- Clinic for Cardiac and Thoracic Vascular Surgery, University Medical Center Schleswig Holstein, Campus Lübeck, Lübeck, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site North, Lübeck, Germany
| | - Stephan Ensminger
- Clinic for Cardiac and Thoracic Vascular Surgery, University Medical Center Schleswig Holstein, Campus Lübeck, Lübeck, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site North, Lübeck, Germany
| | - Felix Bremmer
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Amir Moussavi
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Functional Imaging Laboratory, German Primate Center, Göttingen, Germany
| | - Huaxiao Yang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Xulei Qin
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Sophie Mißbach
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Platform Degenerative Diseases, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
- Laboratory Animal Science Unit, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Charis Drummer
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Platform Degenerative Diseases, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Hassina Baraki
- Department of Cardiothoracic and Vascular Surgery, University Medical Center Göttingen, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
| | - Susann Boretius
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Functional Imaging Laboratory, German Primate Center, Göttingen, Germany
| | - Christopher Hasenauer
- Institute of Diagnostic and Interventional Radiology, University Medical Center Göttingen, Göttingen, Germany
| | - Tobias Nette
- Institute of Diagnostic and Interventional Radiology, University Medical Center Göttingen, Göttingen, Germany
| | - Johannes Kowallick
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Institute of Diagnostic and Interventional Radiology, University Medical Center Göttingen, Göttingen, Germany
| | - Christian O Ritter
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Institute of Diagnostic and Interventional Radiology, University Medical Center Göttingen, Göttingen, Germany
| | - Joachim Lotz
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Institute of Diagnostic and Interventional Radiology, University Medical Center Göttingen, Göttingen, Germany
| | - Michael Didié
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Mathias Mietsch
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Laboratory Animal Science Unit, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Tim Meyer
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
| | - George Kensah
- Department of Cardiothoracic and Vascular Surgery, University Medical Center Göttingen, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
| | - Dennis Krüger
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Md Sadman Sakib
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Lalit Kaurani
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Andre Fischer
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Ralf Dressel
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Ignacio Rodriguez-Polo
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Platform Degenerative Diseases, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Michael Stauske
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Platform Degenerative Diseases, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Sebastian Diecke
- Pluripotent Stem Cells Platform, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Kerstin Maetz-Rensing
- Pathology Unit, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Eva Gruber-Dujardin
- Pathology Unit, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Martina Bleyer
- Pathology Unit, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Beatrix Petersen
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Primate Genetics Laboratory, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Christian Roos
- Primate Genetics Laboratory, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Liye Zhang
- Primate Genetics Laboratory, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Lutz Walter
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Primate Genetics Laboratory, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Silke Kaulfuß
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Gökhan Yigit
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Bernd Wollnik
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Elif Levent
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
| | - Berit Roshani
- Unit of Infection Models, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Christiane Stahl-Henning
- Unit of Infection Models, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Tobias Legler
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Department of Transfusion Medicine, University Medical Center Göttingen, Göttingen, Germany
| | - Joachim Riggert
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Department of Transfusion Medicine, University Medical Center Göttingen, Göttingen, Germany
| | - Kristian Hellenkamp
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Jens-Uwe Voigt
- Department of Cardiovascular Sciences, Catholic University of Leuven and Department of Cardiovascular Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Gerd Hasenfuß
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Rabea Hinkel
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Laboratory Animal Science Unit, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Rüdiger Behr
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany
- Platform Degenerative Diseases, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Wolfram-Hubertus Zimmermann
- German Centre for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Göttingen, Germany.
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany.
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Göttingen, Germany.
| |
Collapse
|
12
|
Sun X, Wu J, Mourad O, Li R, Nunes SS. Microvessel co-transplantation improves poor remuscularization by hiPSC-cardiomyocytes in a complex disease model of myocardial infarction and type 2 diabetes. Stem Cell Reports 2025; 20:102394. [PMID: 39855203 PMCID: PMC11864147 DOI: 10.1016/j.stemcr.2024.102394] [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: 06/17/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/27/2025] Open
Abstract
People with type 2 diabetes (T2D) are at a higher risk for myocardial infarction (MI) than age-matched healthy individuals. Here, we studied cell-based cardiac regeneration post MI in T2D rats modeling the co-morbid conditions in patients with MI. We recapitulated the T2D hallmarks and clinical aspects of diabetic cardiomyopathy using high-fat diet and streptozotocin in athymic rats, which were then subjected to MI and intramyocardial implantation of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) with or without rat adipose-derived microvessels (MVs). hiPSC-CM alone engrafted poorly. Co-delivery of hiPSC-CMs with MVs yielded a smaller infarct area and a thicker left ventricle wall. Additionally, MVs robustly integrated into the infarcted hearts, improved the survival of hiPSC-CMs, and improved cardiac function. MV-conditioned media also promoted hiPSC-CM maturation in vitro, increasing cardiomyocyte (CM) size in an interleukin (IL)-6-dependent manner. Given the availability of MVs from human adipose tissue, MVs present great translational potential for the treatment of heart failure in people with T2D.
Collapse
MESH Headings
- Induced Pluripotent Stem Cells/cytology
- Induced Pluripotent Stem Cells/metabolism
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/transplantation
- Myocytes, Cardiac/metabolism
- Animals
- Diabetes Mellitus, Type 2/therapy
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/metabolism
- Humans
- Myocardial Infarction/therapy
- Myocardial Infarction/pathology
- Myocardial Infarction/complications
- Microvessels/transplantation
- Microvessels/metabolism
- Rats
- Disease Models, Animal
- Male
- Rats, Nude
- Cell Differentiation
- Diabetic Cardiomyopathies/therapy
- Diabetes Mellitus, Experimental/therapy
Collapse
Affiliation(s)
- Xuetao Sun
- Toronto General Hospital Research Institute, University Health Network, 101 College St., Toronto, ON M5G 1L7, Canada
| | - Jun Wu
- Toronto General Hospital Research Institute, University Health Network, 101 College St., Toronto, ON M5G 1L7, Canada
| | - Omar Mourad
- Toronto General Hospital Research Institute, University Health Network, 101 College St., Toronto, ON M5G 1L7, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Renke Li
- Toronto General Hospital Research Institute, University Health Network, 101 College St., Toronto, ON M5G 1L7, Canada; Laboratory of Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Sara S Nunes
- Toronto General Hospital Research Institute, University Health Network, 101 College St., Toronto, ON M5G 1L7, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Laboratory of Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Heart & Stroke/Richard Lewar Centre of Excellence, University of Toronto, Toronto, ON, Canada; Ajmera Transplant Center, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
13
|
Dwyer KD, Snyder CA, Coulombe KLK. Cardiomyocytes in Hypoxia: Cellular Responses and Implications for Cell-Based Cardiac Regenerative Therapies. Bioengineering (Basel) 2025; 12:154. [PMID: 40001674 PMCID: PMC11851968 DOI: 10.3390/bioengineering12020154] [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: 01/04/2025] [Revised: 01/28/2025] [Accepted: 02/02/2025] [Indexed: 02/27/2025] Open
Abstract
Myocardial infarction (MI) is a severe hypoxic event, resulting in the loss of up to one billion cardiomyocytes (CMs). Due to the limited intrinsic regenerative capacity of the heart, cell-based regenerative therapies, which feature the implantation of stem cell-derived cardiomyocytes (SC-CMs) into the infarcted myocardium, are being developed with the goal of restoring lost muscle mass, re-engineering cardiac contractility, and preventing the progression of MI into heart failure (HF). However, such cell-based therapies are challenged by their susceptibility to oxidative stress in the ischemic environment of the infarcted heart. To maximize the therapeutic benefits of cell-based approaches, a better understanding of the heart environment at the cellular, tissue, and organ level throughout MI is imperative. This review provides a comprehensive summary of the cardiac pathophysiology occurring during and after MI, as well as how these changes define the cardiac environment to which cell-based cardiac regenerative therapies are delivered. This understanding is then leveraged to frame how cell culture treatments may be employed to enhance SC-CMs' hypoxia resistance. In this way, we synthesize both the complex experience of SC-CMs upon implantation and the engineering techniques that can be utilized to develop robust SC-CMs for the clinical translation of cell-based cardiac therapies.
Collapse
Affiliation(s)
| | | | - Kareen L. K. Coulombe
- Institute for Biology, Engineering, and Medicine, School of Engineering, Brown University, Providence, RI 02912, USA; (K.D.D.); (C.A.S.)
| |
Collapse
|
14
|
Tariq H, Bukhari SZ, An R, Dong J, Ihsan A, Younis MR. Stem cell-derived exosome delivery systems for treating atherosclerosis: The new frontier of stem cell therapy. Mater Today Bio 2025; 30:101440. [PMID: 39866781 PMCID: PMC11758955 DOI: 10.1016/j.mtbio.2024.101440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/14/2024] [Accepted: 12/30/2024] [Indexed: 01/28/2025] Open
Abstract
Cardiovascular diseases (CVDs) are a leading cause of mortality worldwide. As a chronic inflammatory disease with a complicated pathophysiology marked by abnormal lipid metabolism and arterial plaque formation, atherosclerosis is a major contributor to CVDs and can induce abrupt cardiac events. The discovery of exosomes' role in intercellular communication has sparked a great deal of interest in them recently. Exosomes are involved in strategic phases of the onset and development of atherosclerosis because they have been identified to control pathophysiologic pathways including inflammation, angiogenesis, or senescence. This review investigates the potential role of stem cell-derived exosomes in atherosclerosis management. We briefly introduced atherosclerosis and stem cell therapy including stem cell-derived exosomes. The biogenesis of exosomes along with their secretion and isolation have been elaborated. The design engineering of exosomes has been summarized to present how drug loading and surface modification with targeting ligands can improve the therapeutic and targeting capacity of exosomes, demonstrating atheroprotective action. Moreover, the mechanism of action (endothelial dysfunction, reduction of dyslipidemia, macrophage polarization, vascular calcification, and angiogenesis) of drug-loaded exosomes to treat atherosclerosis has been discussed in detail. In the end, a comparative and balanced viewpoint has been given regarding the current challenges and potential solutions to advance exosome engineering for cardiovascular therapeutic applications.
Collapse
Affiliation(s)
- Hassan Tariq
- Department of Molecular, Cell and Developmental Biology, University of California - Los Angeles, Los Angeles, CA, 90095, USA
| | - Syeda Zunaira Bukhari
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Ruibing An
- Institute of Optical Functional Materials for Biomedical Imaging, School of Chemistry and Pharmaceutical Engineering, Shandong First Medical University & Shandong Academy of Medical Science, Taian, Shandong, 271016, PR China
| | - Jian Dong
- Institute of Optical Functional Materials for Biomedical Imaging, School of Chemistry and Pharmaceutical Engineering, Shandong First Medical University & Shandong Academy of Medical Science, Taian, Shandong, 271016, PR China
| | - Ayesha Ihsan
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Muhammad Rizwan Younis
- Institute of Optical Functional Materials for Biomedical Imaging, School of Chemistry and Pharmaceutical Engineering, Shandong First Medical University & Shandong Academy of Medical Science, Taian, Shandong, 271016, PR China
- Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, CA, 90095, USA
- Department of Molecular, Cell and Developmental Biology, University of California - Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
15
|
Wang W, Lu L, Ma H, Li Z, Lu X, Xie Y. Self-template manufacturing of on-skin electrodes with 3D multi-channel structure for standard 3-limb-lead ECG suit. MICROSYSTEMS & NANOENGINEERING 2024; 10:196. [PMID: 39681565 DOI: 10.1038/s41378-024-00838-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/12/2024] [Accepted: 11/10/2024] [Indexed: 12/18/2024]
Abstract
Wearable electrocardiogram (ECG) devices are the mainstream technology in the diagnosis of various cardiovascular diseases, in which soft, flexible, permeable electrodes are the key link in human-machine interface to capture bioelectrical signals. Herein, we propose a self-template strategy to fabricate silver-coated fiber/silicone (AgCF-S) electrodes. With a simple dissolving-curing-redissolving process, the polyvinyl acetate shell around the AgCF core is in-situ removed to form a three-dimensional (3D) multi-channel structure. The conductive fibers overlap each other and pass through the silicon substrate in a network state, so that the electrode can be bent to 180° or stretched to 30%. The 3D multi-channels in AgCF-S adhesive is further coupled with a Kirigami-design structure of flexible substrate, to maintain high flexibility without sacrificing air-permeability, enabling an excellent water evaporation rate of 1.8 μg/mm2/min, and non-allergenic adhere on pigskin after 24 h. Combined with the self-developed standard 3-limb-lead ECG suit, multi-lead signals with high signal-to-noise ratio (SNR) and low variance (σ2), can be transmitted in real-time via Bluetooth and displayed in the client. Typical heart diseases such as coronary, arrhythmia, myocardial infarction, etc., are detected by our ECG equipment, revealing a huge promise in future medical electronics.
Collapse
Affiliation(s)
- Wentao Wang
- School of Automotive and Mechanical Engineering, Changsha University of Science and Technology, Changsha, 410114, China
| | - Longsheng Lu
- Guangdong Key Laboratory of Precision Equipment and Manufacturing Technology, South China University of Technology, Guangzhou, 510641, China
| | - Huan Ma
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangzhou, 510080, China
| | - Zehong Li
- Guangdong Key Laboratory of Precision Equipment and Manufacturing Technology, South China University of Technology, Guangzhou, 510641, China
| | - Xiaoyu Lu
- Guangdong Key Laboratory of Precision Equipment and Manufacturing Technology, South China University of Technology, Guangzhou, 510641, China
| | - Yingxi Xie
- Guangdong Key Laboratory of Precision Equipment and Manufacturing Technology, South China University of Technology, Guangzhou, 510641, China.
| |
Collapse
|
16
|
Cojocaru C, Dorobanțu M, Vătășescu R. Pre-ablation and Post-ablation Factors Influencing the Prognosis of Patients with Electrical Storm Treated by Radiofrequency Catheter Ablation: An Update. Rev Cardiovasc Med 2024; 25:432. [PMID: 39742218 PMCID: PMC11683710 DOI: 10.31083/j.rcm2512432] [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: 06/26/2024] [Revised: 09/18/2024] [Accepted: 09/25/2024] [Indexed: 01/03/2025] Open
Abstract
Catheter ablation-based management strategies for the drug-refractory electrical storm (ES) have been proven to abolish acute ventricular arrhythmic episodes and improve long-term outcomes. However, this effect is highly influenced by multiple independently acting factors, which, if identified and addressed, may allow a more tailored management to each particular case to improve results. This review synthesizes existing evidence concerning ES outcome predictors of patients undergoing ablation and introduces the role of novel scoring algorithms to refine risk stratification. The presence of these factors should be assessed during two distinct phases in relation to the ablation procedure: before (based on preprocedural multimodal evaluation of the patient's structural heart disease and comorbidities) and after the ablation procedure (in terms of information derived from the invasive substrate characterization, procedural results, postprocedural recurrences (spontaneous or during non-invasive testing), and complications).
Collapse
Affiliation(s)
- Cosmin Cojocaru
- Department of Cardiothoracic Pathology, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Cardiology, Emergency Clinical Hospital of Bucharest, 014461 Bucharest, Romania
| | - Maria Dorobanțu
- Department of Cardiothoracic Pathology, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Romanian Academy, 010071 Bucharest, Romania
| | - Radu Vătășescu
- Department of Cardiothoracic Pathology, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Cardiology, Emergency Clinical Hospital of Bucharest, 014461 Bucharest, Romania
| |
Collapse
|
17
|
Soma Y, Tohyama S, Kubo A, Yamasaki T, Kabasawa N, Haga K, Tani H, Morita-Umei Y, Umei TC, Sekine O, Nakamura M, Moriwaki T, Tanosaki S, Someya S, Kawai Y, Ohno M, Kishino Y, Kanazawa H, Fujita J, Zhang MR, Suematsu M, Fukuda K, Ieda M. Metabolic changes of human induced pluripotent stem cell-derived cardiomyocytes and teratomas after transplantation. iScience 2024; 27:111234. [PMID: 39569381 PMCID: PMC11576393 DOI: 10.1016/j.isci.2024.111234] [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: 05/30/2024] [Revised: 08/23/2024] [Accepted: 10/21/2024] [Indexed: 11/22/2024] Open
Abstract
Cardiac regenerative therapy using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) has been applied in clinical settings. Herein, we aimed to investigate the in vivo metabolic profiles of hiPSC-CM grafts. RNA sequencing and imaging mass spectrometry were performed in the present study, which revealed that hiPSC-CM grafts matured metabolically over time after transplantation. Glycolysis, which was active in the hiPSC-CM grafts immediately after transplantation, shifted to fatty acid oxidation. Additionally, we examined the metabolic profile of teratomas that may form when non-CMs, including undifferentiated human induced pluripotent stem cells (hiPSCs), remain in transplanted cells. The upregulated gene expression of amino acid transporters and the high accumulation of amino acids, such as methionine and aromatic amino acids, were observed in the teratomas. We show that subcutaneous teratomas derived from undifferentiated hiPSCs can be detected in vivo using positron emission tomography with [18F]fluorophenylalanine ([18F]fPhe). These results provided insights into the clinical application of cardiac regenerative therapy.
Collapse
Affiliation(s)
- Yusuke Soma
- Department of Clinical Regenerative Medicine, Fujita Medical Innovation Center, Fujita Health University, Ota-ku, Tokyo 144-0041, Japan
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shugo Tohyama
- Department of Clinical Regenerative Medicine, Fujita Medical Innovation Center, Fujita Health University, Ota-ku, Tokyo 144-0041, Japan
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Akiko Kubo
- Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Tomoteru Yamasaki
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Inage-ku, Chiba 263-8555, Japan
| | - Noriko Kabasawa
- Department of Clinical Regenerative Medicine, Fujita Medical Innovation Center, Fujita Health University, Ota-ku, Tokyo 144-0041, Japan
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Heartseed Inc, Minato-ku, Tokyo 105-0023, Japan
| | - Kotaro Haga
- Department of Clinical Regenerative Medicine, Fujita Medical Innovation Center, Fujita Health University, Ota-ku, Tokyo 144-0041, Japan
| | - Hidenori Tani
- Department of Clinical Regenerative Medicine, Fujita Medical Innovation Center, Fujita Health University, Ota-ku, Tokyo 144-0041, Japan
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Center for Prevention Medicine, Keio University School of Medicine, Minato-ku, Tokyo 106-0041, Japan
| | - Yuika Morita-Umei
- Department of Clinical Regenerative Medicine, Fujita Medical Innovation Center, Fujita Health University, Ota-ku, Tokyo 144-0041, Japan
- Kanagawa Institute of Industrial Science and Technology (KISTEC), Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Tomohiko C Umei
- Department of Clinical Regenerative Medicine, Fujita Medical Innovation Center, Fujita Health University, Ota-ku, Tokyo 144-0041, Japan
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Otoya Sekine
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Masashi Nakamura
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Taijun Moriwaki
- Department of Clinical Regenerative Medicine, Fujita Medical Innovation Center, Fujita Health University, Ota-ku, Tokyo 144-0041, Japan
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Sho Tanosaki
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shota Someya
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yujiro Kawai
- Department of Cardiovascular Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Masatoshi Ohno
- Department of Clinical Regenerative Medicine, Fujita Medical Innovation Center, Fujita Health University, Ota-ku, Tokyo 144-0041, Japan
- Department of Cardiovascular Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yoshikazu Kishino
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hideaki Kanazawa
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Jun Fujita
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Inage-ku, Chiba 263-8555, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- WPI-Bio2Q, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Central Institute for Experimental Medicine and Life Science, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Heartseed Inc, Minato-ku, Tokyo 105-0023, Japan
| | - Masaki Ieda
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
18
|
Abusalah MAH, Priyanka, Abd Rahman ENSE, Choudhary OP. Evolving trends in stem cell therapy: an emerging and promising approach against various diseases. Int J Surg 2024; 110:6862-6868. [PMID: 39699861 DOI: 10.1097/js9.0000000000001948] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 07/01/2024] [Indexed: 12/20/2024]
Affiliation(s)
- Mai Abdel Haleem Abusalah
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, Jordan
| | - Priyanka
- Department of Veterinary Microbiology, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University
| | - Engku Nur Syafirah Engku Abd Rahman
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian, Kota Bharu, Kelantan, Malaysia
| | - Om Prakash Choudhary
- Department of Veterinary Anatomy, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University, Rampura Phul, Bathinda, Punjab, India
| |
Collapse
|
19
|
Snyder CA, Dwyer KD, Coulombe KLK. Advancing Human iPSC-Derived Cardiomyocyte Hypoxia Resistance for Cardiac Regenerative Therapies through a Systematic Assessment of In Vitro Conditioning. Int J Mol Sci 2024; 25:9627. [PMID: 39273573 PMCID: PMC11395605 DOI: 10.3390/ijms25179627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 08/23/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Acute myocardial infarction (MI) is a sudden, severe cardiac ischemic event that results in the death of up to one billion cardiomyocytes (CMs) and subsequent decrease in cardiac function. Engineered cardiac tissues (ECTs) are a promising approach to deliver the necessary mass of CMs to remuscularize the heart. However, the hypoxic environment of the heart post-MI presents a critical challenge for CM engraftment. Here, we present a high-throughput, systematic study targeting several physiological features of human induced pluripotent stem cell-derived CMs (hiPSC-CMs), including metabolism, Wnt signaling, substrate, heat shock, apoptosis, and mitochondrial stabilization, to assess their efficacy in promoting ischemia resistance in hiPSC-CMs. The results of 2D experiments identify hypoxia preconditioning (HPC) and metabolic conditioning as having a significant influence on hiPSC-CM function in normoxia and hypoxia. Within 3D engineered cardiac tissues (ECTs), metabolic conditioning with maturation media (MM), featuring high fatty acid and calcium concentration, results in a 1.5-fold increase in active stress generation as compared to RPMI/B27 control ECTs in normoxic conditions. Yet, this functional improvement is lost after hypoxia treatment. Interestingly, HPC can partially rescue the function of MM-treated ECTs after hypoxia. Our systematic and iterative approach provides a strong foundation for assessing and leveraging in vitro culture conditions to enhance the hypoxia resistance, and thus the successful clinical translation, of hiPSC-CMs in cardiac regenerative therapies.
Collapse
Affiliation(s)
- Caroline A Snyder
- Institute for Biology, Engineering and Medicine, School of Engineering, Brown University, Providence, RI 02912, USA
| | - Kiera D Dwyer
- Institute for Biology, Engineering and Medicine, School of Engineering, Brown University, Providence, RI 02912, USA
| | - Kareen L K Coulombe
- Institute for Biology, Engineering and Medicine, School of Engineering, Brown University, Providence, RI 02912, USA
| |
Collapse
|
20
|
Kistamás K, Lamberto F, Vaiciuleviciute R, Leal F, Muenthaisong S, Marte L, Subías-Beltrán P, Alaburda A, Arvanitis DN, Zana M, Costa PF, Bernotiene E, Bergaud C, Dinnyés A. The Current State of Realistic Heart Models for Disease Modelling and Cardiotoxicity. Int J Mol Sci 2024; 25:9186. [PMID: 39273136 PMCID: PMC11394806 DOI: 10.3390/ijms25179186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/18/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
One of the many unresolved obstacles in the field of cardiovascular research is an uncompromising in vitro cardiac model. While primary cell sources from animal models offer both advantages and disadvantages, efforts over the past half-century have aimed to reduce their use. Additionally, obtaining a sufficient quantity of human primary cardiomyocytes faces ethical and legal challenges. As the practically unlimited source of human cardiomyocytes from induced pluripotent stem cells (hiPSC-CM) is now mostly resolved, there are great efforts to improve their quality and applicability by overcoming their intrinsic limitations. The greatest bottleneck in the field is the in vitro ageing of hiPSC-CMs to reach a maturity status that closely resembles that of the adult heart, thereby allowing for more appropriate drug developmental procedures as there is a clear correlation between ageing and developing cardiovascular diseases. Here, we review the current state-of-the-art techniques in the most realistic heart models used in disease modelling and toxicity evaluations from hiPSC-CM maturation through heart-on-a-chip platforms and in silico models to the in vitro models of certain cardiovascular diseases.
Collapse
Affiliation(s)
- Kornél Kistamás
- BioTalentum Ltd., Aulich Lajos Str 26, H-2100 Gödöllő, Hungary
| | - Federica Lamberto
- BioTalentum Ltd., Aulich Lajos Str 26, H-2100 Gödöllő, Hungary
- Department of Physiology and Animal Health, Institute of Physiology and Animal Nutrition, Hungarian University of Agriculture and Life Sciences, Páter Károly Str 1, H-2100 Gödöllő, Hungary
| | - Raminta Vaiciuleviciute
- Department of Regenerative Medicine, State Research Institute Innovative Medicine Centre, Santariskiu g. 5, LT-08406 Vilnius, Lithuania
| | - Filipa Leal
- Biofabics Lda, Rua Alfredo Allen 455, 4200-135 Porto, Portugal
| | | | - Luis Marte
- Digital Health Unit, Eurecat-Centre Tecnològic de Catalunya, 08005 Barcelona, Spain
| | - Paula Subías-Beltrán
- Digital Health Unit, Eurecat-Centre Tecnològic de Catalunya, 08005 Barcelona, Spain
| | - Aidas Alaburda
- Department of Regenerative Medicine, State Research Institute Innovative Medicine Centre, Santariskiu g. 5, LT-08406 Vilnius, Lithuania
- Institute of Biosciences, Life Sciences Center, Vilnius University, Sauletekio al. 7, LT-10257 Vilnius, Lithuania
| | - Dina N Arvanitis
- Laboratory for Analysis and Architecture of Systems-French National Centre for Scientific Research (LAAS-CNRS), 7 Avenue du Colonel Roche, F-31400 Toulouse, France
| | - Melinda Zana
- BioTalentum Ltd., Aulich Lajos Str 26, H-2100 Gödöllő, Hungary
| | - Pedro F Costa
- Biofabics Lda, Rua Alfredo Allen 455, 4200-135 Porto, Portugal
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Innovative Medicine Centre, Santariskiu g. 5, LT-08406 Vilnius, Lithuania
- Faculty of Fundamental Sciences, Vilnius Tech, Sauletekio al. 11, LT-10223 Vilnius, Lithuania
| | - Christian Bergaud
- Laboratory for Analysis and Architecture of Systems-French National Centre for Scientific Research (LAAS-CNRS), 7 Avenue du Colonel Roche, F-31400 Toulouse, France
| | - András Dinnyés
- BioTalentum Ltd., Aulich Lajos Str 26, H-2100 Gödöllő, Hungary
- Department of Physiology and Animal Health, Institute of Physiology and Animal Nutrition, Hungarian University of Agriculture and Life Sciences, Páter Károly Str 1, H-2100 Gödöllő, Hungary
| |
Collapse
|
21
|
Kobayashi H, Tohyama S, Ichimura H, Ohashi N, Chino S, Soma Y, Tani H, Tanaka Y, Yang X, Shiba N, Kadota S, Haga K, Moriwaki T, Morita-Umei Y, Umei TC, Sekine O, Kishino Y, Kanazawa H, Kawagishi H, Yamada M, Narita K, Naito T, Seto T, Kuwahara K, Shiba Y, Fukuda K. Regeneration of Nonhuman Primate Hearts With Human Induced Pluripotent Stem Cell-Derived Cardiac Spheroids. Circulation 2024; 150:611-621. [PMID: 38666382 DOI: 10.1161/circulationaha.123.064876] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 03/21/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND The clinical application of human induced pluripotent stem cell-derived cardiomyocytes (CMs) for cardiac repair commenced with the epicardial delivery of engineered cardiac tissue; however, the feasibility of the direct delivery of human induced pluripotent stem cell-derived CMs into the cardiac muscle layer, which has reportedly induced electrical integration, is unclear because of concerns about poor engraftment of CMs and posttransplant arrhythmias. Thus, in this study, we prepared purified human induced pluripotent stem cell-derived cardiac spheroids (hiPSC-CSs) and investigated whether their direct injection could regenerate infarcted nonhuman primate hearts. METHODS We performed 2 separate experiments to explore the appropriate number of human induced pluripotent stem cell-derived CMs. In the first experiment, 10 cynomolgus monkeys were subjected to myocardial infarction 2 weeks before transplantation and were designated as recipients of hiPSC-CSs containing 2×107 CMs or the vehicle. The animals were euthanized 12 weeks after transplantation for histological analysis, and cardiac function and arrhythmia were monitored during the observational period. In the second study, we repeated the equivalent transplantation study using more CMs (6×107 CMs). RESULTS Recipients of hiPSC-CSs containing 2×107 CMs showed limited CM grafts and transient increases in fractional shortening compared with those of the vehicle (fractional shortening at 4 weeks after transplantation [mean ± SD]: 26.2±2.1%; 19.3±1.8%; P<0.05), with a low incidence of posttransplant arrhythmia. Transplantation of increased dose of CMs resulted in significantly greater engraftment and long-term contractile benefits (fractional shortening at 12 weeks after transplantation: 22.5±1.0%; 16.6±1.1%; P<0.01, left ventricular ejection fraction at 12 weeks after transplantation: 49.0±1.4%; 36.3±2.9%; P<0.01). The incidence of posttransplant arrhythmia slightly increased in recipients of hiPSC-CSs containing 6×107 CMs. CONCLUSIONS We demonstrated that direct injection of hiPSC-CSs restores the contractile functions of injured primate hearts with an acceptable risk of posttransplant arrhythmia. Although the mechanism for the functional benefits is not fully elucidated, these findings provide a strong rationale for conducting clinical trials using the equivalent CM products.
Collapse
Affiliation(s)
- Hideki Kobayashi
- Department of Cardiovascular Medicine (H. Kobayashi, K.K.), Shinshu University, Matsumoto, Japan
| | - Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
| | - Hajime Ichimura
- Division of Cardiovascular Surgery, Department of Surgery (H.I., N.O., S.C., Y.T., T.S.), Shinshu University, Matsumoto, Japan
- School of Medicine, Department of Regenerative Science and Medicine (H.I., Y.T., X.Y., N.S., S.K., Y. Shiba), Shinshu University, Matsumoto, Japan
| | - Noburo Ohashi
- Division of Cardiovascular Surgery, Department of Surgery (H.I., N.O., S.C., Y.T., T.S.), Shinshu University, Matsumoto, Japan
| | - Shuji Chino
- Division of Cardiovascular Surgery, Department of Surgery (H.I., N.O., S.C., Y.T., T.S.), Shinshu University, Matsumoto, Japan
| | - Yusuke Soma
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
| | - Hidenori Tani
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
| | - Yuki Tanaka
- Division of Cardiovascular Surgery, Department of Surgery (H.I., N.O., S.C., Y.T., T.S.), Shinshu University, Matsumoto, Japan
- School of Medicine, Department of Regenerative Science and Medicine (H.I., Y.T., X.Y., N.S., S.K., Y. Shiba), Shinshu University, Matsumoto, Japan
| | - Xiao Yang
- School of Medicine, Department of Regenerative Science and Medicine (H.I., Y.T., X.Y., N.S., S.K., Y. Shiba), Shinshu University, Matsumoto, Japan
| | - Naoko Shiba
- School of Medicine, Department of Regenerative Science and Medicine (H.I., Y.T., X.Y., N.S., S.K., Y. Shiba), Shinshu University, Matsumoto, Japan
| | - Shin Kadota
- School of Medicine, Department of Regenerative Science and Medicine (H.I., Y.T., X.Y., N.S., S.K., Y. Shiba), Shinshu University, Matsumoto, Japan
- Institute for Biomedical Sciences (S.K., H. Kawagishi, K.K., Y. Shiba), Shinshu University, Matsumoto, Japan
| | - Kotaro Haga
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
| | - Taijun Moriwaki
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
| | - Yuika Morita-Umei
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
- Kanagawa Institute of Industrial Science and Technology, Japan (Y.M.-U.)
| | - Tomohiko C Umei
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
| | - Otoya Sekine
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
| | - Yoshikazu Kishino
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
| | - Hideaki Kanazawa
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
| | - Hiroyuki Kawagishi
- Department of Molecular Pharmacology (H. Kawagishi, M.Y.), Shinshu University, Matsumoto, Japan
- Institute for Biomedical Sciences (S.K., H. Kawagishi, K.K., Y. Shiba), Shinshu University, Matsumoto, Japan
| | - Mitsuhiko Yamada
- Department of Molecular Pharmacology (H. Kawagishi, M.Y.), Shinshu University, Matsumoto, Japan
| | - Kazumasa Narita
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Medicine (K.N., T.N.), Shinshu University, Matsumoto, Japan
- Department of Pharmacy, Shinshu University Hospital, Matsumoto, Japan (K.N., T.N.)
| | - Takafumi Naito
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Medicine (K.N., T.N.), Shinshu University, Matsumoto, Japan
- Department of Pharmacy, Shinshu University Hospital, Matsumoto, Japan (K.N., T.N.)
| | - Tatsuichiro Seto
- Division of Cardiovascular Surgery, Department of Surgery (H.I., N.O., S.C., Y.T., T.S.), Shinshu University, Matsumoto, Japan
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
| | - Koichiro Kuwahara
- Department of Cardiovascular Medicine (H. Kobayashi, K.K.), Shinshu University, Matsumoto, Japan
- Institute for Biomedical Sciences (S.K., H. Kawagishi, K.K., Y. Shiba), Shinshu University, Matsumoto, Japan
| | - Yuji Shiba
- School of Medicine, Department of Regenerative Science and Medicine (H.I., Y.T., X.Y., N.S., S.K., Y. Shiba), Shinshu University, Matsumoto, Japan
- Institute for Biomedical Sciences (S.K., H. Kawagishi, K.K., Y. Shiba), Shinshu University, Matsumoto, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
| |
Collapse
|
22
|
Yahyazadeh R, Baradaran Rahimi V, Askari VR. Stem cell and exosome therapies for regenerating damaged myocardium in heart failure. Life Sci 2024; 351:122858. [PMID: 38909681 DOI: 10.1016/j.lfs.2024.122858] [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: 05/09/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024]
Abstract
Finding novel treatments for cardiovascular diseases (CVDs) is a hot topic in medicine; cell-based therapies have reported promising news for controlling dangerous complications of heart disease such as myocardial infarction (MI) and heart failure (HF). Various progenitor/stem cells were tested in various in-vivo, in-vitro, and clinical studies for regeneration or repairing the injured tissue in the myocardial to accelerate the healing. Fetal, adult, embryonic, and induced pluripotent stem cells (iPSC) have revealed the proper potency for cardiac tissue repair. As an essential communicator among cells, exosomes with specific contacts (proteins, lncRNAs, and miRNAs) greatly promote cardiac rehabilitation. Interestingly, stem cell-derived exosomes have more efficiency than stem cell transplantation. Therefore, stem cells induced pluripotent stem cells (iPSCs), embryonic stem cells (ESCs), cardiac stem cells (CDC), and skeletal myoblasts) and their-derived exosomes will probably be considered an alternative therapy for CVDs remedy. In addition, stem cell-derived exosomes have been used in the diagnosis/prognosis of heart diseases. In this review, we explained the advances of stem cells/exosome-based treatment, their beneficial effects, and underlying mechanisms, which will present new insights in the clinical field in the future.
Collapse
Affiliation(s)
- Roghayeh Yahyazadeh
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
23
|
Stüdemann T, Schwarzová B, Schneidewind T, Geertz B, von Bibra C, Nehring M, Rössinger J, Wiegert JS, Eschenhagen T, Weinberger F. Impulse initiation in engrafted pluripotent stem cell-derived cardiomyocytes can stimulate the recipient heart. Stem Cell Reports 2024; 19:1053-1060. [PMID: 39059379 PMCID: PMC11368679 DOI: 10.1016/j.stemcr.2024.06.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 06/26/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Transplantation of pluripotent stem cell-derived cardiomyocytes is a novel promising cell-based therapeutic approach for patients with heart failure. However, engraftment arrhythmias are a predictable life-threatening complication and represent a major hurdle for clinical translation. Thus, we wanted to experimentally study whether impulse generation by transplanted cardiomyocytes can propagate to the host myocardium and overdrive the recipient rhythm. We transplanted human induced pluripotent stem cell-derived cardiomyocytes expressing the optogenetic actuator Bidirectional Pair of Opsins for Light-induced Excitation and Silencing (BiPOLES) in a guinea pig injury model. Eight weeks after transplantation ex vivo, Langendorff perfusion was used to assess electrical coupling. Pulsed photostimulation was applied to specifically activate the engrafted cardiomyocytes. Photostimulation resulted in ectopic pacemaking that propagated to the host myocardium, caused non-sustained arrhythmia, and stimulated the recipient heart with higher pacing frequency (4/9 hearts). Our study demonstrates that transplanted cardiomyocytes can (1) electrically couple to the host myocardium and (2) stimulate the recipient heart. Thus, our results provide experimental evidence for an important aspect of engraftment-induced arrhythmia induction and thereby support the current hypothesis that cardiomyocyte automaticity can serve as a trigger for ventricular arrhythmias.
Collapse
Affiliation(s)
- Tim Stüdemann
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| | - Barbora Schwarzová
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| | - Till Schneidewind
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| | - Birgit Geertz
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Constantin von Bibra
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| | - Marie Nehring
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| | - Judith Rössinger
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| | - J Simon Wiegert
- Research Group Synaptic Wiring and Information Processing, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neurophysiology, MCTN, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| | - Florian Weinberger
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany; Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain.
| |
Collapse
|
24
|
Matsumoto R, Enzhi Y, Takeda K, Morimoto K, Yogo K, Harada M, Tokushige K, Maehara Y, Hirota S, Kojima Y, Ito M, Sougawa N, Miyagawa S, Sawa Y, Okumura K, Uchida K. CD8 + T cell-mediated rejection of allogenic human-induced pluripotent stem cell-derived cardiomyocyte sheets in human PBMC-transferred NOG MHC double knockout mice. J Heart Lung Transplant 2024; 43:1348-1357. [PMID: 38657776 DOI: 10.1016/j.healun.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 03/28/2024] [Accepted: 04/15/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Transplantation of human-induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) has emerged as a promising therapy to treat end-stage heart failure. However, the immunogenicity of hiPS-CMs in transplanted patients has not been fully elucidated. Thus, in vivo models are required to estimate immune responses against hiPS-CMs in transplant recipients. METHODS We transferred human peripheral blood mononuclear cells (hPBMCs) into NOD/Shi-scid IL-2rgnull (NOG) MHC class I/II double knockout (NOG-ΔMHC) mice, which were reported to accept hPBMCs without xenogeneic-graft-versus-host disease (xeno-GVHD). Then, hiPS-CM sheets generated from the hiPS cell line 201B7 harboring a luciferase transgene were transplanted into the subcutaneous space of NOG-ΔMHC mice. Graft survival was monitored by bioluminescent images using a Xenogen In Vivo Imaging System. RESULTS The human immune cells were engrafted for more than 3 months in NOG-ΔMHC mice without lethal xeno-GVHD. The hiPS-CMs expressed a moderate level of human leukocyte antigen (HLA)-class I, but not HLA-class II, molecules even after interferon-gamma (IFN-γ) stimulation. Consistently, the allogenic IFN-γ-treated hiPS-CMs induced weak CD8+ but not CD4+ T cell responses in vitro. hiPS-CM sheets disappeared approximately 17 to 24 days after transplantation in hPBMC-transferred NOG-ΔMHC mice, and CD8+ T cell depletion significantly prolonged graft survival, similar to what was observed following tacrolimus treatment. CONCLUSIONS hiPS-CMs are less immunogenic in vitro but induce sufficient CD8+ T cell-mediated immune responses for graft rejection in vivo.
Collapse
Affiliation(s)
- Ryu Matsumoto
- Center for Immune Therapeutics and Diagnosis, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Yin Enzhi
- Center for Immune Therapeutics and Diagnosis, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kazuyoshi Takeda
- Center for Immune Therapeutics and Diagnosis, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Biofunctional Microbiota, Graduate School of Medicine, Juntendo University, Tokyo, Japan; Laboratory of Cell Biology, Research Support Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Kodai Morimoto
- Center for Immune Therapeutics and Diagnosis, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kyoko Yogo
- Center for Immune Therapeutics and Diagnosis, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masaki Harada
- Center for Immune Therapeutics and Diagnosis, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Koji Tokushige
- Center for Immune Therapeutics and Diagnosis, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yui Maehara
- Center for Immune Therapeutics and Diagnosis, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Saori Hirota
- Center for Immune Therapeutics and Diagnosis, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuko Kojima
- Laboratory of Morphology and Image Analysis, Biomedical Research Core Facilities, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Mamoru Ito
- Liver Engineering Laboratory, Department of Applied Research for Laboratory Animals, Central Institute for Experimental Animals, Kanagawa, Japan
| | - Nagako Sougawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan; Department of Physiology, Osaka Dental University, Osaka, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ko Okumura
- Center for Immune Therapeutics and Diagnosis, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Biofunctional Microbiota, Graduate School of Medicine, Juntendo University, Tokyo, Japan; Atopy (Allergy) Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Koichiro Uchida
- Center for Immune Therapeutics and Diagnosis, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|
25
|
Wulkan F, Romagnuolo R, Qiang B, Valdman Sadikov T, Kim KP, Quesnel E, Jiang W, Andharia N, Weyers JJ, Ghugre NR, Ozcan B, Alibhai FJ, Laflamme MA. Stem cell-derived cardiomyocytes expressing a dominant negative pacemaker HCN4 channel do not reduce the risk of graft-related arrhythmias. Front Cardiovasc Med 2024; 11:1374881. [PMID: 39045008 PMCID: PMC11263024 DOI: 10.3389/fcvm.2024.1374881] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 06/11/2024] [Indexed: 07/25/2024] Open
Abstract
Background Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) show tremendous promise for cardiac regeneration following myocardial infarction (MI), but their transplantation gives rise to transient ventricular tachycardia (VT) in large-animal MI models, representing a major hurdle to translation. Our group previously reported that these arrhythmias arise from a focal mechanism whereby graft tissue functions as an ectopic pacemaker; therefore, we hypothesized that hPSC-CMs engineered with a dominant negative form of the pacemaker ion channel HCN4 (dnHCN4) would exhibit reduced automaticity and arrhythmogenic risk following transplantation. Methods We used CRISPR/Cas9-mediated gene-editing to create transgenic dnHCN4 hPSC-CMs, and their electrophysiological behavior was evaluated in vitro by patch-clamp recordings and optical mapping. Next, we transplanted WT and homozygous dnHCN4 hPSC-CMs in a pig MI model and compared post-transplantation outcomes including the incidence of spontaneous arrhythmias and graft structure by immunohistochemistry. Results In vitro dnHCN4 hPSC-CMs exhibited significantly reduced automaticity and pacemaker funny current (I f ) density relative to wildtype (WT) cardiomyocytes. Following transplantation with either dnHCN4 or WT hPSC-CMs, all recipient hearts showed transmural infarct scar that was partially remuscularized by scattered islands of human myocardium. However, in contrast to our hypothesis, both dnHCN4 and WT hPSC-CM recipients exhibited frequent episodes of ventricular tachycardia (VT). Conclusions While genetic silencing of the pacemaker ion channel HCN4 suppresses the automaticity of hPSC-CMs in vitro, this intervention is insufficient to reduce VT risk post-transplantation in the pig MI model, implying more complex mechanism(s) are operational in vivo.
Collapse
Affiliation(s)
- Fanny Wulkan
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Rocco Romagnuolo
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Beiping Qiang
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | | | | | - Elya Quesnel
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Wenlei Jiang
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Naaz Andharia
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Jill J. Weyers
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Nilesh R. Ghugre
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
- Schulich Heart Program, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Bilgehan Ozcan
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Faisal J. Alibhai
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Michael A. Laflamme
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
26
|
Lin Y, Sato N, Hong S, Nakamura K, Ferrante EA, Yu ZX, Chen MY, Nakamura DS, Yang X, Clevenger RR, Hunt TJ, Taylor JL, Jeffries KR, Keeran KJ, Neidig LE, Mehta A, Schwartzbeck R, Yu SJ, Kelly C, Navarengom K, Takeda K, Adler SS, Choyke PL, Zou J, Murry CE, Boehm M, Dunbar CE. Long-term engraftment and maturation of autologous iPSC-derived cardiomyocytes in two rhesus macaques. Cell Stem Cell 2024; 31:974-988.e5. [PMID: 38843830 PMCID: PMC11227404 DOI: 10.1016/j.stem.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/14/2024] [Accepted: 05/10/2024] [Indexed: 07/08/2024]
Abstract
Cellular therapies with cardiomyocytes produced from induced pluripotent stem cells (iPSC-CMs) offer a potential route to cardiac regeneration as a treatment for chronic ischemic heart disease. Here, we report successful long-term engraftment and in vivo maturation of autologous iPSC-CMs in two rhesus macaques with small, subclinical chronic myocardial infarctions, all without immunosuppression. Longitudinal positron emission tomography imaging using the sodium/iodide symporter (NIS) reporter gene revealed stable grafts for over 6 and 12 months, with no teratoma formation. Histological analyses suggested capability of the transplanted iPSC-CMs to mature and integrate with endogenous myocardium, with no sign of immune cell infiltration or rejection. By contrast, allogeneic iPSC-CMs were rejected within 8 weeks of transplantation. This study provides the longest-term safety and maturation data to date in any large animal model, addresses concerns regarding neoantigen immunoreactivity of autologous iPSC therapies, and suggests that autologous iPSC-CMs would similarly engraft and mature in human hearts.
Collapse
Affiliation(s)
- Yongshun Lin
- iPSC Core, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Noriko Sato
- Laboratory of Cellular Therapeutics, Molecular Imaging Branch, National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Sogun Hong
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Kenta Nakamura
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA; Department of Medicine/Cardiology, University of Washington, Seattle, WA 98195, USA
| | - Elisa A Ferrante
- Translational Vascular Medicine Branch, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Zu Xi Yu
- Pathology Core, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Marcus Y Chen
- Cardiovascular Branch, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Daisy S Nakamura
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA; Department of Medicine/Cardiology, University of Washington, Seattle, WA 98195, USA
| | - Xiulan Yang
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA; Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | | | - Timothy J Hunt
- Animal Surgery and Resources Core, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Joni L Taylor
- Animal Surgery and Resources Core, NHLBI, NIH, Bethesda, MD 20892, USA
| | | | - Karen J Keeran
- Animal Surgery and Resources Core, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Lauren E Neidig
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA; Department of Comparative Medicine, University of Washington, Seattle, WA 98195, USA
| | - Atul Mehta
- Translational Vascular Medicine Branch, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Robin Schwartzbeck
- Translational Vascular Medicine Branch, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Shiqin Judy Yu
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Conor Kelly
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Keron Navarengom
- Translational Vascular Medicine Branch, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Kazuyo Takeda
- Microscopy and Imaging Core, CBER, FDA, Silver Spring, MD, USA
| | - Stephen S Adler
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Peter L Choyke
- Laboratory of Cellular Therapeutics, Molecular Imaging Branch, National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Jizhong Zou
- iPSC Core, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Charles E Murry
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA; Department of Medicine/Cardiology, University of Washington, Seattle, WA 98195, USA; Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA 98195, USA; Department of Bioengineering, University of Washington School of Medicine, Seattle, WA 98195, USA.
| | - Manfred Boehm
- Translational Vascular Medicine Branch, NHLBI, NIH, Bethesda, MD 20892, USA.
| | - Cynthia E Dunbar
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
27
|
Liu T, Hao Y, Zhang Z, Zhou H, Peng S, Zhang D, Li K, Chen Y, Chen M. Advanced Cardiac Patches for the Treatment of Myocardial Infarction. Circulation 2024; 149:2002-2020. [PMID: 38885303 PMCID: PMC11191561 DOI: 10.1161/circulationaha.123.067097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Myocardial infarction is a cardiovascular disease characterized by a high incidence rate and mortality. It leads to various cardiac pathophysiological changes, including ischemia/reperfusion injury, inflammation, fibrosis, and ventricular remodeling, which ultimately result in heart failure and pose a significant threat to global health. Although clinical reperfusion therapies and conventional pharmacological interventions improve emergency survival rates and short-term prognoses, they are still limited in providing long-lasting improvements in cardiac function or reversing pathological progression. Recently, cardiac patches have gained considerable attention as a promising therapy for myocardial infarction. These patches consist of scaffolds or loaded therapeutic agents that provide mechanical reinforcement, synchronous electrical conduction, and localized delivery within the infarct zone to promote cardiac restoration. This review elucidates the pathophysiological progression from myocardial infarction to heart failure, highlighting therapeutic targets and various cardiac patches. The review considers the primary scaffold materials, including synthetic, natural, and conductive materials, and the prevalent fabrication techniques and optimal properties of the patch, as well as advanced delivery strategies. Last, the current limitations and prospects of cardiac patch research are considered, with the goal of shedding light on innovative products poised for clinical application.
Collapse
Affiliation(s)
- Tailuo Liu
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases (T.L., Y.H., H.Z., S.P., D.Z., Y.C., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
- Department of Cardiology (T.L., S.P., D.Z., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, PR China (T.L., K.L., Y.C.)
| | - Ying Hao
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases (T.L., Y.H., H.Z., S.P., D.Z., Y.C., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
| | - Zixuan Zhang
- West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, PR China (Z.Z.)
| | - Hao Zhou
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases (T.L., Y.H., H.Z., S.P., D.Z., Y.C., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
| | - Shiqin Peng
- Department of Cardiology (T.L., S.P., D.Z., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
| | - Dingyi Zhang
- Department of Cardiology (T.L., S.P., D.Z., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
| | - Ka Li
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, PR China (T.L., K.L., Y.C.)
| | - Yuwen Chen
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, PR China (T.L., K.L., Y.C.)
| | - Mao Chen
- Department of Cardiology (T.L., S.P., D.Z., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
| |
Collapse
|
28
|
von Bibra C, Hinkel R. Non-human primate studies for cardiomyocyte transplantation-ready for translation? Front Pharmacol 2024; 15:1408679. [PMID: 38962314 PMCID: PMC11221829 DOI: 10.3389/fphar.2024.1408679] [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: 03/28/2024] [Accepted: 05/21/2024] [Indexed: 07/05/2024] Open
Abstract
Non-human primates (NHP) are valuable models for late translational pre-clinical studies, often seen as a last step before clinical application. The unique similarity between NHPs and humans is often the subject of ethical concerns. However, it is precisely this analogy in anatomy, physiology, and the immune system that narrows the translational gap to other animal models in the cardiovascular field. Cell and gene therapy approaches are two dominant strategies investigated in the research field of cardiac regeneration. Focusing on the cell therapy approach, several xeno- and allogeneic cell transplantation studies with a translational motivation have been realized in macaque species. This is based on the pressing need for novel therapeutic options for heart failure patients. Stem cell-based remuscularization of the injured heart can be achieved via direct injection of cardiomyocytes (CMs) or patch application. Both CM delivery approaches are in the late preclinical stage, and the first clinical trials have started. However, are we already ready for the clinical area? The present review concentrates on CM transplantation studies conducted in NHPs, discusses the main sources and discoveries, and provides a perspective about human translation.
Collapse
Affiliation(s)
- Constantin von Bibra
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behavior, Stiftung Tieraerztliche Hochschule Hannover, University of Veterinary Medicine, Hanover, Germany
- Laboratory Animal Science Unit, German Primate Center, Leibniz Institute for Primate Research, Goettingen, Germany
- DZHK (German Centre of Cardiovascular Research), Partner Site Lower Saxony, Goettingen, Germany
| | - Rabea Hinkel
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behavior, Stiftung Tieraerztliche Hochschule Hannover, University of Veterinary Medicine, Hanover, Germany
- Laboratory Animal Science Unit, German Primate Center, Leibniz Institute for Primate Research, Goettingen, Germany
- DZHK (German Centre of Cardiovascular Research), Partner Site Lower Saxony, Goettingen, Germany
| |
Collapse
|
29
|
Farahzadi R, Fathi E, Valipour B, Ghaffary S. Stem cells-derived exosomes as cardiac regenerative agents. IJC HEART & VASCULATURE 2024; 52:101399. [PMID: 38584674 PMCID: PMC10990901 DOI: 10.1016/j.ijcha.2024.101399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/03/2024] [Accepted: 03/28/2024] [Indexed: 04/09/2024]
Abstract
Heart failure is a root cause of morbidity and mortality worldwide. Due to the limited regenerative capacity of the heart following myocardial injury, stem cell-based therapies have been considered a hopeful approach for improving cardiac regeneration. In recent years, different kinds of cell products have been investigated regarding their potential to treat patients with heart failure. Despite special attention to cell therapy and its products, therapeutic efficacy has been disappointing, and clinical application is not affordable. In the past few years, a subset of small extracellular vehicles (EVs), commonly known as "exosomes," was reported to grant regenerative and cardioprotective signals at a value similar to their donor cells. The conceptual advantage is that they may be ideally used without evoking a relevant recipient immune response or other adverse effects associated with viable cells. The evidence related to their beneficial effects in animal models of heart failure is rapidly growing. However, there is remarkable heterogeneity regarding source cells, isolation process, effective dosage, and delivery mode. This brief review will focus on the latest research and debates on regenerative potential and cardiac repair of exosomes from different sources, such as cardiac/non-cardiac stem, somatic cells, and progenitor cells. Overall, the current state of research on exosomes as an experimental therapy for heart diseases will be discussed.
Collapse
Affiliation(s)
- Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Medical Philosophy and History Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Behnaz Valipour
- Department of Anatomical Sciences, Sarab Faculty of Medical Sciences, Sarab, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saba Ghaffary
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
30
|
Xiao Y, Xia L, Jiang W, Qin J, Zhao L, Li Z, Huang L, Li K, Yu P, Wei L, Jiang X, Chen Z, Yu X. Cardiopulmonary progenitors facilitate cardiac repair via exosomal transfer of miR-27b-3p targeting the SIK1-CREB1 axis. Cell Prolif 2024; 57:e13593. [PMID: 38185757 PMCID: PMC11056695 DOI: 10.1111/cpr.13593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/27/2023] [Accepted: 12/17/2023] [Indexed: 01/09/2024] Open
Abstract
Ischemic heart disease, especially myocardial infarction (MI), is one of the leading causes of death worldwide, and desperately needs effective treatments, such as cell therapy. Cardiopulmonary progenitors (CPPs) are stem cells for both heart and lung, but their repairing role in damaged heart is still unknown. Here, we obtained CPPs from E9.5 mouse embryos, maintained their stemness while expanding, and identified their characteristics by scRNA-seq, flow cytometry, quantitative reverse transcription-polymerase chain reaction, and differentiation assays. Moreover, we employed mouse MI model to investigate whether CPPs could repair the injured heart. Our data identified that CPPs exhibit hybrid fibroblastic, endothelial, and mesenchymal state, and they could differentiate into cell lineages within the cardiopulmonary system. Moreover, intramyocardial injection of CPPs improves cardiac function through CPPs exosomes (CPPs-Exo) by promotion of cardiomyocytic proliferation and vascularization. To uncover the underlying mechanism, we used miRNA-seq, bulk RNA-seq, and bioinformatic approaches, and found the highly expressed miR-27b-3p in CPPs-Exo and its target gene Sik1, which can influence the transcriptional activity of CREB1. Therefore, we postulate that CPPs facilitate cardiac repair partially through the SIK1-CREB1 axis via exosomal miR-27b-3p. Our study offers a novel insight into the role of CPPs-Exo in heart repair and highlights the potential of CPPs-Exo as a promising therapeutic strategy for MI.
Collapse
Affiliation(s)
- Ying‐Ying Xiao
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
- Department of Pharmacy, The First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Luo‐Xing Xia
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Wen‐Jing Jiang
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Jian‐Feng Qin
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Li‐Xin Zhao
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Zhan Li
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Li‐Juan Huang
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Ke‐Xin Li
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Peng‐Jiu Yu
- Department of Pharmacy, The First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Li Wei
- Department of Pharmacy, The First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Xue‐Yan Jiang
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Zhe‐Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Institute for BiotechnologySt. John's UniversityQueensNew YorkUSA
| | - Xi‐Yong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| |
Collapse
|
31
|
Carvalho AB, Kasai-Brunswick TH, Campos de Carvalho AC. Advanced cell and gene therapies in cardiology. EBioMedicine 2024; 103:105125. [PMID: 38640834 PMCID: PMC11052923 DOI: 10.1016/j.ebiom.2024.105125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 04/01/2024] [Accepted: 04/04/2024] [Indexed: 04/21/2024] Open
Abstract
We review the evidence for the presence of stem/progenitor cells in the heart and the preclinical and clinical data using diverse cell types for the therapy of cardiac diseases. We highlight the failure of adult stem/progenitor cells to ameliorate heart function in most cardiac diseases, with the possible exception of refractory angina. The use of pluripotent stem cell-derived cardiomyocytes is analysed as a viable alternative therapeutic option but still needs further research at preclinical and clinical stages. We also discuss the use of direct reprogramming of cardiac fibroblasts into cardiomyocytes and the use of extracellular vesicles as therapeutic agents in ischemic and non-ischemic cardiac diseases. Finally, gene therapies and genome editing for the treatment of hereditary cardiac diseases, ablation of genes responsible for atherosclerotic disease, or modulation of gene expression in the heart are discussed.
Collapse
Affiliation(s)
- Adriana Bastos Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Universidade Federal do RIo de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Tais Hanae Kasai-Brunswick
- Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Universidade Federal do RIo de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Antonio Carlos Campos de Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Universidade Federal do RIo de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
32
|
Eschenhagen T, Weinberger F. Challenges and perspectives of heart repair with pluripotent stem cell-derived cardiomyocytes. NATURE CARDIOVASCULAR RESEARCH 2024; 3:515-524. [PMID: 39195938 DOI: 10.1038/s44161-024-00472-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 04/04/2024] [Indexed: 08/29/2024]
Abstract
Here we aim at providing a concise but comprehensive overview of the perspectives and challenges of heart repair with pluripotent stem cell-derived cardiomyocytes. This Review comes at a time when consensus has been reached about the lack of relevant proliferative capacity of adult mammalian cardiomyocytes and the lack of new heart muscle formation with autologous cell sources. While alternatives to cell-based approaches will be shortly summarized, the focus lies on pluripotent stem cell-derived cardiomyocyte repair, which entered first clinical trials just 2 years ago. In the view of the authors, these early trials are important but have to be viewed as early proof-of-concept trials in humans that will hopefully provide first answers on feasibility, safety and the survival of allogeneic pluripotent stem cell-derived cardiomyocyte in the human heart. Better approaches have to be developed to make this approach clinically applicable.
Collapse
Affiliation(s)
- Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany.
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| | - Florian Weinberger
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| |
Collapse
|
33
|
Cai S, Dai Q. Progress in preclinical research on induced pluripotent stem cell therapy for acute myocardial infarction. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:244-253. [PMID: 38594961 PMCID: PMC11057988 DOI: 10.3724/zdxbyxb-2023-0402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 03/09/2024] [Indexed: 04/11/2024]
Abstract
Induced pluripotent stem cells (iPSCs) are obtained by introducing exogenous genes or adding chemicals to the culture medium to induce somatic cell differentiation. Similarly to embryonic stem cells, iPSCs have the ability to differentiate into all three embryonic cell lines. iPSCs can differentiate into cardiac muscle cells through two-dimensional differentiation methods such as monolayer cell culture and co-culture, or through embryoid body and scaffold-based three-dimensional differentiation methods. In addition, the process of iPSCs differentiation into cardiac muscle cells also requires activation or inhibition of specific signaling pathways,such as Wnt, BMP, Notch signaling pathways to mimic the development of the heart in vivo. In recent years, suspension culturing in bioreactors has been shown to produce large number of iPSCs derived cardiac muscle cells (iPSC-CMs). Before transplantation, it is necessary to purify iPSC-CMs through metabolic regulation or cell sorting to eliminate undifferentiated iPSCs, which may lead to teratoma formation. The transplantation methods for iPSC-CMs are mainly injection of cell suspension and transplantation of cell patches into the infarcted myocardium. Animal studies have shown that transplantation of iPSC-CMs into the infarcted myocardium can improve cardiac function. This article reviews the progress in preclinical studies on iPSC-CMs therapy for acute myocardial infarction and discusses the limitations and challenges of its clinical application to provide references for further clinical research and application.
Collapse
Affiliation(s)
- Songyan Cai
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China.
| | - Qingyuan Dai
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China.
| |
Collapse
|
34
|
Luo ZR, Meng WT, Li H, Wang Y, Wang YC, Zhao Y, Lu PP, Yuan Y, Huang W, Guo HD. Transplantation of induced pluripotent stem cells-derived cardiomyocytes combined with modified Taohong Siwu decoction improved heart repair after myocardial infarction. Heliyon 2024; 10:e26700. [PMID: 38434034 PMCID: PMC10906439 DOI: 10.1016/j.heliyon.2024.e26700] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 02/02/2024] [Accepted: 02/19/2024] [Indexed: 03/05/2024] Open
Abstract
Objective This study aimed to study whether modified Taohong Siwu decoction (MTHSWD) combined with human induced pluripotent stem cells-derived cardiomyocytes (iPS-CMs) transplantation can promote cardiac function in myocardial infarction (MI) nude mouse model and explore its possible mechanism. Methods The MI mouse model was established by the ligation of left anterior descending coronary artery. After 4 weeks of gavage of MTHSWD combined with iPS-CMs transplantation, the changes in heart function of mice were examined by echocardiography. The histological changes were observed by Masson's trichrome staining. The survival and differentiation of transplanted cells were detected by double immunofluorescence staining of human nuclear antigen (HNA) and cardiac troponin T (cTnT). The number of c-kit-positive cells in the infarct area were evaluated by immunofluorescent staining. The levels of stromal cell-derived factor 1 (SDF-1), stem cell factor (SCF), vascular endothelial growth factor (VEGF) and basic fibroblast growth factor in infarcted myocardium tissues were detected by ELISA. Results MTHSWD combined with iPS-CMs transplantation can improve the heart function of MI mice, reduce the infarct size and collagen deposition in infarct area. By immunofluorescence double-label detection of HNA and cTnT, it was found that MTHSWD combined with iPS-CMs transplantation can improve the survival and maturation of iPS-CMs. In addition, MTHSWD combined with iPS-CMs transplantation can activate more endogenous c-kit positive cardiac mesenchymal cells, and significantly increase the content of SDF-1, SCF and VEGF in myocardial tissues. Conclusions The combination of MTHSWD with iPS-CMs transplantation promoted cardiac function of nude mice with MI by improving the survival and maturation of iPS-CMs in the infarct area, activating the endogenous c-kit positive cardiac mesenchymal cells, and increasing paracrine.
Collapse
Affiliation(s)
- Zhi-rong Luo
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wan-ting Meng
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Han Li
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yu Wang
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ya-chao Wang
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yue Zhao
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ping-ping Lu
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yuan Yuan
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wei Huang
- Department of Chinese Internal Medicine, Dahua Hospital, Xuhui District, Shanghai, China
| | - Hai-dong Guo
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
35
|
Li J, Hua Y, Liu Y, Qu X, Zhang J, Ishida M, Yoshida N, Tabata A, Miyoshi H, Shiba M, Higo S, Sougawa N, Takeda M, Kawamura T, Matsuura R, Okuzaki D, Toyofuku T, Sawa Y, Liu L, Miyagawa S. Human induced pluripotent stem cell-derived closed-loop cardiac tissue for drug assessment. iScience 2024; 27:108992. [PMID: 38333703 PMCID: PMC10850789 DOI: 10.1016/j.isci.2024.108992] [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: 09/05/2023] [Revised: 11/16/2023] [Accepted: 01/18/2024] [Indexed: 02/10/2024] Open
Abstract
Human iPSC-derived cardiomyocytes (hiPSC-CMs) exhibit functional immaturity, potentially impacting their suitability for assessing drug proarrhythmic potential. We previously devised a traveling wave (TW) system to promote maturation in 3D cardiac tissue. To align with current drug assessment paradigms (CiPA and JiCSA), necessitating a 2D monolayer cardiac tissue, we integrated the TW system with a multi-electrode array. This gave rise to a hiPSC-derived closed-loop cardiac tissue (iCT), enabling spontaneous TW initiation and swift pacing of cardiomyocytes from various cell lines. The TW-paced cardiomyocytes demonstrated heightened sarcomeric and functional maturation, exhibiting enhanced response to isoproterenol. Moreover, these cells showcased diminished sensitivity to verapamil and maintained low arrhythmia rates with ranolazine-two drugs associated with a low risk of torsades de pointes (TdP). Notably, the TW group displayed increased arrhythmia rates with high and intermediate risk TdP drugs (quinidine and pimozide), underscoring the potential utility of this system in drug assessment applications.
Collapse
Affiliation(s)
- Junjun Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Ying Hua
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yuting Liu
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Xiang Qu
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Jingbo Zhang
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Masako Ishida
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Noriko Yoshida
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Akiko Tabata
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Hayato Miyoshi
- Fujifilm Corporation, Ashigarakami 258-8577, Kanagawa, Japan
| | - Mikio Shiba
- Cardiovascular Division, Osaka Police Hospital, Tennoji 543-0035, Osaka, Japan
| | - Shuichiro Higo
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita 565-0871, Osaka, Japan
- Department of Medical Therapeutics for Heart Failure, Osaka University Graduate School of Medicine, Suita 565-0871, Osaka, Japan
| | - Nagako Sougawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
- Department of Physiology, Osaka Dental University, 8-1 Kuzuha Hanazono-cho, Hirakata 573-1121, Osaka, Japan
| | - Maki Takeda
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Takuji Kawamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Ryohei Matsuura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Daisuke Okuzaki
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Research Center, Osaka University, Osaka, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Toshihiko Toyofuku
- Department of Immunology and Molecular Medicine, Graduate School of Medicine, Osaka University, Suita 565-0871, Osaka, Japan
| | - Yoshiki Sawa
- Department of Future Medicine, Division of Health Science, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Li Liu
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| |
Collapse
|
36
|
Soma Y, Tani H, Morita-Umei Y, Kishino Y, Fukuda K, Tohyama S. Pluripotent stem cell-based cardiac regenerative therapy for heart failure. J Mol Cell Cardiol 2024; 187:90-100. [PMID: 38331557 DOI: 10.1016/j.yjmcc.2023.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 12/03/2023] [Accepted: 12/04/2023] [Indexed: 02/10/2024]
Abstract
Cardiac regenerative therapy using human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) is expected to become an alternative to heart transplantation for severe heart failure. It is now possible to produce large numbers of human pluripotent stem cells (hPSCs) and eliminate non-cardiomyocytes, including residual undifferentiated hPSCs, which can cause teratoma formation after transplantation. There are two main strategies for transplanting hPSC-CMs: injection of hPSC-CMs into the myocardium from the epicardial side, and implantation of hPSC-CM patches or engineered heart tissues onto the epicardium. Transplantation of hPSC-CMs into the myocardium of large animals in a myocardial infarction model improved cardiac function. The engrafted hPSC-CMs matured, and microvessels derived from the host entered the graft abundantly. Furthermore, as less invasive methods using catheters, injection into the coronary artery and injection into the myocardium from the endocardium side have recently been investigated. Since transplantation of hPSC-CMs alone has a low engraftment rate, various methods such as transplantation with the extracellular matrix or non-cardiomyocytes and aggregation of hPSC-CMs have been developed. Post-transplant arrhythmias, imaging of engrafted hPSC-CMs, and immune rejection are the remaining major issues, and research is being conducted to address them. The clinical application of cardiac regenerative therapy using hPSC-CMs has just begun and is expected to spread widely if its safety and efficacy are proven in the near future.
Collapse
Affiliation(s)
- Yusuke Soma
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Hidenori Tani
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan; Joint Research Laboratory for Medical Innovation in Heart Disease, Keio University School of Medicine, Tokyo, Japan
| | - Yuika Morita-Umei
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan; Kanagawa Institute of Industrial Science and Technology (KISTEC), Kanagawa, Japan
| | - Yoshikazu Kishino
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan.
| | - Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
37
|
Abstract
Permanent fibrosis and chronic deterioration of heart function in patients after myocardial infarction present a major health-care burden worldwide. In contrast to the restricted potential for cellular and functional regeneration of the adult mammalian heart, a robust capacity for cardiac regeneration is seen during the neonatal period in mammals as well as in the adults of many fish and amphibian species. However, we lack a complete understanding as to why cardiac regeneration takes place more efficiently in some species than in others. The capacity of the heart to regenerate after injury is controlled by a complex network of cellular and molecular mechanisms that form a regulatory landscape, either permitting or restricting regeneration. In this Review, we provide an overview of the diverse array of vertebrates that have been studied for their cardiac regenerative potential and discuss differential heart regeneration outcomes in closely related species. Additionally, we summarize current knowledge about the core mechanisms that regulate cardiac regeneration across vertebrate species.
Collapse
Affiliation(s)
- Michael Weinberger
- Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford, UK
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Paul R Riley
- Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
38
|
Al-attar R, Jargstorf J, Romagnuolo R, Jouni M, Alibhai FJ, Lampe PD, Solan JL, Laflamme MA. Casein Kinase 1 Phosphomimetic Mutations Negatively Impact Connexin-43 Gap Junctions in Human Pluripotent Stem Cell-Derived Cardiomyocytes. Biomolecules 2024; 14:61. [PMID: 38254663 PMCID: PMC10813327 DOI: 10.3390/biom14010061] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/14/2023] [Accepted: 12/25/2023] [Indexed: 01/24/2024] Open
Abstract
The transplantation of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) has shown promise in preclinical models of myocardial infarction, but graft myocardium exhibits incomplete host-graft electromechanical integration and a propensity for pro-arrhythmic behavior. Perhaps contributing to this situation, hPSC-CM grafts show low expression of connexin 43 (Cx43), the major gap junction (GJ) protein, in ventricular myocardia. We hypothesized that Cx43 expression and function could be rescued by engineering Cx43 in hPSC-CMs with a series of phosphatase-resistant mutations at three casein kinase 1 phosphorylation sites (Cx43-S3E) that have been previously reported to stabilize Cx43 GJs and reduce arrhythmias in transgenic mice. However, contrary to our predictions, transgenic Cx43-S3E hPSC-CMs exhibited reduced Cx43 expression relative to wild-type cells, both at baseline and following ischemic challenge. Cx43-S3E hPSC-CMs showed correspondingly slower conduction velocities, increased automaticity, and differential expression of other connexin isoforms and various genes involved in cardiac excitation-contraction coupling. Cx43-S3E hPSC-CMs also had phosphorylation marks associated with Cx43 GJ internalization, a finding that may account for their impaired GJ localization. Taken collectively, our data indicate that the Cx43-S3E mutation behaves differently in hPSC-CMs than in adult mouse ventricular myocytes and that multiple biological factors likely need to be addressed synchronously to ensure proper Cx43 expression, localization, and function.
Collapse
Affiliation(s)
- Rasha Al-attar
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (R.A.-a.); (J.J.); (R.R.); (M.J.); (F.J.A.)
| | - Joseph Jargstorf
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (R.A.-a.); (J.J.); (R.R.); (M.J.); (F.J.A.)
| | - Rocco Romagnuolo
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (R.A.-a.); (J.J.); (R.R.); (M.J.); (F.J.A.)
| | - Mariam Jouni
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (R.A.-a.); (J.J.); (R.R.); (M.J.); (F.J.A.)
| | - Faisal J. Alibhai
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (R.A.-a.); (J.J.); (R.R.); (M.J.); (F.J.A.)
| | - Paul D. Lampe
- Translational Research Program, Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (P.D.L.); (J.L.S.)
| | - Joell L. Solan
- Translational Research Program, Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (P.D.L.); (J.L.S.)
| | - Michael A. Laflamme
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (R.A.-a.); (J.J.); (R.R.); (M.J.); (F.J.A.)
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5G 1L7, Canada
| |
Collapse
|
39
|
Fang YH, Wang SPH, Liao IC, Tsai KJ, Huang PH, Yang PJ, Yen CJ, Liu PY, Shan YS, Liu YW. HLA-E high /HLA-G high /HLA-II low Human iPSC-Derived Cardiomyocytes Exhibit Low Immunogenicity for Heart Regeneration. Adv Healthc Mater 2023; 12:e2301186. [PMID: 37672681 DOI: 10.1002/adhm.202301186] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 08/31/2023] [Indexed: 09/08/2023]
Abstract
Although human pluripotent stem cells (hPSCs)-derived cardiomyocytes (hPSC-CMs) can remuscularize infarcted hearts and restore post-infarct cardiac function, post-transplant rejection resulting from human leukocyte antigen (HLA) mismatching is an enormous obstacle. It is crucial to identify hypoimmunogenic hPSCs for allogeneic cell therapy. This study is conducted to demonstrate the immune privilege of HLA-Ehigh /HLA-Ghigh /HLA-IIlow human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (hiPSC-CMs). Ischemia-reperfusion surgery is done to create transmural myocardial infarction in rats. At post-infarct 4 days, hPSC-CMs (1.0×107 cells per kg), including human embryonic stem cell-derived cardiomyocytes (hESC-CMs), HLA-Elow/HLA-Glow/HLA-IIhigh hiPSC-CMs, and HLA-Ehigh /HLA-Ghigh /HLA-IIlow hiPSC-CMs, are injected into the infarcted myocardium. Under the treatment of very low dose cyclosporine A (CsA), only HLA-Ehigh /HLA-Ghigh /HLA-IIlow hiPSC-CMs survive in vivo and improved post-infarct cardiac function with infarct size reduction. HLA-Ehigh /HLA-Ghigh /HLA-IIlow hiPSC-CMs activate the SHP-1 signaling pathway of natural killer (NK) cells and cytotoxic T cells to evade attack by NK cells and cytotoxic T cells. Herein, it is demonstrated that using a clinically relevant CsA dose, HLA-Ehigh /HLA-Ghigh /HLA-IIlow hiPSC-CMs repair the infarcted myocardium and restore the post-infarct heart function. HLA-Ehigh /HLA-Ghigh /HLA-IIlow hiPSCs are less immunogenic and may serve as platforms for regeneration medicine.
Collapse
Affiliation(s)
- Yi-Hsien Fang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan
- Center of Cell Therapy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
| | - Saprina P H Wang
- Center of Cell Therapy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
| | - I-Chuang Liao
- Department of Pathology, Chi-Mei Medical Center, Tainan, 71004, Taiwan
| | - Kuen-Jer Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan
- Center of Cell Therapy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
| | - Po-Hsien Huang
- Center of Cell Therapy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Pei-Jung Yang
- Center of Cell Therapy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
| | - Chia-Jui Yen
- Center of Cell Therapy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
| | - Ping-Yen Liu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
| | - Yan-Shen Shan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan
- Center of Cell Therapy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
| | - Yen-Wen Liu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan
- Center of Cell Therapy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
| |
Collapse
|
40
|
Cheng YC, Hsieh ML, Lin CJ, Chang CMC, Huang CY, Puntney R, Wu Moy A, Ting CY, Herr Chan DZ, Nicholson MW, Lin PJ, Chen HC, Kim GC, Zhang J, Coonen J, Basu P, Simmons HA, Liu YW, Hacker TA, Kamp TJ, Hsieh PCH. Combined Treatment of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes and Endothelial Cells Regenerate the Infarcted Heart in Mice and Non-Human Primates. Circulation 2023; 148:1395-1409. [PMID: 37732466 PMCID: PMC10683868 DOI: 10.1161/circulationaha.122.061736] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/23/2023] [Indexed: 09/22/2023]
Abstract
BACKGROUND Remuscularization of the mammalian heart can be achieved after cell transplantation of human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs). However, several hurdles remain before implementation into clinical practice. Poor survival of the implanted cells is related to insufficient vascularization, and the potential for fatal arrhythmogenesis is associated with the fetal cell-like nature of immature CMs. METHODS We generated 3 lines of hiPSC-derived endothelial cells (ECs) and hiPSC-CMs from 3 independent donors and tested hiPSC-CM sarcomeric length, gap junction protein, and calcium-handling ability in coculture with ECs. Next, we examined the therapeutic effect of the cotransplantation of hiPSC-ECs and hiPSC-CMs in nonobese diabetic-severe combined immunodeficiency (NOD-SCID) mice undergoing myocardial infarction (n≥4). Cardiac function was assessed by echocardiography, whereas arrhythmic events were recorded using 3-lead ECGs. We further used healthy non-human primates (n=4) with cell injection to study the cell engraftment, maturation, and integration of transplanted hiPSC-CMs, alone or along with hiPSC-ECs, by histological analysis. Last, we tested the cell therapy in ischemic reperfusion injury in non-human primates (n=4, 3, and 4 for EC+CM, CM, and control, respectively). Cardiac function was evaluated by echocardiography and cardiac MRI, whereas arrhythmic events were monitored by telemetric ECG recorders. Cell engraftment, angiogenesis, and host-graft integration of human grafts were also investigated. RESULTS We demonstrated that human iPSC-ECs promote the maturity and function of hiPSC-CMs in vitro and in vivo. When cocultured with ECs, CMs showed more mature phenotypes in cellular structure and function. In the mouse model, cotransplantation augmented the EC-accompanied vascularization in the grafts, promoted the maturity of CMs at the infarct area, and improved cardiac function after myocardial infarction. Furthermore, in non-human primates, transplantation of ECs and CMs significantly enhanced graft size and vasculature and improved cardiac function after ischemic reperfusion. CONCLUSIONS These results demonstrate the synergistic effect of combining iPSC-derived ECs and CMs for therapy in the postmyocardial infarction heart, enabling a promising strategy toward clinical translation.
Collapse
Affiliation(s)
- Yu-Che Cheng
- Institute of Biomedical Sciences, Academia Sinica, Taiwan (Y.C.C., C.J.L., C.Y.H., C.Y.T., D.Z.H.C., M.W.N., P.J.L., H.C.C., P.C.H.H.)
| | - Marvin L Hsieh
- Model Organisms Research Core, Department of Medicine (M.L.H., C.M.C.C., T.A.H.), University of Wisconsin-Madison
| | - Chen-Ju Lin
- Institute of Biomedical Sciences, Academia Sinica, Taiwan (Y.C.C., C.J.L., C.Y.H., C.Y.T., D.Z.H.C., M.W.N., P.J.L., H.C.C., P.C.H.H.)
| | - Cindy M C Chang
- Model Organisms Research Core, Department of Medicine (M.L.H., C.M.C.C., T.A.H.), University of Wisconsin-Madison
| | - Ching-Ying Huang
- Institute of Biomedical Sciences, Academia Sinica, Taiwan (Y.C.C., C.J.L., C.Y.H., C.Y.T., D.Z.H.C., M.W.N., P.J.L., H.C.C., P.C.H.H.)
| | - Riley Puntney
- Wisconsin National Primate Research Center (R.P., A.W.M., J.C., P.B., H.A.S.), University of Wisconsin-Madison
| | - Amy Wu Moy
- Wisconsin National Primate Research Center (R.P., A.W.M., J.C., P.B., H.A.S.), University of Wisconsin-Madison
| | - Chien-Yu Ting
- Institute of Biomedical Sciences, Academia Sinica, Taiwan (Y.C.C., C.J.L., C.Y.H., C.Y.T., D.Z.H.C., M.W.N., P.J.L., H.C.C., P.C.H.H.)
| | - Darien Zhing Herr Chan
- Institute of Biomedical Sciences, Academia Sinica, Taiwan (Y.C.C., C.J.L., C.Y.H., C.Y.T., D.Z.H.C., M.W.N., P.J.L., H.C.C., P.C.H.H.)
| | - Martin W Nicholson
- Institute of Biomedical Sciences, Academia Sinica, Taiwan (Y.C.C., C.J.L., C.Y.H., C.Y.T., D.Z.H.C., M.W.N., P.J.L., H.C.C., P.C.H.H.)
| | - Po-Ju Lin
- Institute of Biomedical Sciences, Academia Sinica, Taiwan (Y.C.C., C.J.L., C.Y.H., C.Y.T., D.Z.H.C., M.W.N., P.J.L., H.C.C., P.C.H.H.)
| | - Hung-Chih Chen
- Institute of Biomedical Sciences, Academia Sinica, Taiwan (Y.C.C., C.J.L., C.Y.H., C.Y.T., D.Z.H.C., M.W.N., P.J.L., H.C.C., P.C.H.H.)
| | - Gina C Kim
- Department of Medicine and Stem Cell and Regenerative Medicine Center (G.C.K., J.Z., T.J.K., P.C.H.H.), University of Wisconsin-Madison
| | - Jianhua Zhang
- Department of Medicine and Stem Cell and Regenerative Medicine Center (G.C.K., J.Z., T.J.K., P.C.H.H.), University of Wisconsin-Madison
| | - Jennifer Coonen
- Wisconsin National Primate Research Center (R.P., A.W.M., J.C., P.B., H.A.S.), University of Wisconsin-Madison
| | - Puja Basu
- Wisconsin National Primate Research Center (R.P., A.W.M., J.C., P.B., H.A.S.), University of Wisconsin-Madison
| | - Heather A Simmons
- Wisconsin National Primate Research Center (R.P., A.W.M., J.C., P.B., H.A.S.), University of Wisconsin-Madison
| | - Yen-Wen Liu
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan (Y.W.L.)
| | - Timothy A Hacker
- Model Organisms Research Core, Department of Medicine (M.L.H., C.M.C.C., T.A.H.), University of Wisconsin-Madison
| | - Timothy J Kamp
- Department of Medicine and Stem Cell and Regenerative Medicine Center (G.C.K., J.Z., T.J.K., P.C.H.H.), University of Wisconsin-Madison
| | - Patrick C H Hsieh
- Institute of Biomedical Sciences, Academia Sinica, Taiwan (Y.C.C., C.J.L., C.Y.H., C.Y.T., D.Z.H.C., M.W.N., P.J.L., H.C.C., P.C.H.H.)
- Department of Medicine and Stem Cell and Regenerative Medicine Center (G.C.K., J.Z., T.J.K., P.C.H.H.), University of Wisconsin-Madison
- Institute of Medical Genomics and Proteomics and Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan (P.C.H.H.)
| |
Collapse
|
41
|
Fukuda K. Establishment and Industrialization of a New Treatment Method Using Regenerative Cardiomyocyte Transplantation for Refractory Severe Heart Failure - Secondary Publication. JMA J 2023; 6:388-392. [PMID: 37941693 PMCID: PMC10628313 DOI: 10.31662/jmaj.2023-0075] [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: 05/15/2023] [Accepted: 06/01/2023] [Indexed: 11/10/2023] Open
Abstract
Cardiomyocytes undergo cell division during the fetal period but do not divide after birth; thus, they grow into adult heart cells by enlarging their size. Therefore, heart failure occurs when a certain number of cardiomyocytes are lost owing to myocardial infarction, myocarditis, sarcoidosis, etc. Through scientific efforts, we have developed methods to safely and efficiently generate induced pluripotent stem (iPS) cells from peripheral blood T cells, generate ventricle-specific cardiomyocytes from iPS cells, and remove residual iPS cells and non-cardiomyocytes using the "metabolic selection method" and purify the cardiomyocytes from iPS cell derivatives. We have also developed the technology to mass-produce and efficiently engraft cardiomyocytes by generating cardiomyocyte spheroids and have developed devices suitable for cell transplantation. We have confirmed the safety and efficacy of these techniques by performing preclinical studies (oncogenesis, arrhythmogenicity, etc.) using immunodeficient mice, rats, pigs, and monkeys. Based on these technologies, we have successfully regenerated human ventricular muscle-specific cardiomyocytes with purity greater than 99%. We have also confirmed that the regenerated myocardium transplanted into immunodeficient mice maintained autonomic beating for more than a year without tumor formation. We are planning to conduct clinical trials to transplant iPS cell-derived cardiomyocytes into patients with heart failure associated with ischemic heart disease, which will, in the near future, enable clinical applications using HLA-deficient iPS cells and iPS cells generated from the patient's own lymphocytes to generate regenerative cardiomyocytes without rejection. It would also help establish personalized medicine for heart failure and usher in the long-awaited treatment for intractable severe heart failure using ventricular muscle supplementation.
Collapse
Affiliation(s)
- Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
42
|
Tanaka Y, Kadota S, Zhao J, Kobayashi H, Okano S, Izumi M, Honda Y, Ichimura H, Shiba N, Uemura T, Wada Y, Chuma S, Nakada T, Tohyama S, Fukuda K, Yamada M, Seto T, Kuwahara K, Shiba Y. Mature human induced pluripotent stem cell-derived cardiomyocytes promote angiogenesis through alpha-B crystallin. Stem Cell Res Ther 2023; 14:240. [PMID: 37679796 PMCID: PMC10486094 DOI: 10.1186/s13287-023-03468-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 08/22/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) can be used to treat heart diseases; however, the optimal maturity of hiPSC-CMs for effective regenerative medicine remains unclear. We aimed to investigate the benefits of long-term cultured mature hiPSC-CMs in injured rat hearts. METHODS Cardiomyocytes were differentiated from hiPSCs via monolayer culturing, and the cells were harvested on day 28 or 56 (D28-CMs or D56-CMs, respectively) after differentiation. We transplanted D28-CMs or D56-CMs into the hearts of rat myocardial infarction models and examined cell retention and engraftment via in vivo bioluminescence imaging and histological analysis. We performed transcriptomic sequencing analysis to elucidate the genetic profiles before and after hiPSC-CM transplantation. RESULTS Upregulated expression of mature sarcomere genes in vitro was observed in D56-CMs compared with D28-CMs. In vivo bioluminescence imaging studies revealed increased bioluminescence intensity of D56-CMs at 8 and 12 weeks post-transplantation. Histological and immunohistochemical analyses showed that D56-CMs promoted engraftment and maturation in the graft area at 12 weeks post-transplantation. Notably, D56-CMs consistently promoted microvessel formation in the graft area from 1 to 12 weeks post-transplantation. Transcriptomic sequencing analysis revealed that compared with the engrafted D28-CMs, the engrafted D56-CMs enriched genes related to blood vessel regulation at 12 weeks post-transplantation. As shown by transcriptomic and western blot analyses, the expression of a small heat shock protein, alpha-B crystallin (CRYAB), was significantly upregulated in D56-CMs compared with D28-CMs. Endothelial cell migration was inhibited by small interfering RNA-mediated knockdown of CRYAB when co-cultured with D56-CMs in vitro. Furthermore, CRYAB overexpression enhanced angiogenesis in the D28-CM grafts at 4 weeks post-transplantation. CONCLUSIONS Long-term cultured mature hiPSC-CMs promoted engraftment, maturation and angiogenesis post-transplantation in infarcted rat hearts. CRYAB, which was highly expressed in D56-CMs, was identified as an angiogenic factor from mature hiPSC-CMs. This study revealed the benefits of long-term culture, which may enhance the therapeutic potential of hiPSC-CMs.
Collapse
Affiliation(s)
- Yuki Tanaka
- Department of Regenerative Science and Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
- Division of Cardiovascular Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | - Shin Kadota
- Department of Regenerative Science and Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan.
- Institute for Biomedical Sciences, Shinshu University, Matsumoto, 390-8621, Japan.
| | - Jian Zhao
- Department of Regenerative Science and Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| | - Hideki Kobayashi
- Department of Regenerative Science and Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
- Department of Cardiovascular Medicine, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | - Satomi Okano
- Department of Regenerative Science and Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
- Department of Physical Therapy, Faculty of Health Sciences, Iryo Sosei University, Iwaki, 970-8551, Japan
| | - Masaki Izumi
- Department of Regenerative Science and Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
- Division of Diabetes, Endocrinology and Metabolism, Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | - Yusuke Honda
- Department of Regenerative Science and Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| | - Hajime Ichimura
- Department of Regenerative Science and Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
- Division of Cardiovascular Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | - Naoko Shiba
- Department of Regenerative Science and Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | - Takeshi Uemura
- Institute for Biomedical Sciences, Shinshu University, Matsumoto, 390-8621, Japan
- Division of Gene Research, Research Center for Advanced Science and Technology, Shinshu University, Matsumoto, 390-8621, Japan
| | - Yuko Wada
- Division of Cardiovascular Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | - Shinichiro Chuma
- Department of Regeneration Science and Engineering, Institute for Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Tsutomu Nakada
- Division of Instrumental Analysis, Research Center for Advanced Science and Technology, Shinshu University, Matsumoto, 390-8621, Japan
| | - Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Mitsuhiko Yamada
- Department of Molecular Pharmacology, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | - Tatsuichiro Seto
- Division of Cardiovascular Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | - Koichiro Kuwahara
- Institute for Biomedical Sciences, Shinshu University, Matsumoto, 390-8621, Japan
- Department of Cardiovascular Medicine, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | - Yuji Shiba
- Department of Regenerative Science and Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan.
- Institute for Biomedical Sciences, Shinshu University, Matsumoto, 390-8621, Japan.
| |
Collapse
|
43
|
Tan Y, Coyle RC, Barrs RW, Silver SE, Li M, Richards DJ, Lin Y, Jiang Y, Wang H, Menick DR, Deleon-Pennell K, Tian B, Mei Y. Nanowired human cardiac organoid transplantation enables highly efficient and effective recovery of infarcted hearts. SCIENCE ADVANCES 2023; 9:eadf2898. [PMID: 37540743 PMCID: PMC10403216 DOI: 10.1126/sciadv.adf2898] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 07/06/2023] [Indexed: 08/06/2023]
Abstract
Human cardiac organoids hold remarkable potential for cardiovascular disease modeling and human pluripotent stem cell-derived cardiomyocyte (hPSC-CM) transplantation. Here, we show cardiac organoids engineered with electrically conductive silicon nanowires (e-SiNWs) significantly enhance the therapeutic efficacy of hPSC-CMs to treat infarcted hearts. We first demonstrated the biocompatibility of e-SiNWs and their capacity to improve cardiac microtissue engraftment in healthy rat myocardium. Nanowired human cardiac organoids were then engineered with hPSC-CMs, nonmyocyte supporting cells, and e-SiNWs. Nonmyocyte supporting cells promoted greater ischemia tolerance of cardiac organoids, and e-SiNWs significantly improved electrical pacing capacity. After transplantation into ischemia/reperfusion-injured rat hearts, nanowired cardiac organoids significantly improved contractile development of engrafted hPSC-CMs, induced potent cardiac functional recovery, and reduced maladaptive left ventricular remodeling. Compared to contemporary studies with an identical injury model, greater functional recovery was achieved with a 20-fold lower dose of hPSC-CMs, revealing therapeutic synergy between conductive nanomaterials and human cardiac organoids for efficient heart repair.
Collapse
Affiliation(s)
- Yu Tan
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Robert C. Coyle
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Ryan W. Barrs
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Sophia E. Silver
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Mei Li
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Dylan J. Richards
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Yiliang Lin
- Department of Chemistry, The James Franck Institute and the Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Yuanwen Jiang
- Department of Chemistry, The James Franck Institute and the Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Hongjun Wang
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Donald R. Menick
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Ralph H. Johnson Veterans Affairs Medical Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Kristine Deleon-Pennell
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Ralph H. Johnson Veterans Affairs Medical Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Bozhi Tian
- Department of Chemistry, The James Franck Institute and the Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
44
|
Zhuo D, Lei I, Li W, Liu L, Li L, Ni J, Liu Z, Fan G. The origin, progress, and application of cell-based cardiac regeneration therapy. J Cell Physiol 2023; 238:1732-1755. [PMID: 37334836 DOI: 10.1002/jcp.31060] [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: 03/17/2023] [Revised: 05/08/2023] [Accepted: 05/29/2023] [Indexed: 06/21/2023]
Abstract
Cardiovascular disease (CVD) has become a severe threat to human health, with morbidity and mortality increasing yearly and gradually becoming younger. When the disease progresses to the middle and late stages, the loss of a large number of cardiomyocytes is irreparable to the body itself, and clinical drug therapy and mechanical support therapy cannot reverse the development of the disease. To explore the source of regenerated myocardium in model animals with the ability of heart regeneration through lineage tracing and other methods, and develop a new alternative therapy for CVDs, namely cell therapy. It directly compensates for cardiomyocyte proliferation through adult stem cell differentiation or cell reprogramming, which indirectly promotes cardiomyocyte proliferation through non-cardiomyocyte paracrine, to play a role in heart repair and regeneration. This review comprehensively summarizes the origin of newly generated cardiomyocytes, the research progress of cardiac regeneration based on cell therapy, the opportunity and development of cardiac regeneration in the context of bioengineering, and the clinical application of cell therapy in ischemic diseases.
Collapse
Affiliation(s)
- Danping Zhuo
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ienglam Lei
- Department of Cardiac Surgery, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Wenjun Li
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Li Liu
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lan Li
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jingyu Ni
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihao Liu
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guanwei Fan
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
45
|
Maas RGC, van den Dolder FW, Yuan Q, van der Velden J, Wu SM, Sluijter JPG, Buikema JW. Harnessing developmental cues for cardiomyocyte production. Development 2023; 150:dev201483. [PMID: 37560977 PMCID: PMC10445742 DOI: 10.1242/dev.201483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2023]
Abstract
Developmental research has attempted to untangle the exact signals that control heart growth and size, with knockout studies in mice identifying pivotal roles for Wnt and Hippo signaling during embryonic and fetal heart growth. Despite this improved understanding, no clinically relevant therapies are yet available to compensate for the loss of functional adult myocardium and the absence of mature cardiomyocyte renewal that underlies cardiomyopathies of multiple origins. It remains of great interest to understand which mechanisms are responsible for the decline in proliferation in adult hearts and to elucidate new strategies for the stimulation of cardiac regeneration. Multiple signaling pathways have been identified that regulate the proliferation of cardiomyocytes in the embryonic heart and appear to be upregulated in postnatal injured hearts. In this Review, we highlight the interaction of signaling pathways in heart development and discuss how this knowledge has been translated into current technologies for cardiomyocyte production.
Collapse
Affiliation(s)
- Renee G. C. Maas
- Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, 3508 GA Utrecht, the Netherlands
| | - Floor W. van den Dolder
- Amsterdam Cardiovascular Sciences, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam University Medical Centers, De Boelelaan 1108, 1081 HZ Amsterdam, the Netherlands
| | - Qianliang Yuan
- Amsterdam Cardiovascular Sciences, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam University Medical Centers, De Boelelaan 1108, 1081 HZ Amsterdam, the Netherlands
| | - Jolanda van der Velden
- Amsterdam Cardiovascular Sciences, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam University Medical Centers, De Boelelaan 1108, 1081 HZ Amsterdam, the Netherlands
| | - Sean M. Wu
- Department of Medicine, Division of Cardiovascular Medicine,Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joost P. G. Sluijter
- Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, 3508 GA Utrecht, the Netherlands
| | - Jan W. Buikema
- Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, 3508 GA Utrecht, the Netherlands
- Amsterdam Cardiovascular Sciences, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam University Medical Centers, De Boelelaan 1108, 1081 HZ Amsterdam, the Netherlands
- Department of Cardiology, Amsterdam Heart Center, Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
46
|
Chen Y, Wu Y, Li J, Chen K, Wang W, Ye Z, Feng K, Yang Y, Xu Y, Kang J, Guo X. Cooperative regulation of Zhx1 and hnRNPA1 drives the cardiac progenitor-specific transcriptional activation during cardiomyocyte differentiation. Cell Death Discov 2023; 9:244. [PMID: 37452012 PMCID: PMC10349095 DOI: 10.1038/s41420-023-01548-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 06/22/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023] Open
Abstract
The zinc finger proteins (ZNFs) mediated transcriptional regulation is critical for cell fate transition. However, it is still unclear how the ZNFs realize their specific regulatory roles in the stage-specific determination of cardiomyocyte differentiation. Here, we reported that the zinc fingers and homeoboxes 1 (Zhx1) protein, transiently expressed during the cell fate transition from mesoderm to cardiac progenitors, was indispensable for the proper cardiomyocyte differentiation of mouse and human embryonic stem cells. Moreover, Zhx1 majorly promoted the specification of cardiac progenitors via interacting with hnRNPA1 and co-activated the transcription of a wide range of genes. In-depth mechanistic studies showed that Zhx1 was bound with hnRNPA1 by the amino acid residues (Thr111-His120) of the second Znf domain, thus participating in the formation of cardiac progenitors. Together, our study highlights the unrevealed interaction of Zhx1/hnRNPA1 for activating gene transcription during cardiac progenitor specification and also provides new evidence for the specificity of cell fate determination in cardiomyocyte differentiation.
Collapse
Affiliation(s)
- Yang Chen
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yukang Wu
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jianguo Li
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Kai Chen
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Wuchan Wang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Zihui Ye
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Ke Feng
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yiwei Yang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yanxin Xu
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jiuhong Kang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Xudong Guo
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
- Institute for Advanced Study, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
47
|
Collet BC, Davis DR. Mechanisms of Cardiac Repair in Cell Therapy. Heart Lung Circ 2023; 32:825-835. [PMID: 37031061 DOI: 10.1016/j.hlc.2023.01.019] [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: 07/29/2022] [Revised: 12/10/2022] [Accepted: 01/04/2023] [Indexed: 04/08/2023]
Abstract
Heart failure is an important cause of morbidity and mortality. More than 20 years ago, special interest was drawn to cell therapy as a means of restoring damaged hearts to working condition. But progress has not been straightforward as many of our initial assumptions turned out to be wrong. In this review, we critically examine the last 20 years of progress in cardiac cell therapy and focus on several of the popular beliefs surrounding cell therapy to illustrate the mechanisms involved in restoring heart function after cardiac injury. Are they true or false?
Collapse
Affiliation(s)
- Bérénice C Collet
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Darryl R Davis
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
48
|
Lin YN, Miguel-Dos-Santos R, Cingolani E. Biological Modification of Arrhythmogenic Substrates by Cell-Free Therapeutics. Heart Lung Circ 2023; 32:844-851. [PMID: 37353457 PMCID: PMC10526725 DOI: 10.1016/j.hlc.2023.05.016] [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: 11/24/2022] [Revised: 03/21/2023] [Accepted: 05/02/2023] [Indexed: 06/25/2023]
Abstract
Ventricular arrhythmias (VAs) represent a major cause of sudden cardiac death and afflict patients with heart failure from both ischaemic and non-ischaemic origins, and inherited cardiomyopathies. Current VA management, including anti-arrhythmic medications, autonomic modulation, implantable cardioverter-defibrillator implantation, and catheter ablation, remains suboptimal. Catheter ablation may even cause significant cardiomyocyte loss. Cell-based therapies and exosome treatment have been proposed as promising strategies to lessen cardiomyocyte death, modulate immune reaction, and reduce myocardial scarring, and, therefore, are potentially beneficial in treating VAs. In this review, we summarise the current cornerstones of VA management. We also discuss recent advances and ongoing evidence regarding cell-based and exosome therapy, with special attention to VA treatment.
Collapse
Affiliation(s)
- Yen-Nien Lin
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Cardiovascular Medicine, Department of Medicine, China Medical University and Hospital, Taipei, Taiwan
| | | | - Eugenio Cingolani
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
49
|
Chi C, Song K. Cellular reprogramming of fibroblasts in heart regeneration. J Mol Cell Cardiol 2023; 180:84-93. [PMID: 36965699 PMCID: PMC10347886 DOI: 10.1016/j.yjmcc.2023.03.009] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/10/2023] [Accepted: 03/21/2023] [Indexed: 03/27/2023]
Abstract
Myocardial infarction causes the loss of cardiomyocytes and the formation of cardiac fibrosis due to the activation of cardiac fibroblasts, leading to cardiac dysfunction and heart failure. Unfortunately, current therapeutic interventions can only slow the disease progression. Furthermore, they cannot fully restore cardiac function, likely because the adult human heart lacks sufficient capacity to regenerate cardiomyocytes. Therefore, intensive efforts have focused on developing therapeutics to regenerate the damaged heart. Several strategies have been intensively investigated, including stimulation of cardiomyocyte proliferation, transplantation of stem cell-derived cardiomyocytes, and conversion of fibroblasts into cardiac cells. Resident cardiac fibroblasts are critical in the maintenance of the structure and contractility of the heart. Fibroblast plasticity makes this type of cells be reprogrammed into many cell types, including but not limited to induced pluripotent stem cells, induced cardiac progenitor cells, and induced cardiomyocytes. Fibroblasts have become a therapeutic target due to their critical roles in cardiac pathogenesis. This review summarizes the reprogramming of fibroblasts into induced pluripotent stem cell-derived cardiomyocytes, induced cardiac progenitor cells, and induced cardiomyocytes to repair a damaged heart, outlines recent findings in utilizing fibroblast-derived cells for heart regeneration, and discusses the limitations and challenges.
Collapse
Affiliation(s)
- Congwu Chi
- Division of Cardiology, Department of Medicine, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kunhua Song
- Division of Cardiology, Department of Medicine, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Gates Center for Regenerative Medicine and Stem Cell Biology, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
50
|
Reyes Gaido OE, Anderson ME. CRISPR Editing Takes Aim at Ischemia/Reperfusion Injury. JAMA Cardiol 2023; 8:522-523. [PMID: 37163250 PMCID: PMC11019924 DOI: 10.1001/jamacardio.2023.0983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Affiliation(s)
- Oscar E Reyes Gaido
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mark E Anderson
- Division of Biological Sciences and the Pritzker School of Medicine, The University of Chicago, Chicago, Illinois
| |
Collapse
|