1
|
Bisson JA, Gordillo M, Kumar R, de Silva N, Yang E, Banks KM, Shi ZD, Lee K, Yang D, Chung WK, Huangfu D, Evans T. GATA6 regulates WNT and BMP programs to pattern precardiac mesoderm during the earliest stages of human cardiogenesis. eLife 2025; 13:RP100797. [PMID: 40080060 PMCID: PMC11906159 DOI: 10.7554/elife.100797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2025] Open
Abstract
Haploinsufficiency for GATA6 is associated with congenital heart disease (CHD) with variable comorbidity of pancreatic or diaphragm defects, although the etiology of disease is not well understood. Here, we used cardiac directed differentiation from human embryonic stem cells (hESCs) as a platform to study GATA6 function during early cardiogenesis. GATA6 loss-of-function hESCs had a profound impairment in cardiac progenitor cell (CPC) specification and cardiomyocyte (CM) generation due to early defects during the mesendoderm and lateral mesoderm patterning stages. Profiling by RNA-seq and CUT&RUN identified genes of the WNT and BMP programs regulated by GATA6 during early mesoderm patterning. Furthermore, interactome analysis detected GATA6 binding with developmental transcription factors and chromatin remodelers, suggesting cooperative regulation of cardiac lineage gene accessibility. We show that modulating WNT and BMP inputs during the first 48 hr of cardiac differentiation is sufficient to partially rescue CPC and CM defects in GATA6 heterozygous and homozygous mutant hESCs. This study provides evidence of the regulatory functions for GATA6 directing human precardiac mesoderm patterning during the earliest stages of cardiogenesis to further our understanding of haploinsufficiency causing CHD and the co-occurrence of cardiac and other organ defects caused by human GATA6 mutations.
Collapse
Affiliation(s)
- Joseph A Bisson
- Department of Surgery, Weill Cornell MedicineNew YorkUnited States
| | - Miriam Gordillo
- Department of Surgery, Weill Cornell MedicineNew YorkUnited States
| | - Ritu Kumar
- Department of Surgery, Weill Cornell MedicineNew YorkUnited States
| | | | - Ellen Yang
- Department of Surgery, Weill Cornell MedicineNew YorkUnited States
| | - Kelly M Banks
- Department of Surgery, Weill Cornell MedicineNew YorkUnited States
| | - Zhong-Dong Shi
- Developmental Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Kihyun Lee
- Developmental Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Dapeng Yang
- Developmental Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Wendy K Chung
- Childrens Hospital, Harvard Medical SchoolBostonUnited States
| | - Danwei Huangfu
- Developmental Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Todd Evans
- Department of Surgery, Weill Cornell MedicineNew YorkUnited States
- Hartman Institute for Therapeutic Organ Regeneration, Weill Cornell MedicineNew YorkUnited States
- Center for Genomic Health, Weill Cornell MedicineNew YorkUnited States
| |
Collapse
|
2
|
Urasawa T, Kawasaki N. Proteomic Approach Using DIA-MS Identifies Morphogenesis-Associated Proteins during Cardiac Differentiation of Human iPS Cells. ACS OMEGA 2025; 10:344-357. [PMID: 39829588 PMCID: PMC11740111 DOI: 10.1021/acsomega.4c06371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025]
Abstract
Human-induced pluripotent stem cell (hiPSC)-derived cardiomyocytes have potential applications in regenerative medicine. The quality by design (QbD) approach enables the efficiency and quality assurance in the manufacturing of hiPSC-derived products. It requires a molecular understanding of hiPSC differentiation throughout the differentiation process; however, information on cardiac differentiation remains limited. Proteins associated with the early stages of cardiac differentiation would be useful in the cardiomyocyte quality assessment. Here, we performed quantitative proteomics of hiPSC intermediate cells in the early phase of cardiac differentiation to better understand their molecular characteristics. Proteomic profiles suggested that day 5-7 cells were in the morphogenetic stage of cardiac differentiation. Trophoblast glycoprotein (TPBG) was the most up-regulated protein in the morphogenetic stage; it was previously shown to be up-regulated during differentiation into neural stem cells. Proteomics of TPBG-knockdown cells revealed that TPBG is involved in cell proliferation and is related to the cardiomyocyte yield, suggesting that it could be used as a marker in QbD development. Our approach helps us understand the molecular basis of hiPSC differentiation and could be a powerful tool in QbD-based manufacturing.
Collapse
Affiliation(s)
- Takaya Urasawa
- Biopharmaceutical and Regenerative
Sciences, Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Nana Kawasaki
- Biopharmaceutical and Regenerative
Sciences, Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| |
Collapse
|
3
|
Bisson JA, Gordillo M, Kumar R, de Silva N, Yang E, Banks KM, Shi ZD, Lee K, Yang D, Chung WK, Huangfu D, Evans T. GATA6 regulates WNT and BMP programs to pattern precardiac mesoderm during the earliest stages of human cardiogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.09.602666. [PMID: 39026742 PMCID: PMC11257636 DOI: 10.1101/2024.07.09.602666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Haploinsufficiency for GATA6 is associated with congenital heart disease (CHD) with variable comorbidity of pancreatic or diaphragm defects, although the etiology of disease is not well understood. Here, we used cardiac directed differentiation from human embryonic stem cells (hESCs) as a platform to study GATA6 function during early cardiogenesis. GATA6 loss-of-function hESCs had a profound impairment in cardiac progenitor cell (CPC) specification and cardiomyocyte (CM) generation due to early defects during the mesendoderm and lateral mesoderm patterning stages. Profiling by RNA-seq and CUT&RUN identified genes of the WNT and BMP programs regulated by GATA6 during early mesoderm patterning. Furthermore, interactome analysis detected GATA6 binding with developmental transcription factors and chromatin remodelers suggesting cooperative regulation of cardiac lineage gene accessibility. We show that modulating WNT and BMP inputs during the first 48 hours of cardiac differentiation is sufficient to partially rescue CPC and CM defects in GATA6 heterozygous and homozygous mutant hESCs. This study provides evidence of the regulatory functions for GATA6 directing human precardiac mesoderm patterning during the earliest stages of cardiogenesis to further our understanding of haploinsufficiency causing CHD and the co-occurrence of cardiac and other organ defects caused by human GATA6 mutations.
Collapse
Affiliation(s)
- Joseph A. Bisson
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Miriam Gordillo
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Ritu Kumar
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
- current address: Gladstone Institutes, San Francisco, CA, 94158, USA
| | - Neranjan de Silva
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Ellen Yang
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Kelly M. Banks
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Zhong-Dong Shi
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, 10065, USA
| | - Kihyun Lee
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, 10065, USA
- current address: College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Dapeng Yang
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, 10065, USA
| | - Wendy K. Chung
- Childrens Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Danwei Huangfu
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, 10065, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
- Hartman Institute for Therapeutic Organ Regeneration, Weill Cornell Medicine, New York, NY 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
4
|
Minakawa T, Yamashita JK. Extracellular vesicles and microRNAs in the regulation of cardiomyocyte differentiation and proliferation. Arch Biochem Biophys 2023; 749:109791. [PMID: 37858665 DOI: 10.1016/j.abb.2023.109791] [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: 08/31/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 10/21/2023]
Abstract
Cardiomyocyte differentiation and proliferation are essential processes for the regeneration of an injured heart. In recent years, there have been several reports highlighting the involvement of extracellular vesicles (EVs) in cardiomyocyte differentiation and proliferation. These EVs originate from mesenchymal stem cells, pluripotent stem cells, and heart constituting cells (cardiomyocytes, cardiac fibroblasts, cardiac progenitor cells, epicardium). Numerous reports also indicate the involvement of microRNAs (miRNAs) in cardiomyocyte differentiation and proliferation. Among them, miRNA-1, miRNA-133, and miRNA-499, recently demonstrated to promote cardiomyocyte differentiation, and miRNA-199, shown to promote cardiomyocyte proliferation, were found effective in various studies. MiRNA-132 and miRNA-133 have been identified as cargo in EVs and are reported to induce cardiomyocyte differentiation. Similarly, miRNA-30a, miRNA-100, miRNA-27a, miRNA-30e, miRNA-294 and miRNA-590 have also been identified as cargo in EVs and are shown to have a role in the promotion of cardiomyocyte proliferation. Regeneration of the heart by EVs or artificial nanoparticles containing functional miRNAs is expected in the future. In this review, we outline recent advancements in understanding the roles of EVs and miRNAs in cardiomyocyte differentiation and proliferation. Additionally, we explore the related challenges when utilizing EVs and miRNAs as a less risky approach to cardiac regeneration compared to cell transplantation.
Collapse
Affiliation(s)
- Tomohiro Minakawa
- Department of Cellular and Tissue Communication, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Jun K Yamashita
- Department of Cellular and Tissue Communication, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan.
| |
Collapse
|
5
|
Bachamanda Somesh D, Klose K, Maring JA, Kunkel D, Jürchott K, Protze SI, Klein O, Nebrich G, Becker M, Krüger U, Nazari-Shafti TZ, Falk V, Kurtz A, Gossen M, Stamm C. Cardiomyocyte precursors generated by direct reprogramming and molecular beacon selection attenuate ventricular remodeling after experimental myocardial infarction. Stem Cell Res Ther 2023; 14:296. [PMID: 37840130 PMCID: PMC10577947 DOI: 10.1186/s13287-023-03519-w] [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: 03/24/2022] [Accepted: 09/25/2023] [Indexed: 10/17/2023] Open
Abstract
BACKGROUND Direct cardiac reprogramming is currently being investigated for the generation of cells with a true cardiomyocyte (CM) phenotype. Based on the original approach of cardiac transcription factor-induced reprogramming of fibroblasts into CM-like cells, various modifications of that strategy have been developed. However, they uniformly suffer from poor reprogramming efficacy and a lack of translational tools for target cell expansion and purification. Therefore, our group has developed a unique approach to generate proliferative cells with a pre-CM phenotype that can be expanded in vitro to yield substantial cell doses. METHODS Cardiac fibroblasts were reprogrammed toward CM fate using lentiviral transduction of cardiac transcriptions factors (GATA4, MEF2C, TBX5, and MYOCD). The resulting cellular phenotype was analyzed by RNA sequencing and immunocytology. Live target cells were purified based on intracellular CM marker expression using molecular beacon technology and fluorescence-activated cell sorting. CM commitment was assessed using 5-azacytidine-based differentiation assays and the therapeutic effect was evaluated in a mouse model of acute myocardial infarction using echocardiography and histology. The cellular secretome was analyzed using mass spectrometry. RESULTS We found that proliferative CM precursor-like cells were part of the phenotype spectrum arising during direct reprogramming of fibroblasts toward CMs. These induced CM precursors (iCMPs) expressed CPC- and CM-specific proteins and were selectable via hairpin-shaped oligonucleotide hybridization probes targeting Myh6/7-mRNA-expressing cells. After purification, iCMPs were capable of extensive expansion, with preserved phenotype when under ascorbic acid supplementation, and gave rise to CM-like cells with organized sarcomeres in differentiation assays. When transplanted into infarcted mouse hearts, iCMPs prevented CM loss, attenuated fibrotic scarring, and preserved ventricular function, which can in part be attributed to their substantial secretion of factors with documented beneficial effect on cardiac repair. CONCLUSIONS Fibroblast reprogramming combined with molecular beacon-based cell selection yields an iCMP-like cell population with cardioprotective potential. Further studies are needed to elucidate mechanism-of-action and translational potential.
Collapse
Affiliation(s)
- Dipthi Bachamanda Somesh
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany.
- Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany.
| | - Kristin Klose
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany.
- Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany.
| | - Janita A Maring
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, 14513, Teltow, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, 13353, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Désirée Kunkel
- Cytometry Core Facility, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Karsten Jürchott
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Institute for Medical Immunology, 13353, Berlin, Germany
| | - Stephanie I Protze
- University Health Network, McEwen Stem Cell Institute, Toronto, ON, M5G 1L7, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Oliver Klein
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- BIH Imaging Mass Spectrometry Core Unit, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Grit Nebrich
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- BIH Imaging Mass Spectrometry Core Unit, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Matthias Becker
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Ulrike Krüger
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Institute for Medical Immunology, 13353, Berlin, Germany
| | - Timo Z Nazari-Shafti
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- German Centre for Cardiovascular Research, Partner Site Berlin, 10785, Berlin, Germany
| | - Volkmar Falk
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- German Centre for Cardiovascular Research, Partner Site Berlin, 10785, Berlin, Germany
- Department of Health Sciences and Technology, ETH Zurich, 8092, Zurich, Switzerland
| | - Andreas Kurtz
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Manfred Gossen
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, 14513, Teltow, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, 13353, Berlin, Germany
| | - Christof Stamm
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany.
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, 14513, Teltow, Germany.
- Berlin-Brandenburg Center for Regenerative Therapies, 13353, Berlin, Germany.
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
- German Centre for Cardiovascular Research, Partner Site Berlin, 10785, Berlin, Germany.
| |
Collapse
|
6
|
Takahi M, Taira R, Onozuka J, Sunamura H, Kondow A, Nakade K, Nakashima K, Sato I, Hayashi Y, Patra C, Ohnuma K. Xenograft of human pluripotent stem cell-derived cardiac lineage cells on zebrafish embryo heart. Biochem Biophys Res Commun 2023; 674:190-198. [PMID: 37532637 DOI: 10.1016/j.bbrc.2023.06.040] [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: 05/13/2023] [Accepted: 06/13/2023] [Indexed: 08/04/2023]
Abstract
Cardiomyocytes derived from human induced pluripotent stem cells (hiPSCs) are a promising cell source for regenerative medicine and drug discovery. However, the use of animal models for studying human cardiomyocytes derived from hiPSCs in vivo is limited and challenging. Given the shared properties between humans and zebrafish, their ethical advantages over mammalian models, and their immature immune system that is rejection-free against xenografted human cells, zebrafish provide a suitable alternative model for xenograft studies. We microinjected fluorescence-labeled cardiac lineage cells derived from hiPSCs, specifically mesoderm or cardiac mesoderm cells, into the yolk and the area proximal to the outflow tract of the linear heart at 24 hours post-fertilization (hpf). The cells injected into the yolk survived and did not migrate to other tissues. In contrast, the cells injected contiguous with the outflow tract of the linear heart migrated into the pericardial cavity and heart. After 1 day post injection (1 dpi, 22-24 hpi), the injected cells migrated into the pericardial cavity and heart. Importantly, we observed heartbeat-like movements of some injected cells in the zebrafish heart after 1 dpi. These results suggested successful xenografting of hiPSC-derived cardiac lineage cells into the zebrafish embryo heart. Thus, we developed a valuable tool using zebrafish embryos as a model organism for investigating the molecular and cellular mechanisms involved in the grafting process. This is essential in developing cell transplantation-based cardiac therapeutics as well as for drug testing, notably contributing to advancements in the field of cardio-medicine.
Collapse
Affiliation(s)
- Mika Takahi
- 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.
| | - Jo Onozuka
- Department of Materials Science and Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan.
| | - Haruka Sunamura
- Department of Materials Science and Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan.
| | - Akiko Kondow
- Advanced Comprehensive Research Organization, Teikyo University, 2-21-1 Kaga, Itabashi-ku, Tokyo, 173-0003, Japan.
| | - Koji Nakade
- Gene Engineering Division, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan.
| | - Kenichi Nakashima
- Gene Engineering Division, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan.
| | - Iori Sato
- iPS Cell Advanced Characterization and Development Team, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan.
| | - Yohei Hayashi
- iPS Cell Advanced Characterization and Development Team, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, 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
|
7
|
Yang X, Chen D, Sun Q, Wang Y, Xia Y, Yang J, Lin C, Dang X, Cen Z, Liang D, Wei R, Xu Z, Xi G, Xue G, Ye C, Wang LP, Zou P, Wang SQ, Rivera-Fuentes P, Püntener S, Chen Z, Liu Y, Zhang J, Zhao Y. A live-cell image-based machine learning strategy for reducing variability in PSC differentiation systems. Cell Discov 2023; 9:53. [PMID: 37280224 DOI: 10.1038/s41421-023-00543-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 03/13/2023] [Indexed: 06/08/2023] Open
Abstract
The differentiation of pluripotent stem cells (PSCs) into diverse functional cell types provides a promising solution to support drug discovery, disease modeling, and regenerative medicine. However, functional cell differentiation is currently limited by the substantial line-to-line and batch-to-batch variabilities, which severely impede the progress of scientific research and the manufacturing of cell products. For instance, PSC-to-cardiomyocyte (CM) differentiation is vulnerable to inappropriate doses of CHIR99021 (CHIR) that are applied in the initial stage of mesoderm differentiation. Here, by harnessing live-cell bright-field imaging and machine learning (ML), we realize real-time cell recognition in the entire differentiation process, e.g., CMs, cardiac progenitor cells (CPCs), PSC clones, and even misdifferentiated cells. This enables non-invasive prediction of differentiation efficiency, purification of ML-recognized CMs and CPCs for reducing cell contamination, early assessment of the CHIR dose for correcting the misdifferentiation trajectory, and evaluation of initial PSC colonies for controlling the start point of differentiation, all of which provide a more invulnerable differentiation method with resistance to variability. Moreover, with the established ML models as a readout for the chemical screen, we identify a CDK8 inhibitor that can further improve the cell resistance to the overdose of CHIR. Together, this study indicates that artificial intelligence is able to guide and iteratively optimize PSC differentiation to achieve consistently high efficiency across cell lines and batches, providing a better understanding and rational modulation of the differentiation process for functional cell manufacturing in biomedical applications.
Collapse
Affiliation(s)
- Xiaochun Yang
- State Key Laboratory of Natural and Biomimetic Drugs, MOE Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Daichao Chen
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Qiushi Sun
- Beijing Key Lab of Traffic Data Analysis and Mining, School of Computer and Information Technology, Beijing Jiaotong University, Beijing, China
| | - Yao Wang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yu Xia
- College of Engineering, Peking University, Beijing, China
| | - Jinyu Yang
- College of Engineering, Peking University, Beijing, China
| | - Chang Lin
- College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, China
| | - Xin Dang
- State Key Laboratory of Natural and Biomimetic Drugs, MOE Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Zimu Cen
- State Key Laboratory of Natural and Biomimetic Drugs, MOE Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Dongdong Liang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Rong Wei
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Ze Xu
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing, China
| | - Guangyin Xi
- State Key Laboratory of Natural and Biomimetic Drugs, MOE Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Gang Xue
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Can Ye
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Li-Peng Wang
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing, China
| | - Peng Zou
- College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Shi-Qiang Wang
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing, China
| | | | - Salome Püntener
- Department of Chemistry, University of Zurich, Zurich, Switzerland
- Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédéral de Lausanne, Lausanne, Switzerland
| | - Zhixing Chen
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- Institute of Molecular Medicine, National Biomedical Imaging Center, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Yi Liu
- Beijing Key Lab of Traffic Data Analysis and Mining, School of Computer and Information Technology, Beijing Jiaotong University, Beijing, China.
| | - Jue Zhang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
- College of Engineering, Peking University, Beijing, China.
| | - Yang Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, MOE Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
| |
Collapse
|
8
|
Urasawa T, Koizumi T, Kimura K, Ohta Y, Kawasaki N. Quantitative Proteomics for the Development and Manufacturing of Human-Induced Pluripotent Stem Cell-Derived Neural Stem Cells Using Data-Independent Acquisition Mass Spectrometry. J Proteome Res 2023. [PMID: 37097202 DOI: 10.1021/acs.jproteome.2c00841] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
Human-induced pluripotent stem cell (iPSC)-derived neural stem cells (NSCs) have several potential applications in regenerative medicine. A deep understanding of stem cell characteristics is critical for developing appropriate products for use in the clinic. This study aimed to develop approaches for characterizing iPSC-derived NSCs. Data-independent acquisition mass spectrometry (DIA-MS) was used to obtain temporal proteomic profiles of differentiating cells. Principal component analysis of the proteome profiles allowed for the discrimination of cells cultured for different periods. Cells were characterized by Gene Ontology analysis to annotate the upregulated proteins based on their functions. We found that trophoblast glycoprotein (TPBG), a membrane glycoprotein that inhibits the Wnt/β-catenin pathway, was elevated in NSC and that silencing TPBG promoted proliferation rather than neuronal differentiation. Treatment with Wnt/β-catenin pathway activators and inhibitors showed that modulating the Wnt/β-catenin pathway is crucial for differentiation into NSC. These results suggest that the level of TPBG is critical for differentiation into NSC, and TPBG is a potentially critical quality attribute of differentiating cells. In summary, DIA-MS-based proteomics is a promising multi-attribute method for characterizing stem cell-derived products.
Collapse
Affiliation(s)
- Takaya Urasawa
- Biopharmaceutical and Regenerative Sciences, Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045 Japan
| | - Takumi Koizumi
- Biopharmaceutical and Regenerative Sciences, Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045 Japan
| | - Kazumasa Kimura
- Biopharmaceutical and Regenerative Sciences, Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045 Japan
| | - Yuki Ohta
- Biopharmaceutical and Regenerative Sciences, Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045 Japan
| | - Nana Kawasaki
- Biopharmaceutical and Regenerative Sciences, Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045 Japan
| |
Collapse
|
9
|
Small Extracellular Vesicles Derived from Induced Pluripotent Stem Cells in the Treatment of Myocardial Injury. Int J Mol Sci 2023; 24:ijms24054577. [PMID: 36902008 PMCID: PMC10003569 DOI: 10.3390/ijms24054577] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 03/02/2023] Open
Abstract
Induced pluripotent stem cell (iPSC) therapy brings great hope to the treatment of myocardial injuries, while extracellular vesicles may be one of the main mechanisms of its action. iPSC-derived small extracellular vesicles (iPSCs-sEVs) can carry genetic and proteinaceous substances and mediate the interaction between iPSCs and target cells. In recent years, more and more studies have focused on the therapeutic effect of iPSCs-sEVs in myocardial injury. IPSCs-sEVs may be a new cell-free-based treatment for myocardial injury, including myocardial infarction, myocardial ischemia-reperfusion injury, coronary heart disease, and heart failure. In the current research on myocardial injury, the extraction of sEVs from mesenchymal stem cells induced by iPSCs was widely used. Isolation methods of iPSCs-sEVs for the treatment of myocardial injury include ultracentrifugation, isodensity gradient centrifugation, and size exclusion chromatography. Tail vein injection and intraductal administration are the most widely used routes of iPSCs-sEV administration. The characteristics of sEVs derived from iPSCs which were induced from different species and organs, including fibroblasts and bone marrow, were further compared. In addition, the beneficial genes of iPSC can be regulated through CRISPR/Cas9 to change the composition of sEVs and improve the abundance and expression diversity of them. This review focused on the strategies and mechanisms of iPSCs-sEVs in the treatment of myocardial injury, which provides a reference for future research and the application of iPSCs-sEVs.
Collapse
|
10
|
Cho SW, Kim HK, Sung JH, Kim Y, Kim JH, Han J. Mitochondrial energy metabolic transcriptome profiles during cardiac differentiation from mouse and human pluripotent stem cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY 2022; 26:357-365. [PMID: 36039736 PMCID: PMC9437366 DOI: 10.4196/kjpp.2022.26.5.357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/13/2022] [Accepted: 07/21/2022] [Indexed: 11/15/2022]
Abstract
Simultaneous myofibril and mitochondrial development is crucial for the cardiac differentiation of pluripotent stem cells (PSCs). Specifically, mitochondrial energy metabolism (MEM) development in cardiomyocytes is essential for the beating function. Although previous studies have reported that MEM is correlated with cardiac differentiation, the process and timing of MEM regulation for cardiac differentiation remain poorly understood. Here, we performed transcriptome analysis of cells at specific stages of cardiac differentiation from mouse embryonic stem cells (mESCs) and human induced PSCs (hiPSCs). We selected MEM genes strongly upregulated at cardiac lineage commitment and in a time-dependent manner during cardiac maturation and identified the protein-protein interaction networks. Notably, MEM proteins were found to interact closely with cardiac maturation-related proteins rather than with cardiac lineage commitment-related proteins. Furthermore, MEM proteins were found to primarily interact with cardiac muscle contractile proteins rather than with cardiac transcription factors. We identified several candidate MEM regulatory genes involved in cardiac lineage commitment (Cck, Bdnf, Fabp4, Cebpα, and Cdkn2a in mESC-derived cells, and CCK and NOS3 in hiPSC-derived cells) and cardiac maturation (Ppargc1α, Pgam2, Cox6a2, and Fabp3 in mESC-derived cells, and PGAM2 and SLC25A4 in hiPSC-derived cells). Therefore, our findings show the importance of MEM in cardiac maturation.
Collapse
Affiliation(s)
- Sung Woo Cho
- Division of Cardiology, Department of Internal Medicine, Inje University College of Medicine, Ilsan Paik Hospital, Cardiac & Vascular Center, Goyang 10380, Korea
- Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutics Center, Inje University College of Medicine, Busan 47392, Korea
| | - Hyoung Kyu Kim
- Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutics Center, Inje University College of Medicine, Busan 47392, Korea
- Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, Inje University College of Medicine, Busan 47392, Korea
| | - Ji Hee Sung
- Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutics Center, Inje University College of Medicine, Busan 47392, Korea
- Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, Inje University College of Medicine, Busan 47392, Korea
| | - Yeseul Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Jae Ho Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea
| | - Jin Han
- Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutics Center, Inje University College of Medicine, Busan 47392, Korea
- Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, Inje University College of Medicine, Busan 47392, Korea
| |
Collapse
|
11
|
Cho SW, Kim HK, Sung JH, Han J. Stage specific transcriptome profiles at cardiac lineage commitment during cardiomyocyte differentiation from mouse and human pluripotent stem cells. BMB Rep 2021. [PMID: 34120677 PMCID: PMC8505231 DOI: 10.5483/bmbrep.2021.54.9.046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cardiomyocyte differentiation occurs through complex and finely regulated processes including cardiac lineage commitment and maturation from pluripotent stem cells (PSCs). To gain some insight into the genome-wide characteristics of cardiac lineage commitment, we performed transcriptome analysis on both mouse embryonic stem cells (mESCs) and human induced PSCs (hiPSCs) at specific stages of cardiomyocyte differentiation. Specifically, the gene expression profiles and the protein–protein interaction networks of the mESC-derived platelet-derived growth factor receptor-alpha (PDGFRα)+ cardiac lineage-committed cells (CLCs) and hiPSC-derived kinase insert domain receptor (KDR)+ and PDGFRα+ cardiac progenitor cells (CPCs) at cardiac lineage commitment were compared with those of mesodermal cells and differentiated cardiomyocytes. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses revealed that the genes significantly upregulated at cardiac lineage commitment were associated with responses to organic substances and external stimuli, extracellular and myocardial contractile components, receptor binding, gated channel activity, PI3K‑AKT signaling, and cardiac hypertrophy and dilation pathways. Protein–protein interaction network analysis revealed that the expression levels of genes that regulate cardiac maturation, heart contraction, and calcium handling showed a consistent increase during cardiac differentiation; however, the expression levels of genes that regulate cell differentiation and multicellular organism development decreased at the cardiac maturation stage following lineage commitment. Additionally, we identified for the first time the protein–protein interaction network connecting cardiac development, the immune system, and metabolism during cardiac lineage commitment in both mESC-derived PDGFRα+ CLCs and hiPSC-derived KDR+PDGFRα+ CPCs. These findings shed light on the regulation of cardiac lineage commitment and the pathogenesis of cardiometabolic diseases.
Collapse
Affiliation(s)
- Sung Woo Cho
- Division of Cardiology, Department of Internal Medicine, Inje University College of Medicine, Ilsan Paik Hospital Vision 21 Cardiac & Vascular Center, Goyang 10380, Korea
- Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutics Center, Inje University College of Medicine, Busan 47392, Korea
| | - Hyoung Kyu Kim
- Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutics Center, Inje University College of Medicine, Busan 47392, Korea
- Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, Inje University College of Medicine, Busan 47392, Korea
| | - Ji Hee Sung
- Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutics Center, Inje University College of Medicine, Busan 47392, Korea
- Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, Inje University College of Medicine, Busan 47392, Korea
| | - Jin Han
- Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutics Center, Inje University College of Medicine, Busan 47392, Korea
- Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, Inje University College of Medicine, Busan 47392, Korea
| |
Collapse
|
12
|
Sun G, Chen J, Ding Y, Wren JD, Xu F, Lu L, Wang Y, Wang DW, Zhang XA. A Bioinformatics Perspective on the Links Between Tetraspanin-Enriched Microdomains and Cardiovascular Pathophysiology. Front Cardiovasc Med 2021; 8:630471. [PMID: 33860000 PMCID: PMC8042132 DOI: 10.3389/fcvm.2021.630471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/15/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Tetraspanins and integrins are integral membrane proteins. Tetraspanins interact with integrins to modulate the dynamics of adhesion, migration, proliferation, and signaling in the form of membrane domains called tetraspanin-enriched microdomains (TEMs). TEMs also contain other cell adhesion proteins like immunoglobulin superfamily (IgSF) proteins and claudins. Cardiovascular functions of these TEM proteins have emerged and remain to be further revealed. Objectives: The aims of this study are to explore the roles of these TEM proteins in the cardiovascular system using bioinformatics tools and databases and to highlight the TEM proteins that may functionally associate with cardiovascular physiology and pathology. Methods: For human samples, three databases-GTEx, NCBI-dbGaP, and NCBI-GEO-were used for the analyses. The dbGaP database was used for GWAS analysis to determine the association between target genes and human phenotypes. GEO is an NCBI public repository that archives genomics data. GTEx was used for the analyses of tissue-specific mRNA expression levels and eQTL. For murine samples, GeneNetwork was used to find gene-phenotype correlations and gene-gene correlations of expression levels in mice. The analysis of cardiovascular data was the focus of this study. Results: Some integrins and tetraspanins, such as ITGA8 and Cd151, are highly expressed in the human cardiovascular system. TEM components are associated with multiple cardiovascular pathophysiological events in humans. GWAS and GEO analyses showed that human Cd82 and ITGA9 are associated with blood pressure. Data from mice also suggest that various cardiovascular phenotypes are correlated with integrins and tetraspanins. For instance, Cd82 and ITGA9, again, have correlations with blood pressure in mice. Conclusion: ITGA9 is related to blood pressure in both species. KEGG analysis also linked ITGA9 to metabolism and MAPK signaling pathway. This work provides an example of using integrated bioinformatics approaches across different species to identify the connections of structurally and/or functionally related molecules to certain categories of diseases.
Collapse
Affiliation(s)
- Ge Sun
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Junxiong Chen
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Yingjun Ding
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Jonathan D. Wren
- Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Fuyi Xu
- University of Tennessee Health Science Center, Memphis, TN, United States
| | - Lu Lu
- University of Tennessee Health Science Center, Memphis, TN, United States
| | - Yan Wang
- Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Dao-wen Wang
- Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Xin A. Zhang
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
13
|
Wang AYL. Human Induced Pluripotent Stem Cell-Derived Exosomes as a New Therapeutic Strategy for Various Diseases. Int J Mol Sci 2021; 22:1769. [PMID: 33578948 PMCID: PMC7916646 DOI: 10.3390/ijms22041769] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/01/2021] [Accepted: 02/07/2021] [Indexed: 02/07/2023] Open
Abstract
Recently, an increasing number of studies have demonstrated that induced pluripotent stem cells (iPSCs) and iPSC-derived cells display therapeutic effects, mainly via the paracrine mechanism in addition to their transdifferentiation ability. Exosomes have emerged as an important paracrine factor for iPSCs to repair injured cells through the delivery of bioactive components. Animal reports of iPSC-derived exosomes on various disease models are increasing, such as in heart, limb, liver, skin, bone, eye and neurological disease and so forth. This review aims to summarize the therapeutic effects of iPSC-derived exosomes on various disease models and their properties, such as angiogenesis, cell proliferation and anti-apoptosis, with the hopes of improving their potential role in clinical applications and functional restoration.
Collapse
Affiliation(s)
- Aline Yen Ling Wang
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, 5 Fu-hsing Street, Gueishan, Taoyuan 333, Taiwan
| |
Collapse
|
14
|
Fukushima H, Yoshioka M, Kawatou M, López-Dávila V, Takeda M, Kanda Y, Sekino Y, Yoshida Y, Yamashita JK. Specific induction and long-term maintenance of high purity ventricular cardiomyocytes from human induced pluripotent stem cells. PLoS One 2020; 15:e0241287. [PMID: 33137106 PMCID: PMC7605685 DOI: 10.1371/journal.pone.0241287] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 10/13/2020] [Indexed: 12/28/2022] Open
Abstract
Currently, cardiomyocyte (CM) differentiation methods require a purification step after CM induction to ensure the high purity of the cell population. Here we show an improved human CM differentiation protocol with which high-purity ventricular-type CMs can be obtained and maintained without any CM purification process. We induced and collected a mesodermal cell population (platelet-derived growth factor receptor-α (PDGFRα)-positive cells) that can respond to CM differentiation cues, and then stimulated CM differentiation by means of Wnt inhibition. This method reproducibly generated CMs with purities above 95% in several human pluripotent stem cell lines. Furthermore, these CM populations were maintained in culture at such high purity without any further CM purification step for over 200 days. The majority of these CMs (>95%) exhibited a ventricular-like phenotype with a tendency to structural and electrophysiological maturation, including T-tubule-like structure formation and the ability to respond to QT prolongation drugs. This is a simple and valuable method to stably generate CM populations suitable for cardiac toxicology testing, disease modeling and regenerative medicine.
Collapse
Affiliation(s)
- Hiroyuki Fukushima
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Miki Yoshioka
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Masahide Kawatou
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Department of Cardiovascular Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | - Víctor López-Dávila
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Masafumi Takeda
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Yuko Sekino
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Yoshinori Yoshida
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Jun K. Yamashita
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- * E-mail:
| |
Collapse
|
15
|
Optimizing the Use of iPSC-CMs for Cardiac Regeneration in Animal Models. Animals (Basel) 2020; 10:ani10091561. [PMID: 32887495 PMCID: PMC7552322 DOI: 10.3390/ani10091561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 12/29/2022] Open
Abstract
Simple Summary In 2006, the first induced pluripotent stem cells were generated by reprogramming skin cells. Induced pluripotent stem cells undergo fast cell division, can differentiate into many different cell types, can be patient-specific, and do not raise ethical issues. Thus, they offer great promise as in vitro disease models, drug toxicity testing platforms, and for autologous tissue regeneration. Heart failure is one of the major causes of death worldwide. It occurs when the heart cannot meet the body’s metabolic demands. Induced pluripotent stem cells can be differentiated into cardiac myocytes, can form patches resembling native cardiac tissue, and can engraft to the damaged heart. However, despite correct host/graft coupling, most animal studies demonstrate an arrhythmogenicity of the engrafted tissue and variable survival. This is partially because of the heterogeneity and immaturity of the cells. New evidence suggests that by modulating induced pluripotent stem cells-cardiac myocytes (iPSC-CM) metabolism by switching substrates and changing metabolic pathways, you can decrease iPSC-CM heterogeneity and arrhythmogenicity. Novel culture methods and tissue engineering along with animal models of heart failure are needed to fully unlock the potential of cardiac myocytes derived from induced pluripotent stem cells for cardiac regeneration. Abstract Heart failure (HF) is a common disease in which the heart cannot meet the metabolic demands of the body. It mostly occurs in individuals 65 years or older. Cardiac transplantation is the best option for patients with advanced HF. High numbers of patient-specific cardiac myocytes (CMs) can be generated from induced pluripotent stem cells (iPSCs) and can possibly be used to treat HF. While some studies found iPSC-CMS can couple efficiently to the damaged heart and restore cardiac contractility, almost all found iPSC-CM transplantation is arrhythmogenic, thus hampering the use of iPSC-CMs for cardiac regeneration. Studies show that iPSC-CM cultures are highly heterogeneous containing atrial-, ventricular- and nodal-like CMs. Furthermore, they have an immature phenotype, resembling more fetal than adult CMs. There is an urgent need to overcome these issues. To this end, a novel and interesting avenue to increase CM maturation consists of modulating their metabolism. Combined with careful engineering and animal models of HF, iPSC-CMs can be assessed for their potential for cardiac regeneration and a cure for HF.
Collapse
|
16
|
Branco MA, Cabral JM, Diogo MM. From Human Pluripotent Stem Cells to 3D Cardiac Microtissues: Progress, Applications and Challenges. Bioengineering (Basel) 2020; 7:E92. [PMID: 32785039 PMCID: PMC7552661 DOI: 10.3390/bioengineering7030092] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/30/2020] [Accepted: 08/06/2020] [Indexed: 12/19/2022] Open
Abstract
The knowledge acquired throughout the years concerning the in vivo regulation of cardiac development has promoted the establishment of directed differentiation protocols to obtain cardiomyocytes (CMs) and other cardiac cells from human pluripotent stem cells (hPSCs), which play a crucial role in the function and homeostasis of the heart. Among other developments in the field, the transition from homogeneous cultures of CMs to more complex multicellular cardiac microtissues (MTs) has increased the potential of these models for studying cardiac disorders in vitro and for clinically relevant applications such as drug screening and cardiotoxicity tests. This review addresses the state of the art of the generation of different cardiac cells from hPSCs and the impact of transitioning CM differentiation from 2D culture to a 3D environment. Additionally, current methods that may be employed to generate 3D cardiac MTs are reviewed and, finally, the adoption of these models for in vitro applications and their adaptation to medium- to high-throughput screening settings are also highlighted.
Collapse
Affiliation(s)
| | | | - Maria Margarida Diogo
- iBB-Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; (M.A.B.); (J.M.S.C.)
| |
Collapse
|
17
|
Sung TC, Su HC, Ling QD, Kumar SS, Chang Y, Hsu ST, Higuchi A. Efficient differentiation of human pluripotent stem cells into cardiomyocytes on cell sorting thermoresponsive surface. Biomaterials 2020; 253:120060. [PMID: 32450407 DOI: 10.1016/j.biomaterials.2020.120060] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 03/18/2020] [Accepted: 04/17/2020] [Indexed: 12/11/2022]
Abstract
The current differentiation process of human pluripotent stem cells (hPSCs) into cardiomyocytes to enhance the purity of hPSC-derived cardiomyocytes requires some purification processes, which are laborious processes. We developed cell sorting plates, which are prepared from coating thermoresponsive poly(N-isopropylacrylamide) and extracellular matrix proteins. After hPSCs were induced into cardiomyocytes on the thermoresponsive surface coated with laminin-521 for 15 days, the temperature of the cell culture plates was decreased to 8-9 °C to detach the cells partially from the thermoresponsive surface. The detached cells exhibited a higher cardiomyocyte marker of cTnT than the remaining cells on the thermoresponsive surface as well as the cardiomyocytes after purification using conventional cell selection. The detached cells expressed several cardiomyocyte markers, such as α-actinin, MLC2a and NKX2.5. This study suggested that the purification of hPSC-derived cardiomyocytes using cell sorting plates with the thermoresponsive surface is a promising method for the purification of hPSC-derived cardiomyocytes without conventional laborious processes.
Collapse
Affiliation(s)
- Tzu-Cheng Sung
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China; Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan, 32001, Taiwan
| | - Huan Chiao Su
- Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan, 32001, Taiwan
| | - Qing-Dong Ling
- Cathay Medical Research Institute, Cathay General Hospital, No. 32, Ln 160, Jian-Cheng Road, Hsi-Chi City, Taipei 221, Taiwan
| | - S Suresh Kumar
- Department of Medical Microbiology and Parasitology, Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia
| | - Yung Chang
- Department of Chemical Engineering, R&D Center for Membrane Technology, Chung Yuan Christian University, 200, Chung-Bei Rd., Chungli, Taoyuan, 320, Taiwan
| | - Shih-Tien Hsu
- Department of Internal Medicine, Taiwan Landseed Hospital, 77, Kuangtai Road, Pingjen City, Taoyuan, 32405, Taiwan
| | - Akon Higuchi
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China; Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan, 32001, Taiwan; Department of Chemical Engineering, R&D Center for Membrane Technology, Chung Yuan Christian University, 200, Chung-Bei Rd., Chungli, Taoyuan, 320, Taiwan; Wenzhou Institute, University of Chinese Academy of Science, No. 16, Xinsan Road, Hi-tech Industry Park, Wenzhou, Zhejiang, China; Center for Emergent Matter Science, Riken, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
| |
Collapse
|
18
|
Pan J, Lee YC, Lee HHC, Sung TC, Jen SH, Ban LK, Su HY, Chen DC, Hsu ST, Higuchi A, Chen H. Culture and differentiation of purified human adipose-derived stem cells by membrane filtration via nylon mesh filters. J Mater Chem B 2020; 8:5204-5214. [PMID: 32490480 DOI: 10.1039/d0tb00947d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Human adipose derived stem cells purified by the membrane migration method through filter membranes coated with vitronectin showed high osteogenic differentiation.
Collapse
|
19
|
Sunohara T, Morizane A, Matsuura S, Miyamoto S, Saito H, Takahashi J. MicroRNA-Based Separation of Cortico-Fugal Projection Neuron-Like Cells Derived From Embryonic Stem Cells. Front Neurosci 2019; 13:1141. [PMID: 31708734 PMCID: PMC6819314 DOI: 10.3389/fnins.2019.01141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 10/10/2019] [Indexed: 12/26/2022] Open
Abstract
The purification of pluripotent stem cell-derived cortico-fugal projection neurons (PSC-CFuPNs) is useful for disease modeling and cell therapies related to the dysfunction of cortical motor neurons, such as amyotrophic lateral sclerosis (ALS) or stroke. However, no CFuPN-specific surface markers for the purification are known. Recently, microRNAs (miRNAs) have been reported as alternatives to surface markers. Here, we investigated this possibility by applying the miRNA switch, an mRNA technology, to enrich PSC-CFuPNs. An array study of miRNAs in mouse fetal brain tissue revealed that CFuPNs highly express miRNA-124-3p at E14.5 and E16.5. In response, we designed a miRNA switched that responds to miRNA-124-3p and applied it to mouse embryonic stem cell (ESC)-derived cortical neurons. Flow cytometry and quantitative polymerase chain reaction (qPCR) analyses showed the miRNA-124-3p switch enriched CFuPN-like cells from this population. Immunocytechemical analysis confirmed vGlut1/Emx1/Bcl11b triple positive CFuPN-like cells were increased from 6.5 to 42%. Thus, our miRNA-124-3p switch can uniquely enrich live CFuPN-like cells from mouse ESC-derived cortical neurons.
Collapse
Affiliation(s)
- Tadashi Sunohara
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.,Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Asuka Morizane
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Satoshi Matsuura
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Susumu Miyamoto
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hirohide Saito
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Jun Takahashi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.,Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
20
|
Ohashi F, Miyagawa S, Yasuda S, Miura T, Kuroda T, Itoh M, Kawaji H, Ito E, Yoshida S, Saito A, Sameshima T, Kawai J, Sawa Y, Sato Y. CXCL4/PF4 is a predictive biomarker of cardiac differentiation potential of human induced pluripotent stem cells. Sci Rep 2019; 9:4638. [PMID: 30874579 PMCID: PMC6420577 DOI: 10.1038/s41598-019-40915-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 02/21/2019] [Indexed: 12/23/2022] Open
Abstract
Selection of human induced pluripotent stem cell (hiPSC) lines with high cardiac differentiation potential is important for regenerative therapy and drug screening. We aimed to identify biomarkers for predicting cardiac differentiation potential of hiPSC lines by comparing the gene expression profiles of six undifferentiated hiPSC lines with different cardiac differentiation capabilities. We used three platforms of gene expression analysis, namely, cap analysis of gene expression (CAGE), mRNA array, and microRNA array to efficiently screen biomarkers related to cardiac differentiation of hiPSCs. Statistical analysis revealed candidate biomarker genes with significant correlation between the gene expression levels in the undifferentiated hiPSCs and their cardiac differentiation potential. Of the candidate genes, PF4 was validated as a biomarker expressed in undifferentiated hiPSCs with high potential for cardiac differentiation in 13 additional hiPSC lines. Our observations suggest that PF4 may be a useful biomarker for selecting hiPSC lines appropriate for the generation of cardiomyocytes.
Collapse
Affiliation(s)
- Fumiya Ohashi
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan.,Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.,Department of Cellular & Gene Therapy Products, Osaka University Graduate School of Pharmaceutical Sciences, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Terumo Corporation, 1500 Inokuchi, Nakai-machi, Ashigarakami-gun, Kanagawa, 259-0151, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Satoshi Yasuda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan
| | - Takumi Miura
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan
| | - Takuya Kuroda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan
| | - Masayoshi Itoh
- Preventive Medicine and Diagnosis Innovation Program, RIKEN Center, 1-7-22, Suehirocho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Hideya Kawaji
- Preventive Medicine and Diagnosis Innovation Program, RIKEN Center, 1-7-22, Suehirocho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan.,Preventive Medicine and Applied Genomics Unit, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehirocho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Emiko Ito
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Shohei Yoshida
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Atsuhiro Saito
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Tadashi Sameshima
- Terumo Corporation, 1500 Inokuchi, Nakai-machi, Ashigarakami-gun, Kanagawa, 259-0151, Japan
| | - Jun Kawai
- Preventive Medicine and Diagnosis Innovation Program, RIKEN Center, 1-7-22, Suehirocho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Yoji Sato
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan. .,Department of Cellular & Gene Therapy Products, Osaka University Graduate School of Pharmaceutical Sciences, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan. .,Department of Quality Assurance Science for Pharmaceuticals, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan. .,Department of Translational Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka, 812-8582, Japan. .,LiSE Laboratory, Kanagawa Institute of Industrial Science and Technology, 3-25-13 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-0821, Japan.
| |
Collapse
|
21
|
Hong SP, Song S, Lee S, Jo H, Kim HK, Han J, Park JH, Cho SW. Regenerative potential of mouse embryonic stem cell-derived PDGFRα + cardiac lineage committed cells in infarcted myocardium. World J Stem Cells 2019; 11:44-54. [PMID: 30705714 PMCID: PMC6354102 DOI: 10.4252/wjsc.v11.i1.44] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 12/06/2018] [Accepted: 01/06/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pluripotent stem cell-derived cardiomyocytes (CMs) have become one of the most attractive cellular resources for cell-based therapy to rescue damaged cardiac tissue.
AIM We investigated the regenerative potential of mouse embryonic stem cell (ESC)-derived platelet-derived growth factor receptor-α (PDGFRα)+ cardiac lineage-committed cells (CLCs), which have a proliferative capacity but are in a morphologically and functionally immature state compared with differentiated CMs.
METHODS We induced mouse ESCs into PDGFRα+ CLCs and αMHC+ CMs using a combination of the small molecule cyclosporin A, the rho-associated coiled-coil kinase inhibitor Y27632, the antioxidant Trolox, and the ALK5 inhibitor EW7197. We implanted PDGFRα+ CLCs and differentiated αMHC+ CMs into a myocardial infarction (MI) murine model and performed functional analysis using transthoracic echocardiography (TTE) and histologic analysis.
RESULTS Compared with the untreated MI hearts, the anterior and septal regional wall motion and systolic functional parameters were notably and similarly improved in the MI hearts implanted with PDGFRα+ CLCs and αMHC+ CMs based on TTE. In histologic analysis, the untreated MI hearts contained a thinner ventricular wall than did the controls, while the ventricular walls of MI hearts implanted with PDGFRα+ CLCs and αMHC+ CMs were similarly thicker compared with that of the untreated MI hearts. Furthermore, implanted PDGFRα+ CLCs aligned and integrated with host CMs and were mostly differentiated into α-actinin+ CMs, and they did not convert into CD31+ endothelial cells or αSMA+ mural cells.
CONCLUSION PDGFRα+ CLCs from mouse ESCs exhibiting proliferative capacity showed a regenerative effect in infarcted myocardium. Therefore, mouse ESC-derived PDGFRα+ CLCs may represent a potential cellular resource for cardiac regeneration.
Collapse
Affiliation(s)
- Seon Pyo Hong
- Center for Vascular Research, Institute of Basic Science (IBS), Daejeon 34141, South Korea
| | - Sukhyun Song
- Center for Vascular Research, Institute of Basic Science (IBS), Daejeon 34141, South Korea
| | - Seungjoo Lee
- Department of Neurosurgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Hyeonju Jo
- Cardiovascular and Metabolic Disease Center, Department of Physiology, Department of Health Sciences and Technology, BK21 plus Project Team, Inje University College of Medicine, Busan 47392, South Korea
| | - Hyoung Kyu Kim
- Cardiovascular and Metabolic Disease Center, Department of Physiology, Department of Health Sciences and Technology, BK21 plus Project Team, Inje University College of Medicine, Busan 47392, South Korea
| | - Jin Han
- Cardiovascular and Metabolic Disease Center, Department of Physiology, Department of Health Sciences and Technology, BK21 plus Project Team, Inje University College of Medicine, Busan 47392, South Korea
| | - Jae-Hyeong Park
- Department of Cardiology in Internal Medicine, School of Medicine, Chungnam National University Hospital, Chungnam National University, Daejeon 35015, South Korea
| | - Sung Woo Cho
- Division of Cardiology, Department of Internal Medicine, Inje University College of Medicine, Seoul Paik Hospital, Seoul 04551, South Korea
- Cardiovascular and Metabolic Disease Center, Inje University College of Medicine, Busan 47392, South Korea
| |
Collapse
|
22
|
Hong SP, Song S, Lee S, Jo H, Kim HK, Han J, Park JH, Cho SW. Regenerative potential of mouse embryonic stem cell-derived PDGFRα + cardiac lineage committed cells in infarcted myocardium. World J Stem Cells 2019. [DOI: 10.4252/wjsc.v11.i1.45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
23
|
Human ISL1 + Ventricular Progenitors Self-Assemble into an In Vivo Functional Heart Patch and Preserve Cardiac Function Post Infarction. Mol Ther 2018; 26:1644-1659. [PMID: 29606507 PMCID: PMC6035340 DOI: 10.1016/j.ymthe.2018.02.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 02/09/2018] [Accepted: 02/11/2018] [Indexed: 11/26/2022] Open
Abstract
The generation of human pluripotent stem cell (hPSC)-derived ventricular progenitors and their assembly into a 3-dimensional in vivo functional ventricular heart patch has remained an elusive goal. Herein, we report the generation of an enriched pool of hPSC-derived ventricular progenitors (HVPs), which can expand, differentiate, self-assemble, and mature into a functional ventricular patch in vivo without the aid of any gel or matrix. We documented a specific temporal window, in which the HVPs will engraft in vivo. On day 6 of differentiation, HVPs were enriched by depleting cells positive for pluripotency marker TRA-1-60 with magnetic-activated cell sorting (MACS), and 3 million sorted cells were sub-capsularly transplanted onto kidneys of NSG mice where, after 2 months, they formed a 7 mm × 3 mm × 4 mm myocardial patch resembling the ventricular wall. The graft acquired several features of maturation: expression of ventricular marker (MLC2v), desmosomes, appearance of T-tubule-like structures, and electrophysiological action potential signature consistent with maturation, all this in a non-cardiac environment. We further demonstrated that HVPs transplanted into un-injured hearts of NSG mice remain viable for up to 8 months. Moreover, transplantation of 2 million HVPs largely preserved myocardial contractile function following myocardial infarction. Taken together, our study reaffirms the promising idea of using progenitor cells for regenerative therapy.
Collapse
|