1
|
Jang JH, Kim MS, Antao AM, Jo WJ, Kim HJ, Kim SJ, Choi MJ, Ramakrishna S, Kim KS. Bioactive Lipid O-cyclic phytosphingosine-1-phosphate Promotes Differentiation of Human Embryonic Stem Cells into Cardiomyocytes via ALK3/BMPR Signaling. Int J Mol Sci 2021; 22:ijms22137015. [PMID: 34209900 PMCID: PMC8267745 DOI: 10.3390/ijms22137015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 12/04/2022] Open
Abstract
Adult human cardiomyocytes have an extremely limited proliferative capacity, which poses a great barrier to regenerative medicine and research. Human embryonic stem cells (hESCs) have been proposed as an alternative source to generate large numbers of clinical grade cardiomyocytes (CMs) that can have potential therapeutic applications to treat cardiac diseases. Previous studies have shown that bioactive lipids are involved in diverse cellular responses including cardiogenesis. In this study, we explored the novel function of the chemically synthesized bioactive lipid O-cyclic phytosphingosine-1-phosphate (cP1P) as an inducer of cardiac differentiation. Here, we identified cP1P as a novel factor that significantly enhances the differentiation potential of hESCs into cardiomyocytes. Treatment with cP1P augments the beating colony number and contracting area of CMs. Furthermore, we elucidated the molecular mechanism of cP1P regulating SMAD1/5/8 signaling via the ALK3/BMP receptor cascade during cardiac differentiation. Our result provides a new insight for cP1P usage to improve the quality of CM differentiation for regenerative therapies.
Collapse
Affiliation(s)
- Ji-Hye Jang
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (J.-H.J.); (M.-S.K.); (A.M.A.); (W.-J.J.); (H.-J.K.)
| | - Min-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (J.-H.J.); (M.-S.K.); (A.M.A.); (W.-J.J.); (H.-J.K.)
| | - Ainsley Mike Antao
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (J.-H.J.); (M.-S.K.); (A.M.A.); (W.-J.J.); (H.-J.K.)
| | - Won-Jun Jo
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (J.-H.J.); (M.-S.K.); (A.M.A.); (W.-J.J.); (H.-J.K.)
| | - Hyung-Joon Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (J.-H.J.); (M.-S.K.); (A.M.A.); (W.-J.J.); (H.-J.K.)
| | - Su-Jin Kim
- Axceso Biopharma Co., Ltd., Yongin 16914, Korea; (S.-J.K.); (M.-J.C.)
| | - Myeong-Jun Choi
- Axceso Biopharma Co., Ltd., Yongin 16914, Korea; (S.-J.K.); (M.-J.C.)
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (J.-H.J.); (M.-S.K.); (A.M.A.); (W.-J.J.); (H.-J.K.)
- College of Medicine, Hanyang University, Seoul 04763, Korea
- Correspondence: or (S.R.); (K.-S.K.)
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (J.-H.J.); (M.-S.K.); (A.M.A.); (W.-J.J.); (H.-J.K.)
- College of Medicine, Hanyang University, Seoul 04763, Korea
- Correspondence: or (S.R.); (K.-S.K.)
| |
Collapse
|
2
|
Barreto S, Hamel L, Schiatti T, Yang Y, George V. Cardiac Progenitor Cells from Stem Cells: Learning from Genetics and Biomaterials. Cells 2019; 8:E1536. [PMID: 31795206 PMCID: PMC6952950 DOI: 10.3390/cells8121536] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiac Progenitor Cells (CPCs) show great potential as a cell resource for restoring cardiac function in patients affected by heart disease or heart failure. CPCs are proliferative and committed to cardiac fate, capable of generating cells of all the cardiac lineages. These cells offer a significant shift in paradigm over the use of human induced pluripotent stem cell (iPSC)-derived cardiomyocytes owing to the latter's inability to recapitulate mature features of a native myocardium, limiting their translational applications. The iPSCs and direct reprogramming of somatic cells have been attempted to produce CPCs and, in this process, a variety of chemical and/or genetic factors have been evaluated for their ability to generate, expand, and maintain CPCs in vitro. However, the precise stoichiometry and spatiotemporal activity of these factors and the genetic interplay during embryonic CPC development remain challenging to reproduce in culture, in terms of efficiency, numbers, and translational potential. Recent advances in biomaterials to mimic the native cardiac microenvironment have shown promise to influence CPC regenerative functions, while being capable of integrating with host tissue. This review highlights recent developments and limitations in the generation and use of CPCs from stem cells, and the trends that influence the direction of research to promote better application of CPCs.
Collapse
Affiliation(s)
- Sara Barreto
- Guy Hilton Research Centre, School of Pharmacy & Bioengineering, Keele University, Staffordshire ST4 7QB, UK; (S.B.); (T.S.); (Y.Y.)
| | | | - Teresa Schiatti
- Guy Hilton Research Centre, School of Pharmacy & Bioengineering, Keele University, Staffordshire ST4 7QB, UK; (S.B.); (T.S.); (Y.Y.)
| | - Ying Yang
- Guy Hilton Research Centre, School of Pharmacy & Bioengineering, Keele University, Staffordshire ST4 7QB, UK; (S.B.); (T.S.); (Y.Y.)
| | - Vinoj George
- Guy Hilton Research Centre, School of Pharmacy & Bioengineering, Keele University, Staffordshire ST4 7QB, UK; (S.B.); (T.S.); (Y.Y.)
| |
Collapse
|
3
|
Cardiac Progenitors Induced from Human Induced Pluripotent Stem Cells with Cardiogenic Small Molecule Effectively Regenerate Infarcted Hearts and Attenuate Fibrosis. Shock 2019; 50:627-639. [PMID: 29485473 DOI: 10.1097/shk.0000000000001133] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Cardiac progenitor cells (CPCs) being multipotent offer a promising source for cardiac repair due to their ability to proliferate and multiply into cardiac lineage cells. Here, we explored a novel strategy for human CPCs generation from human induced pluripotent stem cells (hiPSCs) using a cardiogenic small molecule, isoxazole (ISX-9) and their ability to grow in the scar tissue for functional improvement in the infarcted myocardium. CPCs were induced from hiPSCs with ISX-9. CPCs were characterized by immunocytochemistry and RT-PCR. The CPC survival and differentiation in the infarcted hearts were determined by in vivo transplantation in immunodeficient mice following left anterior descending artery ligation and their effects were determined on fibrosis and functional improvement. ISX-9 simultaneously induced expression of cardiac transcription factors, NK2 homeobox 5, islet-1, GATA binding protein 4, myocyte enhancer factor-2 in hiPSCs within 3 days of treatment and successfully differentiated into three cardiac lineages in vitro. Messenger RNA and microRNA-sequencing results showed that ISX-9 targeted multiple cardiac differentiation, proliferation signaling pathways and upregulated myogenesis and cardiac hypertrophy related-microRNA. ISX-9 activated multiple pathways including transforming growth factor β induced epithelial-mesenchymal transition signaling, canonical, and non-canonical Wnt signaling at different stages of cardiac differentiation. CPCs transplantation promoted myoangiogenesis, attenuated fibrosis, and led to functional improvement in treated mice.
Collapse
|
4
|
Li L, Wang Q, Yuan Z, Chen A, Liu Z, Li H, Wang Z. Long non-coding RNA H19 contributes to hypoxia-induced CPC injury by suppressing Sirt1 through miR-200a-3p. Acta Biochim Biophys Sin (Shanghai) 2018; 50:950-959. [PMID: 30137188 DOI: 10.1093/abbs/gmy093] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Indexed: 12/28/2022] Open
Abstract
Cardiomyocyte death is the chief obstacle that prevents the heart function recovery in myocardial infarction (MI)-induced heart failure (HF). Cardiac progenitor cells (CPCs)-based myocardial regeneration has provided a promising method for heart function recovery after MI. However, CPCs can easily lose their proliferation ability due to oxygen deficiency in infarcted myocardium. Revealing the underlying molecular mechanism for CPC proliferation is critical for effective MI therapy. In the present study, we set up a CoCl2-induced hypoxia model in CPCs. We found that the expression of long non-coding RNA H19 was significantly down-regulated in CPCs after hypoxia stimuli. In addition, H19 suppression attenuated the proliferation and migration of CPCs under hypoxia stress. Furthermore, we discovered that H19 regulated the proliferation and migration of CPCs through mediating the expression of Sirt1 which is a target of miR-200a-3p under hypoxia. In conclusion, our findings demonstrate a novel regulatory mechanism for the proliferation and migration of CPCs under hypoxia condition, which provides useful information for the development of new therapeutic targets for MI therapy.
Collapse
Affiliation(s)
- Linlin Li
- College of Life Sciences, Peking University, Beijing, China
| | - Qiuyun Wang
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai, China
| | - Zhize Yuan
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai, China
| | - Anqing Chen
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai, China
| | - Zuyun Liu
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai, China
| | - Haiqing Li
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai, China
| | - Zhe Wang
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai, China
| |
Collapse
|
5
|
Zhang Y, Cao N, Huang Y, Spencer CI, Fu JD, Yu C, Liu K, Nie B, Xu T, Li K, Xu S, Bruneau BG, Srivastava D, Ding S. Expandable Cardiovascular Progenitor Cells Reprogrammed from Fibroblasts. Cell Stem Cell 2016; 18:368-81. [PMID: 26942852 DOI: 10.1016/j.stem.2016.02.001] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 12/11/2015] [Accepted: 02/09/2016] [Indexed: 12/31/2022]
Abstract
Stem cell-based approaches to cardiac regeneration are increasingly viable strategies for treating heart failure. Generating abundant and functional autologous cells for transplantation in such a setting, however, remains a significant challenge. Here, we isolated a cell population with extensive proliferation capacity and restricted cardiovascular differentiation potentials during cardiac transdifferentiation of mouse fibroblasts. These induced expandable cardiovascular progenitor cells (ieCPCs) proliferated extensively for more than 18 passages in chemically defined conditions, with 10(5) starting fibroblasts robustly producing 10(16) ieCPCs. ieCPCs expressed cardiac signature genes and readily differentiated into functional cardiomyocytes (CMs), endothelial cells (ECs), and smooth muscle cells (SMCs) in vitro, even after long-term expansion. When transplanted into mouse hearts following myocardial infarction, ieCPCs spontaneously differentiated into CMs, ECs, and SMCs and improved cardiac function for up to 12 weeks after transplantation. Thus, ieCPCs are a powerful system to study cardiovascular specification and provide strategies for regenerative medicine in the heart.
Collapse
Affiliation(s)
- Yu Zhang
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Nan Cao
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yu Huang
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | - C Ian Spencer
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | - Ji-Dong Fu
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Department of Medicine, Heart and Vascular Research Center, MetroHealth Campus, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Chen Yu
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kai Liu
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Baoming Nie
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Tao Xu
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ke Li
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Shaohua Xu
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Benoit G Bruneau
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Roddenberry Center for Stem Cell Biology and Medicine, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Deepak Srivastava
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Roddenberry Center for Stem Cell Biology and Medicine, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sheng Ding
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
6
|
Bao X, Lian X, Hacker TA, Schmuck EG, Qian T, Bhute VJ, Han T, Shi M, Drowley L, Plowright A, Wang QD, Goumans MJ, Palecek SP. Long-term self-renewing human epicardial cells generated from pluripotent stem cells under defined xeno-free conditions. Nat Biomed Eng 2016; 1. [PMID: 28462012 PMCID: PMC5408455 DOI: 10.1038/s41551-016-0003] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The epicardium contributes both multi-lineage descendants and paracrine factors to the heart during cardiogenesis and cardiac repair, underscoring its potential for cardiac regenerative medicine. Yet little is known about the cellular and molecular mechanisms that regulate human epicardial development and regeneration. Here, we show that the temporal modulation of canonical Wnt signaling is sufficient for epicardial induction from 6 different human pluripotent stem cell (hPSC) lines, including a WT1-2A-eGFP knock-in reporter line, under chemically-defined, xeno-free conditions. We also show that treatment with transforming growth factor beta (TGF-β)-signalling inhibitors permitted long-term expansion of the hPSC-derived epicardial cells, resulting in a more than 25 population doublings of WT1+ cells in homogenous monolayers. The hPSC-derived epicardial cells were similar to primary epicardial cells both in vitro and in vivo, as determined by morphological and functional assays, including RNA-seq. Our findings have implications for the understanding of self-renewal mechanisms of the epicardium and for epicardial regeneration using cellular or small-molecule therapies.
Collapse
Affiliation(s)
- Xiaoping Bao
- Department of Chemical & Biological Engineering, University of Wisconsin, Madison, WI 53706, USA
| | - Xiaojun Lian
- Department of Chemical & Biological Engineering, University of Wisconsin, Madison, WI 53706, USA.,Departments of Biomedical Engineering, Biology and Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Timothy A Hacker
- Department of Medicine, University of Wisconsin, Madison, WI 53706, USA
| | - Eric G Schmuck
- Department of Medicine, University of Wisconsin, Madison, WI 53706, USA
| | - Tongcheng Qian
- Department of Chemical & Biological Engineering, University of Wisconsin, Madison, WI 53706, USA
| | - Vijesh J Bhute
- Department of Chemical & Biological Engineering, University of Wisconsin, Madison, WI 53706, USA
| | - Tianxiao Han
- Department of Chemical & Biological Engineering, University of Wisconsin, Madison, WI 53706, USA
| | - Mengxuan Shi
- Department of Chemical & Biological Engineering, University of Wisconsin, Madison, WI 53706, USA
| | - Lauren Drowley
- Department of Cardiovascular and Metabolic Diseases Innovative Medicine Unit, AstraZeneca, Mölndal, 43183, Sweden
| | - Alleyn Plowright
- Department of Cardiovascular and Metabolic Diseases Innovative Medicine Unit, AstraZeneca, Mölndal, 43183, Sweden
| | - Qing-Dong Wang
- Department of Cardiovascular and Metabolic Diseases Innovative Medicine Unit, AstraZeneca, Mölndal, 43183, Sweden
| | - Marie-Jose Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, 2333 ZC Leiden, the Netherlands
| | - Sean P Palecek
- Department of Chemical & Biological Engineering, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
7
|
Månsson-Broberg A, Rodin S, Bulatovic I, Ibarra C, Löfling M, Genead R, Wärdell E, Felldin U, Granath C, Alici E, Le Blanc K, Smith CIE, Salašová A, Westgren M, Sundström E, Uhlén P, Arenas E, Sylvén C, Tryggvason K, Corbascio M, Simonson OE, Österholm C, Grinnemo KH. Wnt/β-Catenin Stimulation and Laminins Support Cardiovascular Cell Progenitor Expansion from Human Fetal Cardiac Mesenchymal Stromal Cells. Stem Cell Reports 2016; 6:607-617. [PMID: 27052314 PMCID: PMC4834052 DOI: 10.1016/j.stemcr.2016.02.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 02/19/2016] [Accepted: 02/22/2016] [Indexed: 11/29/2022] Open
Abstract
The intrinsic regenerative capacity of human fetal cardiac mesenchymal stromal cells (MSCs) has not been fully characterized. Here we demonstrate that we can expand cells with characteristics of cardiovascular progenitor cells from the MSC population of human fetal hearts. Cells cultured on cardiac muscle laminin (LN)-based substrata in combination with stimulation of the canonical Wnt/β-catenin pathway showed increased gene expression of ISL1, OCT4, KDR, and NKX2.5. The majority of cells stained positive for PDGFR-α, ISL1, and NKX2.5, and subpopulations also expressed the progenitor markers TBX18, KDR, c-KIT, and SSEA-1. Upon culture of the cardiac MSCs in differentiation media and on relevant LNs, portions of the cells differentiated into spontaneously beating cardiomyocytes, and endothelial and smooth muscle-like cells. Our protocol for large-scale culture of human fetal cardiac MSCs enables future exploration of the regenerative functions of these cells in the context of myocardial injury in vitro and in vivo. Cells with progenitor properties can be expanded from human fetal cardiac MSCs Specific LNs support expansion and differentiation of cardiac MSCs The fetal cardiac MSCs express ISL1, PDGFR-α, and NKX2.5 Subpopulations express the progenitor markers KDR, SSEA-1, c-KIT, and TBX18
Collapse
Affiliation(s)
- Agneta Månsson-Broberg
- Division of Cardiology, Department of Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Sergey Rodin
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Ivana Bulatovic
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Cristián Ibarra
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; Cardiovascular & Metabolic Diseases, Innovative Medicines and Early Development, AstraZeneca R&D, 43150 Mölndal, Sweden
| | - Marie Löfling
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Rami Genead
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Eva Wärdell
- Division of Cardiology, Department of Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Ulrika Felldin
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Carl Granath
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Evren Alici
- Division of Hematology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, 17177 Stockholm, Sweden; Cell Therapy Institute, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| | - Katarina Le Blanc
- Division of Hematology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, 17177 Stockholm, Sweden; Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - C I Edvard Smith
- Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - Alena Salašová
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Magnus Westgren
- CLINTEC, Division of Obstetrics and Gynecology, Karolinska Institutet, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - Erik Sundström
- Division of Neurodegeneration, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - Per Uhlén
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Ernest Arenas
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Christer Sylvén
- Division of Cardiology, Department of Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Karl Tryggvason
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; Duke-NUS Graduate Medical School, Durham, NC 27710, USA
| | - Matthias Corbascio
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Oscar E Simonson
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Cecilia Österholm
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden; Cell Therapy Institute, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| | - Karl-Henrik Grinnemo
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden; Cell Therapy Institute, Nova Southeastern University, Fort Lauderdale, FL 33314, USA.
| |
Collapse
|
8
|
Lalit PA, Hei DJ, Raval AN, Kamp TJ. Induced pluripotent stem cells for post-myocardial infarction repair: remarkable opportunities and challenges. Circ Res 2014; 114:1328-45. [PMID: 24723658 DOI: 10.1161/circresaha.114.300556] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Coronary artery disease with associated myocardial infarction continues to be a major cause of death and morbidity around the world, despite significant advances in therapy. Patients who have large myocardial infarctions are at highest risk for progressive heart failure and death, and cell-based therapies offer new hope for these patients. A recently discovered cell source for cardiac repair has emerged as a result of a breakthrough reprogramming somatic cells to induced pluripotent stem cells (iPSCs). The iPSCs can proliferate indefinitely in culture and can differentiate into cardiac lineages, including cardiomyocytes, smooth muscle cells, endothelial cells, and cardiac progenitors. Thus, large quantities of desired cell products can be generated without being limited by cellular senescence. The iPSCs can be obtained from patients to allow autologous therapy or, alternatively, banks of human leukocyte antigen diverse iPSCs are possible for allogeneic therapy. Preclinical animal studies using a variety of cell preparations generated from iPSCs have shown evidence of cardiac repair. Methodology for the production of clinical grade products from human iPSCs is in place. Ongoing studies for the safety of various iPSC preparations with regard to the risk of tumor formation, immune rejection, induction of arrhythmias, and formation of stable cardiac grafts are needed as the field advances toward the first-in-man trials of iPSCs after myocardial infarction.
Collapse
Affiliation(s)
- Pratik A Lalit
- From the Department of Medicine (P.A.L., A.N.R., T.J.K.), Molecular and Cellular Pharmacology Program (P.A.L., T.J.K.), and Stem Cell and Regenerative Medicine Center (P.A.L., D.J.H., A.N.R., T.J.K.), Waisman Biomanufacturing at University of Wisconsin, Madison (D.J.H.)
| | | | | | | |
Collapse
|
9
|
Cao N, Liang H, Huang J, Wang J, Chen Y, Chen Z, Yang HT. Highly efficient induction and long-term maintenance of multipotent cardiovascular progenitors from human pluripotent stem cells under defined conditions. Cell Res 2013; 23:1119-32. [PMID: 23896987 DOI: 10.1038/cr.2013.102] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 05/05/2013] [Accepted: 05/17/2013] [Indexed: 12/23/2022] Open
Abstract
Cardiovascular progenitor cells (CVPCs) derived from human pluripotent stem cells (hPSCs), including human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), hold great promise for the study of cardiovascular development and cell-based therapy of heart diseases, but their applications are challenged by the difficulties in their efficient generation and stable maintenance. This study aims to develop chemically defined systems for robust generation and stable propagation of hPSC-derived CVPCs by modulating the key early developmental pathways involved in human cardiovascular specification and CVPC self-renewal. Herein we report that a combination of bone morphogenetic protein 4 (BMP4), glycogen synthase kinase 3 (GSK3) inhibitor CHIR99021 and ascorbic acid is sufficient to rapidly convert monolayer-cultured hPSCs, including hESCs and hiPSCs, into homogeneous CVPCs in a chemically defined medium under feeder- and serum-free culture conditions. These CVPCs stably self-renewed under feeder- and serum-free conditions and expanded over 10(7)-fold when the differentiation-inducing signals from BMP, GSK3 and Activin/Nodal pathways were simultaneously eliminated. Furthermore, these CVPCs exhibited expected genome-wide molecular features of CVPCs, retained potentials to generate major cardiovascular lineages including cardiomyocytes, smooth muscle cells and endothelial cells in vitro, and were non-tumorigenic in vivo. Altogether, the established systems reported here permit efficient generation and stable maintenance of hPSC-derived CVPCs, which represent a powerful tool to study early embryonic cardiovascular development and provide a potentially safe source of cells for myocardial regenerative medicine.
Collapse
Affiliation(s)
- Nan Cao
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | | | | | | | | | | | | |
Collapse
|
10
|
Brenner C, David R, Franz WM. Cardiovascular Stem Cells. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
11
|
Abstract
The heart as a functional organ first appeared in bilaterians as a single peristaltic pump and evolved through arthropods, fish, amphibians, and finally mammals into a four-chambered engine controlling blood-flow within the body. The acquisition of cardiac complexity in the evolving heart was a product of gene duplication events and the co-option of novel signaling pathways to an ancestral cardiac-specific gene network. T-box factors belong to an evolutionary conserved family of transcriptional regulators with diverse roles in development. Their regulatory functions are integral in the initiation and potentiation of heart development, and mutations in these genes are associated with congenital heart defects. In this review we will discuss the evolutionary conserved cardiac regulatory functions of this family as well as their implication in disease in an aim to facilitate future gene-targeted and regenerative therapeutic remedies.
Collapse
Affiliation(s)
- Fadi Hariri
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, C.P. 6128, Succursale, Centre-ville Montréal, Quebec, H3C3J7, Canada
| | | | | |
Collapse
|
12
|
Zhou Y, Kim J, Yuan X, Braun T. Epigenetic modifications of stem cells: a paradigm for the control of cardiac progenitor cells. Circ Res 2011; 109:1067-81. [PMID: 21998298 DOI: 10.1161/circresaha.111.243709] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Stem cells of all types are characterized by the ability to self-renew and to differentiate. Multiple lines of evidence suggest that both maintenance of stemness and lineage commitment, including determination of the cardiomyogenic lineage, are tightly controlled by epigenetic mechanisms such as DNA methylation, histone modifications, and ATP-dependent chromatin remodeling. Epigenetic mechanisms are intrinsically reversible, interdependent, and highly dynamic in regulation of chromatin structure and specific gene transcription programs, thereby contributing to stem cell homeostasis. Here, we review the current understanding of epigenetic mechanisms involved in regulation of stem cell self-renewal and differentiation and in the control of cardiac progenitor cell commitment during heart development. Further progress in this area will help to decipher the epigenetic landscape in stem and progenitor cells and facilitate manipulation of stem cells for regenerative applications.
Collapse
Affiliation(s)
- Yonggang Zhou
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim Germany.
| | | | | | | |
Collapse
|
13
|
Cao N, Liu Z, Chen Z, Wang J, Chen T, Zhao X, Ma Y, Qin L, Kang J, Wei B, Wang L, Jin Y, Yang HT. Ascorbic acid enhances the cardiac differentiation of induced pluripotent stem cells through promoting the proliferation of cardiac progenitor cells. Cell Res 2011; 22:219-36. [PMID: 22143566 DOI: 10.1038/cr.2011.195] [Citation(s) in RCA: 178] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Generation of induced pluripotent stem cells (iPSCs) has opened new avenues for the investigation of heart diseases, drug screening and potential autologous cardiac regeneration. However, their application is hampered by inefficient cardiac differentiation, high interline variability, and poor maturation of iPSC-derived cardiomyocytes (iPS-CMs). To identify efficient inducers for cardiac differentiation and maturation of iPSCs and elucidate the mechanisms, we systematically screened sixteen cardiomyocyte inducers on various murine (m) iPSCs and found that only ascorbic acid (AA) consistently and robustly enhanced the cardiac differentiation of eleven lines including eight without spontaneous cardiogenic potential. We then optimized the treatment conditions and demonstrated that differentiation day 2-6, a period for the specification of cardiac progenitor cells (CPCs), was a critical time for AA to take effect. This was further confirmed by the fact that AA increased the expression of cardiovascular but not mesodermal markers. Noteworthily, AA treatment led to approximately 7.3-fold (miPSCs) and 30.2-fold (human iPSCs) augment in the yield of iPS-CMs. Such effect was attributed to a specific increase in the proliferation of CPCs via the MEK-ERK1/2 pathway by through promoting collagen synthesis. In addition, AA-induced cardiomyocytes showed better sarcomeric organization and enhanced responses of action potentials and calcium transients to β-adrenergic and muscarinic stimulations. These findings demonstrate that AA is a suitable cardiomyocyte inducer for iPSCs to improve cardiac differentiation and maturation simply, universally, and efficiently. These findings also highlight the importance of stimulating CPC proliferation by manipulating extracellular microenvironment in guiding cardiac differentiation of the pluripotent stem cells.
Collapse
Affiliation(s)
- Nan Cao
- Key Laboratory of Stem Cell Biology, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) & Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Zeng B, Ren XF, Cao F, Zhou XY, Zhang J. Developmental patterns and characteristics of epicardial cell markers Tbx18 and Wt1 in murine embryonic heart. J Biomed Sci 2011; 18:67. [PMID: 21871065 PMCID: PMC3182971 DOI: 10.1186/1423-0127-18-67] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2011] [Accepted: 08/26/2011] [Indexed: 01/05/2023] Open
Abstract
Background Although recent studies have highlighted the role of epicardial cells during cardiac development and regeneration, their cardiomyogenic potential is still controversial due to the question of lineage tracing of epicardial cells. The present study therefore aimed to examine the the expression of Tbx18 and Wt1 in embryonic heart and to identify whether Tbx18 and Wt1 themselves expressed in the cardiomyocyte. Methods Mouse embryonic hearts were collected at different stages for immunofluorescence costaining with either Tbx18 and the cardiac transcription factor Nkx2.5 or Wilms tumor 1 (Wt1) and Nkx2.5. Results Tbx18 and Wt1, but not Nkx2.5, were expressed in the proepicardium and epicardium. Tbx18 was expressed in cells within the heart from E10.5 to at least E14.5; these Tbx18-expressing cells were Nkx2.5 positive, except for a few cells that were Nkx2.5 negative at E14.5. Wt1 was expressed in cells within the heart from E12.5 to at least E14.5, but these Wt1-expressing cells were Nkx2.5 negative. Conclusion The data obtained in this study demonstrate that Tbx18 is expressed in the cardiomyocytes from E10.5 to at least E14.5, and Wt1 is expressed within the heart from E12.5 to at least E14.5, but not in the cardiomyocyte. These findings may provide new insights on the role of the epicardial cells in cardiac regeneration.
Collapse
Affiliation(s)
- Bin Zeng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China.
| | | | | | | | | |
Collapse
|
15
|
Liu Y, Li Y, Li T, Lu H, Jia Z, Wang W, Chen P, Ma K, Zhou C. POU homeodomain protein OCT1 modulates islet 1 expression during cardiac differentiation of P19CL6 cells. Cell Mol Life Sci 2011; 68:1969-82. [PMID: 20960024 PMCID: PMC11115038 DOI: 10.1007/s00018-010-0544-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 09/02/2010] [Accepted: 09/28/2010] [Indexed: 11/30/2022]
Abstract
Islet 1 (ISL1), a marker of cardiac progenitors, plays a crucial role in cardiogenesis. However, the precise mechanism underlying the activation of its expression is not fully understood. Using the cardiac differentiation model of P19CL6 cells, we show that POU homeodomain protein, OCT1, modulates Isl1 expression in the process of cardiac differentiation. Oct1 knock-down resulted in reduction of Isl1 expression and downregulated mesodermal, cardiac-specific, and signal pathway gene expression. Additionally, the octamer motif located in the proximal region of Isl1 promoter is essential to Isl1 transcriptional activation. Mutation of this motif remarkably decreased Isl1 transcription. Although both OCT1 and OCT4 bound to this motif, it was OCT1 rather than OCT4 that modulated Isl1 expression. Furthermore, the correlation of OCT1 in regulation of Isl1 was revealed by in situ hybridization in early embryos. Collectively, our data highlight a novel role of OCT1 in the regulation of Isl1 expression.
Collapse
Affiliation(s)
- Yinan Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District Beijing, 100191 China
| | - Yanming Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District Beijing, 100191 China
| | - Tao Li
- Department of Biology, College of Chemistry and Life Science, Zhejiang Normal University, 688 Yingbing Road, Jinhua, 321004 Zhejiang Province China
| | - Huafei Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District Beijing, 100191 China
| | - Zhuqing Jia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District Beijing, 100191 China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University, Beijing, 100191 China
| | - Weiping Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District Beijing, 100191 China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University, Beijing, 100191 China
| | - Ping Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District Beijing, 100191 China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University, Beijing, 100191 China
| | - Kangtao Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District Beijing, 100191 China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University, Beijing, 100191 China
| | - Chunyan Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District Beijing, 100191 China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University, Beijing, 100191 China
| |
Collapse
|
16
|
Cardiovascular Stem Cells. Regen Med 2011. [DOI: 10.1007/978-90-481-9075-1_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|