1
|
Extracellular vesicles from human embryonic stem cell-derived cardiovascular progenitor cells promote cardiac infarct healing through reducing cardiomyocyte death and promoting angiogenesis. Cell Death Dis 2020; 11:354. [PMID: 32393784 PMCID: PMC7214429 DOI: 10.1038/s41419-020-2508-y] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 04/12/2020] [Accepted: 04/14/2020] [Indexed: 02/07/2023]
Abstract
Human pluripotent stem cells (hPSCs)-derived cardiovascular progenitor cells (CVPCs) are a promising source for myocardial repair, while the mechanisms remain largely unknown. Extracellular vesicles (EVs) are known to mediate cell–cell communication, however, the efficacy and mechanisms of hPSC-CVPC-secreted EVs (hCVPC-EVs) in the infarct healing when given at the acute phase of myocardial infarction (MI) are unknown. Here, we report the cardioprotective effects of the EVs secreted from hESC-CVPCs under normoxic (EV-N) and hypoxic (EV-H) conditions in the infarcted heart and the long noncoding RNA (lncRNA)-related mechanisms. The hCVPC-EVs were confirmed by electron microscopy, nanoparticle tracking, and immunoblotting analysis. Injection of hCVPC-EVs into acutely infracted murine myocardium significantly improved cardiac function and reduced fibrosis at day 28 post MI, accompanied with the improved vascularization and cardiomyocyte survival at border zones. Consistently, hCVPC-EVs enhanced the tube formation and migration of human umbilical vein endothelial cells (HUVECs), improved the cell viability, and attenuated the lactate dehydrogenase release of neonatal rat cardiomyocytes (NRCMs) with oxygen glucose deprivation (OGD) injury. Moreover, the improvement of the EV-H in cardiomyocyte survival and tube formation of HUVECs was significantly better than these in the EV-N. RNA-seq analysis revealed a high abundance of the lncRNA MALAT1 in the EV-H. Its abundance was upregulated in the infarcted myocardium and cardiomyocytes treated with hCVPC-EVs. Overexpression of human MALAT1 improved the cell viability of NRCM with OGD injury, while knockdown of MALAT1 inhibited the hCVPC-EV-promoted tube formation of HUVECs. Furthermore, luciferase activity assay, RNA pull-down, and manipulation of miR-497 levels showed that MALAT1 improved NRCMs survival and HUVEC tube formation through targeting miR-497. These results reveal that hCVPC-EVs promote the infarct healing through improvement of cardiomyocyte survival and angiogenesis. The cardioprotective effects of hCVPC-EVs can be enhanced by hypoxia-conditioning of hCVPCs and are partially contributed by MALAT1 via targeting the miRNA.
Collapse
|
2
|
Wang J, Liu M, Wu Q, Li Q, Gao L, Jiang Y, Deng B, Huang W, Bi W, Chen Z, Chin YE, Paul C, Wang Y, Yang HT. Human Embryonic Stem Cell-Derived Cardiovascular Progenitors Repair Infarcted Hearts Through Modulation of Macrophages via Activation of Signal Transducer and Activator of Transcription 6. Antioxid Redox Signal 2019; 31:369-386. [PMID: 30854870 PMCID: PMC6602123 DOI: 10.1089/ars.2018.7688] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Aims: Human embryonic stem cell derived-cardiovascular progenitor cells (hESC-CVPCs) are a promising cell source for cardiac repair, while the underlying mechanisms need to be elucidated. We recently observed cardioprotective effects of human pluripotent stem cell (hPSC)-CVPCs in infarcted nonhuman primates, but their effects on inflammation during early phase of myocardial infarction (MI) and the contribution of such effect to the cardioprotection are unclear. Results: Injection of hESC-CVPCs into acutely infarcted myocardium significantly ameliorated the functional worsening and scar formation, concomitantly with reduced inflammatory reactions and cardiomyocyte apoptosis as well as increased vascularization. Moreover, hESC-CVPCs modulated cardiac macrophages toward a reparative phenotype in the infarcted hearts, and such modulation was further confirmed in vitro using human cardiovascular progenitor cell (hCVPC)-conditioned medium (hCVPC-CdM) and highly contained interleukin (IL)-4/IL-13. Furthermore, signal transducer and activator of transcription 6 (STAT6) was activated in hCVPC-CdM- and IL-4/IL-13-treated macrophages in vitro and in hESC-CVPC-implanted MI hearts, resulting in the polarization of macrophages toward a reparative phenotype in the post-MI hearts. However, hESC-CVPC-mediated modulation on macrophages and cardioprotection were abolished in STAT6-deficient MI mice. Innovation: This is the first report about the immunoregulatory role played by hESC-CVPCs in the macrophage polarization in the infarcted hearts, its importance for the infarct repair, and the underlying signaling pathway. The findings provide new insight into the mechanism of microenvironmental regulation of stem cell-based therapy during acute MI. Conclusions: Implantion of hESC-CVPCs during the early phase of MI promotes infarct repair via the modulation of macrophage polarization through secreted cytokine-mediated STAT6 activation. The findings suggest a therapeutic potential by modulating macrophage polarization during acute phase of MI.
Collapse
Affiliation(s)
- Jinxi Wang
- 1 CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Jiao Tong University School of Medicine and Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences (CAS), Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Meilan Liu
- 1 CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Jiao Tong University School of Medicine and Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences (CAS), Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Qiang Wu
- 1 CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Jiao Tong University School of Medicine and Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences (CAS), Chinese Academy of Sciences, Shanghai, People's Republic of China.,2 Institute for Stem Cell and Regeneration, Chinese Academy of Sciences (CAS), Beijing, China
| | - Qiang Li
- 1 CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Jiao Tong University School of Medicine and Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences (CAS), Chinese Academy of Sciences, Shanghai, People's Republic of China.,2 Institute for Stem Cell and Regeneration, Chinese Academy of Sciences (CAS), Beijing, China
| | - Ling Gao
- 1 CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Jiao Tong University School of Medicine and Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences (CAS), Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Yun Jiang
- 1 CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Jiao Tong University School of Medicine and Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences (CAS), Chinese Academy of Sciences, Shanghai, People's Republic of China.,2 Institute for Stem Cell and Regeneration, Chinese Academy of Sciences (CAS), Beijing, China
| | - Boxiong Deng
- 3 CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Tumor and Stem Cell, SIBS, Chinese Academy of Sciences (CAS), Shanghai, People's Republic of China
| | - Wei Huang
- 4 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio
| | - Wei Bi
- 1 CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Jiao Tong University School of Medicine and Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences (CAS), Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Zhongyan Chen
- 1 CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Jiao Tong University School of Medicine and Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences (CAS), Chinese Academy of Sciences, Shanghai, People's Republic of China.,2 Institute for Stem Cell and Regeneration, Chinese Academy of Sciences (CAS), Beijing, China
| | - Y Eugene Chin
- 3 CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Tumor and Stem Cell, SIBS, Chinese Academy of Sciences (CAS), Shanghai, People's Republic of China
| | - Christian Paul
- 4 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio
| | - Yigang Wang
- 4 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio
| | - Huang-Tian Yang
- 1 CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Jiao Tong University School of Medicine and Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences (CAS), Chinese Academy of Sciences, Shanghai, People's Republic of China.,2 Institute for Stem Cell and Regeneration, Chinese Academy of Sciences (CAS), Beijing, China
| |
Collapse
|
3
|
Jebeniani I, Ding S, Pucéat M. Improved Protocol for Cardiac Differentiation and Maturation of Pluripotent Stem Cells. Methods Mol Biol 2019; 1994:71-77. [PMID: 31124105 DOI: 10.1007/978-1-4939-9477-9_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Pluripotent stem cells feature the capacity to differentiate into any somatic cell types including cardiomyocytes. We report a cost-effective and simple protocol for the differentiation of specific ventricular cardiomyocytes. These cells are elongated, do not spontaneously beat, and do not feature any Ca2+-transient, an index of their stage of maturation toward adult cardiac cells. They represent a suitable model to screen both the efficiency and toxicology of drugs.
Collapse
Affiliation(s)
- Imen Jebeniani
- INSERM U1251 Université Aix-Marseille, MMG, Marseille, France
| | - Shunli Ding
- INSERM U1251 Université Aix-Marseille, MMG, Marseille, France
| | - Michel Pucéat
- INSERM U1251 Université Aix-Marseille, MMG, Marseille, France.
| |
Collapse
|
4
|
Scalable Cardiac Differentiation of Pluripotent Stem Cells Using Specific Growth Factors and Small Molecules. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2018; 163:39-69. [PMID: 29071404 DOI: 10.1007/10_2017_30] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The envisioned routine application of human pluripotent stem cell (hPSC)-derived cardiomyocytes (CMs) for therapies and industry-compliant screening approaches will require efficient and highly reproducible processes for the mass production of well-characterized CM batches.On their way toward beating CMs, hPSCs initially undergo an epithelial-to-mesenchymal transition into a primitive-streak (PS)-like population that later gives rise to all endodermal and mesodermal lineages, including cardiovascular progenies (CVPs). CVPs are multipotent and possess the capability to give rise to all major cell types of the heart, including CMs, endothelial cells, cardiac fibroblasts, and smooth muscle cells. This article provides an historical overview and describes the stepwise development of protocols that typically result in the appearance of beating CMs within 7-12 days of hPSC differentiation.We describe the development of directed and closely controlled cardiomyogenic differentiation, which now enables the induction of >90% CM purity without further lineage enrichment. Although secreted lineage specifiers (revealed from developmental biology) were initially used, we outline the advantages of chemical pathway modulators, as defined by more recent screening approaches. Subsequently, we discuss the use of defined culture media for upscaling the production of hPSC-CMs in controlled bioreactors and how this, in principle, unlimited source of human CMs can be used to progress heart regeneration and stimulate the drug discovery pipeline. Graphical Abstract.
Collapse
|
5
|
Pane LS, Fulcoli FG, Cirino A, Altomonte A, Ferrentino R, Bilio M, Baldini A. Tbx1 represses Mef2c gene expression and is correlated with histone 3 deacetylation of the anterior heart field enhancer. Dis Model Mech 2018; 11:11/9/dmm029967. [PMID: 30166330 PMCID: PMC6176997 DOI: 10.1242/dmm.029967] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 07/09/2018] [Indexed: 12/19/2022] Open
Abstract
The TBX1 gene is haploinsufficient in 22q11.2 deletion syndrome (22q11.2DS), and genetic evidence from human patients and mouse models points to a major role of this gene in the pathogenesis of this syndrome. Tbx1 can activate and repress transcription, and previous work has shown that one of its functions is to negatively modulate cardiomyocyte differentiation. Tbx1 occupies the anterior heart field (AHF) enhancer of the Mef2c gene, which encodes a key cardiac differentiation transcription factor. Here, we show that increased dosage of Tbx1 correlates with downregulation of Mef2c expression and reduced acetylation of its AHF enhancer in cultured mouse myoblasts. Consistently, 22q11.2DS-derived and in vitro-differentiated human induced pluripotent stem cells (hiPSCs) expressed higher levels of MEF2C and showed increased AHF acetylation, compared with hiPSCs from a healthy donor. Most importantly, we show that in mouse embryos, loss of Tbx1 enhances the expression of the Mef2c-AHF-Cre transgene in a specific region of the splanchnic mesoderm, and in a dosage-dependent manner, providing an in vivo correlate of our cell culture data. These results indicate that Tbx1 regulates the Mef2c AHF enhancer by inducing histone deacetylation.
Collapse
Affiliation(s)
- Luna Simona Pane
- CNR Institute of Genetics and Biophysics Adriano Buzzati Traverso, Via Pietro Castellino 111, 80131 Napoli, Italy
| | - Filomena Gabriella Fulcoli
- CNR Institute of Genetics and Biophysics Adriano Buzzati Traverso, Via Pietro Castellino 111, 80131 Napoli, Italy
| | - Andrea Cirino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Napoli, Italy
| | - Alessandra Altomonte
- CNR Institute of Genetics and Biophysics Adriano Buzzati Traverso, Via Pietro Castellino 111, 80131 Napoli, Italy
| | - Rosa Ferrentino
- CNR Institute of Genetics and Biophysics Adriano Buzzati Traverso, Via Pietro Castellino 111, 80131 Napoli, Italy
| | - Marchesa Bilio
- CNR Institute of Genetics and Biophysics Adriano Buzzati Traverso, Via Pietro Castellino 111, 80131 Napoli, Italy
| | - Antonio Baldini
- CNR Institute of Genetics and Biophysics Adriano Buzzati Traverso, Via Pietro Castellino 111, 80131 Napoli, Italy .,Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Napoli, Italy
| |
Collapse
|
6
|
Zhu K, Wu Q, Ni C, Zhang P, Zhong Z, Wu Y, Wang Y, Xu Y, Kong M, Cheng H, Tao Z, Yang Q, Liang H, Jiang Y, Li Q, Zhao J, Huang J, Zhang F, Chen Q, Li Y, Chen J, Zhu W, Yu H, Zhang J, Yang HT, Hu X, Wang J. Lack of Remuscularization Following Transplantation of Human Embryonic Stem Cell-Derived Cardiovascular Progenitor Cells in Infarcted Nonhuman Primates. Circ Res 2018; 122:958-969. [PMID: 29343525 DOI: 10.1161/circresaha.117.311578] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 01/09/2018] [Accepted: 01/12/2018] [Indexed: 01/13/2023]
Abstract
RATIONALE Human pluripotent stem cell-derived cardiovascular progenitor cells (hPSC-CVPCs) should be thoroughly investigated in large animal studies before testing in clinical trials. OBJECTIVE The main of this study is to clarify whether hPSC-CVPCs can engraft for long time in the heart of primates after myocardial infarction (MI) and compare the effectiveness and safety of immunosuppression with cyclosporine alone or multiple-drug regimen (MDR) containing cyclosporine, methylprednisolone, and basiliximab in cynomolgus monkeys that had received intramyocardial injections of 1×107 EGFP (enhanced green fluorescent protein)-expressing hPSC-CVPCs after MI. A third group of animals received the immunosuppression MDR but without cell therapy after MI (MI+MDR group). METHODS AND RESULTS Measurements of EGFP gene levels and EGFP immunofluorescence staining indicated that the hPSC-CVPC engraftment rate was greater in the MI+MDR+CVPC group than that in the MI+cyclosporine+CVPC group. However, even in the MI+MDR+CVPC group, no transplanted cells could be detected at 140 days after transplantation. Concomitantly, immunofluorescent analysis of CD3, CD4, and CD8 expression indicated that T-lymphocyte infiltration in the CVPC-transplanted hearts was less in the MDR-treated animals than in the cyclosporine-alone-treated animals. The recovery of left ventricular function on day 28 post-MI in the MI+MDR+CVPC group was better than that in the MI+MDR group. Apoptotic cardiac cells were also less common in the MI+MDR+CVPC group than in the MI+MDR group, although both immunosuppression regimens were associated with transient hepatic dysfunction. CONCLUSIONS This is the largest study of hPSCs in nonhuman primates in cardiovascular field to date (n=32). Compared with cyclosporine alone, MDR attenuates immune rejection and improves survival of hPSC-CVPCs in primates; this is associated with less apoptosis of native cardiac cells and better recovery of left ventricular function at 28 days. However, even with MDR, transplanted hPSC-CVPCs do not engraft and do not survive at 140 days after transplantation, thereby excluding remuscularization as a mechanism for the functional effect.
Collapse
Affiliation(s)
- Keyang Zhu
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Qiang Wu
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Cheng Ni
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Peng Zhang
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Zhiwei Zhong
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Yan Wu
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Yingchao Wang
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Yinchuan Xu
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Minjian Kong
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Haifeng Cheng
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Zhihua Tao
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Qian Yang
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - He Liang
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Yun Jiang
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Qingju Li
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Jing Zhao
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Jijun Huang
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Fengjiang Zhang
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Qi Chen
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Yi Li
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Jinghai Chen
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Wei Zhu
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Hong Yu
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Jianyi Zhang
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Huang-Tian Yang
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang).
| | - Xinyang Hu
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang).
| | - Jian'an Wang
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang).
| |
Collapse
|
7
|
Skelton RJP, Kamp TJ, Elliott DA, Ardehali R. Biomarkers of Human Pluripotent Stem Cell-Derived Cardiac Lineages. Trends Mol Med 2017; 23:651-668. [PMID: 28576602 DOI: 10.1016/j.molmed.2017.05.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 04/24/2017] [Accepted: 05/04/2017] [Indexed: 02/07/2023]
Abstract
Human pluripotent stem cells (hPSCs) offer a practical source for the de novo generation of cardiac tissues and a unique opportunity to investigate cardiovascular lineage commitment. Numerous strategies have focused on the in vitro production of cardiomyocytes, smooth muscle, and endothelium from hPSCs. However, these differentiation protocols often yield undesired cell types. Thus, establishing a set of stage-specific markers for pure cardiac subpopulations will assist in defining the hierarchy of cardiac differentiation, aid in the development of cellular therapy, and facilitate drug screening and disease modeling. The recent characterization of many such markers is enabling the isolation of major cardiac lineages and subpopulations from differentiating hPSCs. We provide here a comprehensive review detailing the suite of biomarkers used to differentiate cardiac lineages from mixed hPSC-derived populations.
Collapse
Affiliation(s)
- Rhys J P Skelton
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA
| | - Timothy J Kamp
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - David A Elliott
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, Victoria 3052, Australia
| | - Reza Ardehali
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
8
|
Lee SH, Chen TY, Dhar SS, Gu B, Chen K, Kim YZ, Li W, Lee MG. A feedback loop comprising PRMT7 and miR-24-2 interplays with Oct4, Nanog, Klf4 and c-Myc to regulate stemness. Nucleic Acids Res 2016; 44:10603-10618. [PMID: 27625395 PMCID: PMC5159542 DOI: 10.1093/nar/gkw788] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 08/10/2016] [Accepted: 08/30/2016] [Indexed: 12/27/2022] Open
Abstract
Self-renewal and pluripotency are two fundamental characteristics of embryonic stem cells (ESCs) and are controlled by diverse regulatory factors, including pluripotent factors, epigenetic regulators and microRNAs (miRNAs). Although histone methyltransferases are key epigenetic regulators, whether and how a histone methyltransferase forms a network with miRNAs and the core pluripotent factor system to regulate ESC stemness is little known. Here, we show that the protein arginine methyltransferase 7 (PRMT7) is a pluripotent factor essential for the stemness of mouse ESCs. PRMT7 repressed the miR-24-2 gene encoding miR-24-3p and miR-24-2-5p by upregulating the levels of symmetrically dimethylated H4R3. Notably, miR-24-3p targeted the 3′ untranslated regions (UTRs) of the major pluripotent factors Oct4, Nanog, Klf4 and c-Myc, whereas miR-24-2-5p silenced Klf4 and c-Myc expression. miR-24-3p and miR-24-2-5p also targeted the 3′UTR of their repressor gene Prmt7. miR-24-3p and miR-24-2-5p induced mouse ESC differentiation, and their anti-sense inhibitors substantially reversed spontaneous differentiation of PRMT7-depleted mouse ESCs. Oct4, Nanog, Klf4 and c-Myc positively regulated Prmt7 expression. These findings define miR-24-3p and miR-24-2-5p as new anti-pluripotent miRNAs and also reveal a novel epigenetic stemness-regulatory mechanism in which a double-negative feedback loop consisting of PRMT7 and miR-24-3p/miR24-2-5p interplays with Oct4, Nanog, Klf4 and c-Myc to control ESC stemness.
Collapse
Affiliation(s)
- Sung-Hun Lee
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Tsai-Yu Chen
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.,Cancer Biology Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA
| | - Shilpa S Dhar
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Bingnan Gu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Kaifu Chen
- Institutefor Academic Medicine, The Methodist Hospital Research Institute, Houston, TX77030, USA.,Centerfor Cardiovascular Regeneration, Department of Cardiovascular Sciences, TheMethodist Hospital Research Institute, Houston, TX 77030, USA.,WeillCornell Medical College, Cornell University, New York, NY 10065, USA
| | - Young Zoon Kim
- Division of Neurooncology and Department of Neurosurgery, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, 158, Paryong-ro, Masan Hoiwon-Gu, Changwon, Gyeongsangnam-do, 630-723, Republic of Korea
| | - Wei Li
- Division of Biostatistics, Dan L. Duncan Cancer Center and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Min Gyu Lee
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA .,Cancer Biology Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA
| |
Collapse
|
9
|
Kempf H, Andree B, Zweigerdt R. Large-scale production of human pluripotent stem cell derived cardiomyocytes. Adv Drug Deliv Rev 2016; 96:18-30. [PMID: 26658242 DOI: 10.1016/j.addr.2015.11.016] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 11/19/2015] [Accepted: 11/25/2015] [Indexed: 12/20/2022]
Abstract
Regenerative medicine, including preclinical studies in large animal models and tissue engineering approaches as well as innovative assays for drug discovery, will require the constant supply of hPSC-derived cardiomyocytes and other functional progenies. Respective cell production processes must be robust, economically viable and ultimately GMP-compliant. Recent research has enabled transition of lab scale protocols for hPSC expansion and cardiomyogenic differentiation towards more controlled processing in industry-compatible culture platforms. Here, advanced strategies for the cultivation and differentiation of hPSCs will be reviewed by focusing on stirred bioreactor-based techniques for process upscaling. We will discuss how cardiomyocyte mass production might benefit from recent findings such as cell expansion at the cardiovascular progenitor state. Finally, remaining challenges will be highlighted, specifically regarding three dimensional (3D) hPSC suspension culture and critical safety issues ahead of clinical translation.
Collapse
Affiliation(s)
- Henning Kempf
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, Hannover Medical School, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Birgit Andree
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, Hannover Medical School, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, Hannover Medical School, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| |
Collapse
|
10
|
Thies RS, Murry CE. The advancement of human pluripotent stem cell-derived therapies into the clinic. Development 2016; 142:3077-84. [PMID: 26395136 DOI: 10.1242/dev.126482] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Human pluripotent stem cells (hPSCs) offer many potential applications for drug screening and 'disease in a dish' assay capabilities. However, a more ambitious goal is to develop cell therapeutics using hPSCs to generate and replace somatic cells that are lost as a result of disease or injury. This Spotlight article will describe the state of progress of some of the hPSC-derived therapeutics that offer the most promise for clinical use. Lessons from developmental biology have been instrumental in identifying signaling molecules that can guide these differentiation processes in vitro, and will be described in the context of these cell therapy programs.
Collapse
Affiliation(s)
- R Scott Thies
- Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Charles E Murry
- Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA Department of Pathology, University of Washington, Seattle, WA 98195, USA Department of Bioengineering, University of Washington, Seattle, WA 98195, USA Department of Medicine/Cardiology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
11
|
Levine RA, Hagége AA, Judge DP, Padala M, Dal-Bianco JP, Aikawa E, Beaudoin J, Bischoff J, Bouatia-Naji N, Bruneval P, Butcher JT, Carpentier A, Chaput M, Chester AH, Clusel C, Delling FN, Dietz HC, Dina C, Durst R, Fernandez-Friera L, Handschumacher MD, Jensen MO, Jeunemaitre XP, Le Marec H, Le Tourneau T, Markwald RR, Mérot J, Messas E, Milan DP, Neri T, Norris RA, Peal D, Perrocheau M, Probst V, Pucéat M, Rosenthal N, Solis J, Schott JJ, Schwammenthal E, Slaugenhaupt SA, Song JK, Yacoub MH. Mitral valve disease--morphology and mechanisms. Nat Rev Cardiol 2015; 12:689-710. [PMID: 26483167 DOI: 10.1038/nrcardio.2015.161] [Citation(s) in RCA: 260] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mitral valve disease is a frequent cause of heart failure and death. Emerging evidence indicates that the mitral valve is not a passive structure, but--even in adult life--remains dynamic and accessible for treatment. This concept motivates efforts to reduce the clinical progression of mitral valve disease through early detection and modification of underlying mechanisms. Discoveries of genetic mutations causing mitral valve elongation and prolapse have revealed that growth factor signalling and cell migration pathways are regulated by structural molecules in ways that can be modified to limit progression from developmental defects to valve degeneration with clinical complications. Mitral valve enlargement can determine left ventricular outflow tract obstruction in hypertrophic cardiomyopathy, and might be stimulated by potentially modifiable biological valvular-ventricular interactions. Mitral valve plasticity also allows adaptive growth in response to ventricular remodelling. However, adverse cellular and mechanobiological processes create relative leaflet deficiency in the ischaemic setting, leading to mitral regurgitation with increased heart failure and mortality. Our approach, which bridges clinicians and basic scientists, enables the correlation of observed disease with cellular and molecular mechanisms, leading to the discovery of new opportunities for improving the natural history of mitral valve disease.
Collapse
Affiliation(s)
- Robert A Levine
- Cardiac Ultrasound Laboratory, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Yawkey 5E, Boston, MA 02114, USA
| | - Albert A Hagége
- Hôpital Européen Georges Pompidou, Université René Descartes, UMR 970, Paris, France
| | | | | | - Jacob P Dal-Bianco
- Massachusetts General Hospital, Cardiac Ultrasound Laboratory, Harvard Medical School, Boston, MA, USA
| | | | | | | | - Nabila Bouatia-Naji
- Hôpital Européen Georges Pompidou, Université René Descartes, UMR 970, Paris, France
| | - Patrick Bruneval
- Hôpital Européen Georges Pompidou, Université René Descartes, UMR 970, Paris, France
| | | | - Alain Carpentier
- Hôpital Européen Georges Pompidou, Université René Descartes, UMR 970, Paris, France
| | | | | | | | - Francesca N Delling
- Beth Israel Deaconess Medical Centre, Harvard Medical School, Boston, MA, USA
| | | | - Christian Dina
- University of Nantes, Thoracic Institute, INSERM UMR 1097, CNRS UMR 6291, Nantes, France
| | - Ronen Durst
- Hadassah-Hebrew University Medical Centre, Jerusalem, Israel
| | - Leticia Fernandez-Friera
- Hospital Universitario HM Monteprincipe and the Centro Nacional de Investigaciones Cardiovasculares, Carlos III (CNIC), Madrid, Spain
| | - Mark D Handschumacher
- Massachusetts General Hospital, Cardiac Ultrasound Laboratory, Harvard Medical School, Boston, MA, USA
| | | | - Xavier P Jeunemaitre
- Hôpital Européen Georges Pompidou, Université René Descartes, UMR 970, Paris, France
| | - Hervé Le Marec
- University of Nantes, Thoracic Institute, INSERM UMR 1097, CNRS UMR 6291, Nantes, France
| | - Thierry Le Tourneau
- University of Nantes, Thoracic Institute, INSERM UMR 1097, CNRS UMR 6291, Nantes, France
| | | | - Jean Mérot
- University of Nantes, Thoracic Institute, INSERM UMR 1097, CNRS UMR 6291, Nantes, France
| | - Emmanuel Messas
- Hôpital Européen Georges Pompidou, Université René Descartes, UMR 970, Paris, France
| | - David P Milan
- Cardiovascular Research Center, Harvard Medical School, Boston, MA, USA
| | - Tui Neri
- Aix-Marseille University, INSERM UMR 910, Marseille, France
| | | | - David Peal
- Cardiovascular Research Center, Harvard Medical School, Boston, MA, USA
| | - Maelle Perrocheau
- Hôpital Européen Georges Pompidou, Université René Descartes, UMR 970, Paris, France
| | - Vincent Probst
- University of Nantes, Thoracic Institute, INSERM UMR 1097, CNRS UMR 6291, Nantes, France
| | - Michael Pucéat
- Aix-Marseille University, INSERM UMR 910, Marseille, France
| | | | - Jorge Solis
- Hospital Universitario HM Monteprincipe and the Centro Nacional de Investigaciones Cardiovasculares, Carlos III (CNIC), Madrid, Spain
| | - Jean-Jacques Schott
- University of Nantes, Thoracic Institute, INSERM UMR 1097, CNRS UMR 6291, Nantes, France
| | | | - Susan A Slaugenhaupt
- Center for Human Genetic Research, MGH Research Institute, Harvard Medical School, Boston, MA, USA
| | | | | | | |
Collapse
|
12
|
Cutts J, Nikkhah M, Brafman DA. Biomaterial Approaches for Stem Cell-Based Myocardial Tissue Engineering. Biomark Insights 2015; 10:77-90. [PMID: 26052226 PMCID: PMC4451817 DOI: 10.4137/bmi.s20313] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 05/05/2015] [Accepted: 05/06/2015] [Indexed: 12/21/2022] Open
Abstract
Adult and pluripotent stem cells represent a ready supply of cellular raw materials that can be used to generate the functionally mature cells needed to replace damaged or diseased heart tissue. However, the use of stem cells for cardiac regenerative therapies is limited by the low efficiency by which stem cells are differentiated in vitro to cardiac lineages as well as the inability to effectively deliver stem cells and their derivatives to regions of damaged myocardium. In this review, we discuss the various biomaterial-based approaches that are being implemented to direct stem cell fate both in vitro and in vivo. First, we discuss the stem cell types available for cardiac repair and the engineering of naturally and synthetically derived biomaterials to direct their in vitro differentiation to the cell types that comprise heart tissue. Next, we describe biomaterial-based approaches that are being implemented to enhance the in vivo integration and differentiation of stem cells delivered to areas of cardiac damage. Finally, we present emerging trends of using stem cell-based biomaterial approaches to deliver pro-survival factors and fully vascularized tissue to the damaged and diseased cardiac tissue.
Collapse
Affiliation(s)
- Josh Cutts
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - David A Brafman
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
13
|
Farouz Y, Chen Y, Terzic A, Menasché P. Concise Review: Growing Hearts in the Right Place: On the Design of Biomimetic Materials for Cardiac Stem Cell Differentiation. Stem Cells 2015; 33:1021-35. [DOI: 10.1002/stem.1929] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 11/10/2014] [Accepted: 12/01/2014] [Indexed: 12/25/2022]
Affiliation(s)
- Yohan Farouz
- Department of Chemistry, Paris Sciences et Lettres, Ecole Normale Supérieure de Paris; CNRS UMR; Paris France
- Sorbonne Paris Cité; Paris Descartes University; Paris France
- INSERM U970; Paris France
| | - Yong Chen
- Department of Chemistry, Paris Sciences et Lettres, Ecole Normale Supérieure de Paris; CNRS UMR; Paris France
| | | | - Philippe Menasché
- Sorbonne Paris Cité; Paris Descartes University; Paris France
- INSERM U970; Paris France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou; Department of Cardiovascular Surgery; Paris France
| |
Collapse
|
14
|
Jha R, Xu RH, Xu C. Efficient differentiation of cardiomyocytes from human pluripotent stem cells with growth factors. Methods Mol Biol 2015; 1299:115-131. [PMID: 25836579 PMCID: PMC4886660 DOI: 10.1007/978-1-4939-2572-8_9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Human pluripotent stem cells have tremendous replicative capacity and demonstrated potential to generate functional cardiomyocytes. These cardiomyocytes represent a promising source for cell replacement therapy to treat heart disease and may serve as a useful tool for drug discovery and disease modeling. Efficient cardiomyocyte differentiation, a prerequisite for the application of stem cell-derived cardiomyocytes, can be achieved with a growth factor-guided method. Undifferentiated cells are sequentially treated with activin A and BMP4 in a serum-free and insulin-free medium and then maintained in a serum-free medium with insulin. This method yields as much as >75% cardiomyocytes in the differentiation culture within 2 weeks, and the beating cardiomyocytes have expected molecular, cellular, and electrophysiological characteristics. In this chapter, we describe in detail the differentiation protocol and follow-up characterization focusing on immunocytochemistry, quantitative RT-PCR, and flow cytometry analysis.
Collapse
Affiliation(s)
- Rajneesh Jha
- Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Ren-He Xu
- Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Chunhui Xu
- Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, GA, USA
- The Parker H. Petit Institute for Bioengineering and Bioscience, Atlanta, GA, USA
| |
Collapse
|
15
|
Cao N, Liang H, Yang HT. Generation, expansion, and differentiation of cardiovascular progenitor cells from human pluripotent stem cells. Methods Mol Biol 2015; 1212:113-125. [PMID: 25208753 DOI: 10.1007/7651_2014_119] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Cardiovascular progenitor cells (CVPCs) derived from human embryonic stem cells and human induced pluripotent stem cells represent an invaluable potential source for the study of early embryonic cardiovascular development and stem cell-based therapies for congenital and acquired heart diseases. To fully realize their values, it is essential to establish an efficient and stable differentiation system for the induction of these pluripotent stem cells (PSCs) into the CVPCs and robustly expand them in culture, and then further differentiate these CVPCs into multiple cardiovascular cell types. Here we describe the protocols for efficient derivation, expansion, and differentiation of CVPCs from hPSCs in a chemically defined medium under feeder- and serum-free culture conditions.
Collapse
Affiliation(s)
- Nan Cao
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) & Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | | | | |
Collapse
|
16
|
Bellamy V, Vanneaux V, Bel A, Nemetalla H, Emmanuelle Boitard S, Farouz Y, Joanne P, Perier MC, Robidel E, Mandet C, Hagège A, Bruneval P, Larghero J, Agbulut O, Menasché P. Long-term functional benefits of human embryonic stem cell-derived cardiac progenitors embedded into a fibrin scaffold. J Heart Lung Transplant 2014; 34:1198-207. [PMID: 25534019 DOI: 10.1016/j.healun.2014.10.008] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Revised: 09/22/2014] [Accepted: 10/29/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Cardiac-committed cells and biomimetic scaffolds independently improve the therapeutic efficacy of stem cells. In this study we tested the long-term effects of their combination. METHODS Eighty immune-deficient rats underwent permanent coronary artery ligation. Five to 7 weeks later, those with an echocardiographically measured ejection fraction (EF) ≤55% were re-operated on and randomly allocated to receive a cell-free fibrin patch (n = 25), a fibrin patch loaded with 700,000 human embryonic stem cells (ESC) pre-treated to promote early cardiac differentiation (SSEA-1(+) progenitors [n = 30]), or to serve as sham-operated animals (n = 25). Left ventricular function was assessed by echocardiography at baseline and every month thereafter until 4 months. Hearts were then processed for assessment of fibrosis and angiogenesis and a 5-component heart failure score was constructed by integrating the absolute change in left ventricular end-systolic volume (LVESV) between 4 months and baseline, and the quantitative polymerase chain reaction (qPCR)-based expression of natriuretic peptides A and B, myosin heavy chain 7 and periostin. All data were recorded and analyzed in a blinded manner. RESULTS The cell-treated group consistently yielded better functional outcomes than the sham-operated group (p = 0.002 for EF; p = 0.01 for LVESV). Angiogenesis in the border zone was also significantly greater in the cell-fibrin group (p = 0.006), which yielded the lowest heart failure score (p = 0.04 vs sham). Engrafted progenitors were only detected shortly after transplantation; no grafted cells were identified after 4 months. There was no teratoma identified. CONCLUSIONS A fibrin scaffold loaded with ESC-derived cardiac progenitors resulted in sustained improvement in contractility and attenuation of remodeling without sustained donor cell engraftment. A paracrine effect, possibly on innate reparative responses, is a possible mechanism for this enduring effect.
Collapse
Affiliation(s)
- Valérie Bellamy
- INSERM U970, Hôpital Européen Georges Pompidou, Paris, France
| | - Valérie Vanneaux
- Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Cell Therapy Unit and Clinical Investigation Center in Biotherapies (CBT501), INSERM UMR1160, Université Sorbonne Paris Cité, Paris, France
| | - Alain Bel
- INSERM U970, Hôpital Européen Georges Pompidou, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Cardiovascular Surgery, Université Sorbonne Paris Cité, Paris, France
| | - Hany Nemetalla
- INSERM U970, Hôpital Européen Georges Pompidou, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Cardiology, Université Sorbonne Paris Cité, Paris, France
| | - Solène Emmanuelle Boitard
- Sorbonne Universités, UPMC Univ Paris 06, IBPS, UMR CNRS 8256, Biological Adaptation and Ageing, Paris, France
| | - Yohan Farouz
- INSERM U970, Hôpital Européen Georges Pompidou, Paris, France
| | - Pierre Joanne
- Sorbonne Universités, UPMC Univ Paris 06, IBPS, UMR CNRS 8256, Biological Adaptation and Ageing, Paris, France
| | | | - Estelle Robidel
- INSERM U970, Hôpital Européen Georges Pompidou, Paris, France
| | - Chantal Mandet
- INSERM U970, Hôpital Européen Georges Pompidou, Paris, France
| | - Albert Hagège
- INSERM U970, Hôpital Européen Georges Pompidou, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Cardiology, Université Sorbonne Paris Cité, Paris, France
| | - Patrick Bruneval
- INSERM U970, Hôpital Européen Georges Pompidou, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Pathology, Université Sorbonne Paris Cité, Paris, France
| | - Jérôme Larghero
- Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Cell Therapy Unit and Clinical Investigation Center in Biotherapies (CBT501), INSERM UMR1160, Université Sorbonne Paris Cité, Paris, France
| | - Onnik Agbulut
- Sorbonne Universités, UPMC Univ Paris 06, IBPS, UMR CNRS 8256, Biological Adaptation and Ageing, Paris, France
| | - Philippe Menasché
- INSERM U970, Hôpital Européen Georges Pompidou, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Cardiovascular Surgery, Université Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
17
|
Leschik J, Caron L, Yang H, Cowan C, Pucéat M. A view of bivalent epigenetic marks in two human embryonic stem cell lines reveals a different cardiogenic potential. Stem Cells Dev 2014; 24:384-92. [PMID: 25202820 DOI: 10.1089/scd.2014.0345] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Human embryonic stem (HUES) cells are derived from early individual embryos with unique genetic printing. However, how their epigenetic status might affect their potential to differentiate toward specific lineages remains a puzzling question. Using chromatin immunoprecipitation (ChIP)-polymerase chain reaction and ChIP-on-chip, the status of bivalent domains on gene promoters (ie, histone 3 on lysine 4 and histone 3 on lysine 27 trimethylation) was monitored for both undifferentiated and bone morphogenetic protein 2 (BMP2)-induced cardiac-committed cells. A marked difference in the epigenetic profile of HUES cell lines was observed and this was correlated to the pattern of gene expression induced by BMP2 as well as to their potential to generate cardiac progenitors and differentiated myocytes. Thus, the epigenetic H3trimeK4 and H3trimeK27 prints generating bivalent domains on promoters, could be used to predict a preference in their differentiation toward a specific lineage.
Collapse
Affiliation(s)
- Julia Leschik
- 1 INSERM UMR 633, Genopole Evry, University Paris V Descartes , Evry, France
| | | | | | | | | |
Collapse
|
18
|
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: 8.2] [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
|
19
|
Li Y, Higashiyama S, Shimakage M, Kawahara K, Yutsudo M, Watari A. Involvement of NANOG upregulation in malignant progression of human cells. DNA Cell Biol 2013; 32:104-10. [PMID: 23427894 DOI: 10.1089/dna.2012.1908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Previously, we isolated cell lines that display various degrees of transformed phenotypes from a single-cell population of human diploid fibroblasts (RB) containing a large deletion (13q14-22) in one copy of chromosome 13. They included a cell line transfected with SV40 early genes (RBSV), an immortalized cell line (RBI), an anchorage-independent cell line (RBS), and a tumorigenic cell line (RBT). Here, we analyzed gene expression profiles in these cell lines and showed that expression of some fibroblast-specified or mesenchyme-specified genes were downregulated, and those of stem cell-specified genes, including NANOG, were upregulated during malignant progression. When NANOG expression was knocked down with a short hairpin NANOG expression vector (shNANOG vector) in the RBS and RBT cells, the anchorage independency and tumorigenicity were repressed. We next examined various cancer cell lines for NANOG expression and showed that some cancer cell lines expressed a high level of normal and/or variant NANOG proteins. Overexpression of NANOG mRNA in lung adenocarcinoma was also shown by in situ hybridization. All these data indicate the involvement of NANOG in tumorigenesis.
Collapse
Affiliation(s)
- Yang Li
- Research Institute for Microbial Diseases, Osaka University, Yamadaoka, Suita, Osaka, Japan
| | | | | | | | | | | |
Collapse
|
20
|
Calderon D, Planat-Benard V, Bellamy V, Vanneaux V, Kuhn C, Peyrard S, Larghero J, Desnos M, Casteilla L, Pucéat M, Menasché P, Chatenoud L. Immune response to human embryonic stem cell-derived cardiac progenitors and adipose-derived stromal cells. J Cell Mol Med 2012; 16:1544-52. [PMID: 21895965 PMCID: PMC3823222 DOI: 10.1111/j.1582-4934.2011.01435.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Transplantation of allogeneic human embryonic stem cell-derived cardiac progenitors triggers an immune response. We assessed whether this response could be modulated by the concomitant use of adipose-derived stromal cells (ADSC). Peripheral blood mononuclear cells were collected from 40 patients with coronary artery disease (CAD) and nine healthy controls. Cardiac progenitors (CD15+ Mesp1+) were generated as already reported from the I6 cell line treated with bone morphogenetic protein (BMP)-2. Adipose-derived stromal cells were obtained from abdominal dermolipectomies. We assessed the proliferative response of peripheral lymphocytes from patients and controls to cardiac progenitors cultured on a monolayer of ADSC, to allogeneic lymphocytes in mixed lymphocyte culture and to the T cell mitogen phytohemaglutin A in presence or absence of ADSC. Cardiac progenitors cultured on a monolayer of ADSC triggered a proliferation of lymphocytes from both patients and controls albeit lower than that induced by allogeneic lymphocytes. When cultured alone, ADSC did not induce any proliferation of allogeneic lymphocytes. When added to cultures of lymphocytes, ADSC significantly inhibited the alloantigen or mitogen-induced proliferative response. Compared to healthy controls, lymphocytes from patients presenting CAD expressed a decreased proliferative capacity, in particular to mitogen-induced stimulation. Adipose-derived stromal cells express an immunomodulatory effect that limits both alloantigen and mitogen-induced lymphocyte responses. Furthermore, lymphocytes from patients with CAD are low responders to conventional stimuli, possibly because of their age and disease-associated treatment regimens. We propose that, in combination, these factors may limit the in vivo immunogenicity of cardiac progenitors co-implanted with ADSC in patients with CAD.
Collapse
Affiliation(s)
- Damelys Calderon
- INSERM UMR 633, Laboratory of Surgical Research, Höopital Européen Georges Pompidou, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Embryological origin of the endocardium and derived valve progenitor cells: from developmental biology to stem cell-based valve repair. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1833:917-22. [PMID: 23078978 DOI: 10.1016/j.bbamcr.2012.09.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 09/26/2012] [Accepted: 09/29/2012] [Indexed: 11/23/2022]
Abstract
The cardiac valves are targets of both congenital and acquired diseases. The formation of valves during embryogenesis (i.e., valvulogenesis) originates from endocardial cells lining the myocardium. These cells undergo an endothelial-mesenchymal transition, proliferate and migrate within an extracellular matrix. This leads to the formation of bilateral cardiac cushions in both the atrioventricular canal and the outflow tract. The embryonic origin of both the endocardium and prospective valve cells is still elusive. Endocardial and myocardial lineages are segregated early during embryogenesis and such a cell fate decision can be recapitulated in vitro by embryonic stem cells (ESC). Besides genetically modified mice and ex vivo heart explants, ESCs provide a cellular model to study the early steps of valve development and might constitute a human therapeutic cell source for decellularized tissue-engineered valves. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Cardiac Pathways of Differentiation, Metabolism and Contraction.
Collapse
|
22
|
Blazeski A, Zhu R, Hunter DW, Weinberg SH, Boheler KR, Zambidis ET, Tung L. Electrophysiological and contractile function of cardiomyocytes derived from human embryonic stem cells. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2012; 110:178-95. [PMID: 22958937 DOI: 10.1016/j.pbiomolbio.2012.07.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 07/30/2012] [Indexed: 12/23/2022]
Abstract
Human embryonic stem cells have emerged as the prototypical source from which cardiomyocytes can be derived for use in drug discovery and cell therapy. However, such applications require that these cardiomyocytes (hESC-CMs) faithfully recapitulate the physiology of adult cells, especially in relation to their electrophysiological and contractile function. We review what is known about the electrophysiology of hESC-CMs in terms of beating rate, action potential characteristics, ionic currents, and cellular coupling as well as their contractility in terms of calcium cycling and contraction. We also discuss the heterogeneity in cellular phenotypes that arises from variability in cardiac differentiation, maturation, and culture conditions, and summarize present strategies that have been implemented to reduce this heterogeneity. Finally, we present original electrophysiological data from optical maps of hESC-CM clusters.
Collapse
Affiliation(s)
- Adriana Blazeski
- Department of Biomedical Engineering, The Johns Hopkins University, 720 Rutland Ave., Baltimore, MD 21205, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Xu C. Differentiation and enrichment of cardiomyocytes from human pluripotent stem cells. J Mol Cell Cardiol 2012; 52:1203-12. [PMID: 22484618 DOI: 10.1016/j.yjmcc.2012.03.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 03/15/2012] [Accepted: 03/20/2012] [Indexed: 12/20/2022]
Abstract
Human cardiomyocytes derived from pluripotent stem cells hold great promise for cardiac cell therapy, disease modeling, drug discovery, and the study of developmental biology. Reaching these potentials fully requires the development of methods that enable efficient and robust generation of cardiomyocytes with expected characteristics. This review summarizes and discusses up-to-date methods that have been used to derive and enrich human cardiomyocytes from pluripotent stem cells, provides a brief overview of in vitro and in vivo characterization of these cardiomyocytes, and considers future advancement needed to further harness the power of these cells.
Collapse
Affiliation(s)
- Chunhui Xu
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
24
|
Lentiviral vectors and cardiovascular diseases: a genetic tool for manipulating cardiomyocyte differentiation and function. Gene Ther 2012; 19:642-8. [DOI: 10.1038/gt.2012.19] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
25
|
Young DA, DeQuach JA, Christman KL. Human cardiomyogenesis and the need for systems biology analysis. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2011; 3:666-80. [PMID: 21197666 PMCID: PMC3282989 DOI: 10.1002/wsbm.141] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease remains the leading cause of death in the Western world and myocardial infarction is one of the primary facets of this disease. The limited natural self-renewal of cardiac muscle following injury and restricted supply of heart transplants has encouraged researchers to investigate other means to stimulate regeneration of damaged myocardium. The plasticity of stem cells toward multiple lineages offers the potential to repair the heart following injury. Embryonic stem cells have been extensively studied for their ability to differentiate into early cardiomyocytes, however, the pathway has only been partially defined and inadequate efficiency limits their clinical applicability. Some studies have shown cardiomyogenesis from adult mesenchymal stem cells, from both bone marrow and adipose tissue, but their differentiation pathway remains poorly detailed and these results remain controversial. Despite promising results using stem cells in animal models of cardiac injury, the driving mechanisms behind their differentiation down a cardiomyogenic pathway have yet to be determined. Currently, there is a paucity of information regarding cardiomyogenesis on the systemic level. Stem cell differentiation results from multiple signaling parameters operating in a tightly regulated spatiotemporal pattern. Investigating this phenomenon from a systems biology perspective could unveil the abstruse mechanisms controlling cardiomyogenesis that would otherwise require extensive in vitro testing.
Collapse
Affiliation(s)
- D Adam Young
- Department of Bioengineering, University of California, San Diego, CA, USA
| | | | | |
Collapse
|
26
|
Egashira T, Yuasa S, Fukuda K. Induced pluripotent stem cells in cardiovascular medicine. Stem Cells Int 2011; 2011:348960. [PMID: 21977041 PMCID: PMC3184500 DOI: 10.4061/2011/348960] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 05/25/2011] [Accepted: 07/18/2011] [Indexed: 01/31/2023] Open
Abstract
Induced pluripotent stem (iPS) cells are generated by reprogramming human somatic cells through the forced expression of several embryonic stem (ES) cell-specific transcription factors. The potential of iPS cells is having a significant impact on regenerative medicine, with the promise of infinite self-renewal, differentiation into multiple cell types, and no problems concerning ethics or immunological rejection. Human iPS cells are currently generated by transgene introduction principally through viral vectors, which integrate into host genomes, although the associated risk of tumorigenesis is driving research into nonintegration methods. Techniques for pluripotent stem cell differentiation and purification to yield cardiomyocytes are also advancing constantly. Although there remain some unsolved problems, cardiomyocyte transplantation may be a reality in the future. After those problems will be solved, applications of human iPS cells in human cardiovascular regenerative medicine will be envisaged for the future. Furthermore, iPS cell technology has generated new human disease models using disease-specific cells. This paper summarizes the progress of iPS cell technology in cardiovascular research.
Collapse
Affiliation(s)
- Toru Egashira
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | | | | |
Collapse
|
27
|
Gonzales C, Ullrich ND, Gerber S, Berthonneche C, Niggli E, Pedrazzini T. Isolation of cardiovascular precursor cells from the human fetal heart. Tissue Eng Part A 2011; 18:198-207. [PMID: 21902604 DOI: 10.1089/ten.tea.2011.0022] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Weakening of cardiac function in patients with heart failure results from a loss of cardiomyocytes in the damaged heart. Cell replacement therapies as a way to induce myocardial regeneration in humans could represent attractive alternatives to classical drug-based approaches. However, a suitable source of precursor cells, which could produce a functional myocardium after transplantation, remains to be identified. In the present study, we isolated cardiovascular precursor cells from ventricles of human fetal hearts at 12 weeks of gestation. These cells expressed Nkx2.5 but not late cardiac markers such as α-actinin and troponin I. In addition, proliferating cells expressed the mesenchymal stem cell markers CD73, CD90, and CD105. Evidence for functional cardiogenic differentiation in vitro was demonstrated by the upregulation of cardiac gene expression as well as the appearance of cells with organized sarcomeric structures. Importantly, differentiated cells presented spontaneous and triggered calcium signals. Differentiation into smooth muscle cells was also detected. In contrast, precursor cells did not produce endothelial cells. The engraftment and differentiation capacity of green fluorescent protein (GFP)-labeled cardiac precursor cells were then tested in vivo after transfer into the heart of immunodeficient severe combined immunodeficient mice. Engrafted human cells were readily detected in the mouse myocardium. These cells retained their cardiac commitment and differentiated into α-actinin-positive cardiomyocytes. Expression of connexin-43 at the interface between GFP-labeled and endogenous cardiomyocytes indicated that precursor-derived cells connected to the mouse myocardium. Together, these results suggest that human ventricular nonmyocyte cells isolated from fetal hearts represent a suitable source of precursors for cell replacement therapies.
Collapse
Affiliation(s)
- Christine Gonzales
- Experimental Cardiology Unit, Department of Medicine, University of Lausanne Medical School, Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
28
|
Ren Y, Lee MY, Schliffke S, Paavola J, Amos PJ, Ge X, Ye M, Zhu S, Senyei G, Lum L, Ehrlich BE, Qyang Y. Small molecule Wnt inhibitors enhance the efficiency of BMP-4-directed cardiac differentiation of human pluripotent stem cells. J Mol Cell Cardiol 2011; 51:280-7. [PMID: 21569778 PMCID: PMC3334336 DOI: 10.1016/j.yjmcc.2011.04.012] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Revised: 03/31/2011] [Accepted: 04/26/2011] [Indexed: 01/25/2023]
Abstract
Human induced pluripotent stem (iPS) cells potentially provide a unique resource for generating patient-specific cardiomyocytes to study cardiac disease mechanisms and treatments. However, existing approaches to cardiomyocyte production from human iPS cells are inefficient, limiting the application of iPS cells in basic and translational cardiac research. Furthermore, strategies to accurately record changes in iPS cell-derived cardiomyocyte action potential duration (APD) are needed to monitor APD-related cardiac disease and for rapid drug screening. We examined whether modulation of the bone morphogenetic protein 4 (BMP-4) and Wnt/β-catenin signaling pathways could induce efficient cardiac differentiation of human iPS cells. We found that early treatment of human iPS cells with BMP-4 followed by late treatment with small molecule Wnt inhibitors led to a marked increase in production of cardiomyocytes compared to existing differentiation strategies. Using immunocytochemical staining and real-time intracellular calcium imaging, we showed that these induced cardiomyocytes expressed typical sarcomeric markers, exhibited normal rhythmic Ca(2+) transients, and responded to both β-adrenergic and electric stimulation. Furthermore, human iPS cell-derived cardiomyocytes demonstrated characteristic changes in action potential duration in response to cardioactive drugs procainamide and verapamil using voltage-sensitive dye-based optical recording. Thus, modulation of the BMP-4 and Wnt signaling pathways in human iPS cells leads to highly efficient production of cardiomyocytes with typical electrophysiological function and pharmacologic responsiveness. The use of human iPS cell-derived cardiomyocytes and the application of calcium- and voltage-sensitive dyes for the direct, rapid measurement of iPS cell-derived cardiomyocyte activity promise to offer attractive platforms for studying cardiac disease mechanisms and therapeutics.
Collapse
Affiliation(s)
- Yongming Ren
- Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06520
| | - Min Young Lee
- Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06520
| | - Simon Schliffke
- Department of Pharmacology, School of Medicine, Yale University, New Haven, CT 06520
- Department of Anatomy II: Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jere Paavola
- Department of Pharmacology, School of Medicine, Yale University, New Haven, CT 06520
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2U, Tukholmankatu 8, 00290, Helsinki, Finland
| | - Peter J. Amos
- Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06520
| | - Xin Ge
- Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06520
| | - Mingyu Ye
- Department of Cellular and Molecular Physiology, School of Medicine, Yale University, New Haven, CT 06520
| | - Shenjun Zhu
- Cardiovascular Research Center, Massachusetts General Hospital, 149 Thirteenth Street, Charlestown, MA 02129
| | - Grant Senyei
- Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06520
| | - Lawrence Lum
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390
| | | | - Yibing Qyang
- Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06520
- Yale Stem Cell Center, Yale University, New Haven, CT 06520
| |
Collapse
|
29
|
Xu C, Police S, Hassanipour M, Li Y, Chen Y, Priest C, O'Sullivan C, Laflamme MA, Zhu WZ, Van Biber B, Hegerova L, Yang J, Delavan-Boorsma K, Davies A, Lebkowski J, Gold JD. Efficient generation and cryopreservation of cardiomyocytes derived from human embryonic stem cells. Regen Med 2011; 6:53-66. [PMID: 21175287 DOI: 10.2217/rme.10.91] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
AIM Human embryonic stem cells (hESCs) represent a novel cell source to treat diseases such as heart failure and for use in drug screening. In this study, we aim to promote efficient generation of cardiomyocytes from hESCs by combining the current optimal techniques of controlled growth of undifferentiated cells and specific induction for cardiac differentiation. We also aim to examine whether these methods are scalable and whether the differentiated cells can be cryopreserved. METHODS & RESULTS hESCs were maintained without conditioned medium or feeders and were sequentially treated with activin A and bone morphogenetic protein-4 in a serum-free medium. This led to differentiation into cell populations containing high percentages of cardiomyocytes. The differentiated cells expressed appropriate cardiomyocyte markers and maintained contractility in culture, and the majority of the cells displayed working chamber (atrial and ventricular) type electrophysiological properties. In addition, the cell growth and differentiation process was adaptable to large culture formats. Moreover, the cardiomyocytes survived following cryopreservation, and viable cardiac grafts were detected after transplantation of cryopreserved cells into rat hearts following myocardial infarctions. CONCLUSION These results demonstrate that cardiomyocytes of high quality can be efficiently generated and cryopreserved using hESCs maintained in serum-free medium, a step forward towards the application of these cells to human clinical use or drug discovery.
Collapse
Affiliation(s)
- Chunhui Xu
- Geron Corporation, Menlo Park, CA 94025, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Rajala K, Pekkanen-Mattila M, Aalto-Setälä K. Cardiac differentiation of pluripotent stem cells. Stem Cells Int 2011; 2011:383709. [PMID: 21603143 PMCID: PMC3096314 DOI: 10.4061/2011/383709] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 02/01/2011] [Accepted: 02/08/2011] [Indexed: 01/12/2023] Open
Abstract
The ability of human pluripotent stem cells to differentiate towards the cardiac lineage has attracted significant interest, initially with a strong focus on regenerative medicine. The ultimate goal to repair the heart by cardiomyocyte replacement has, however, proven challenging. Human cardiac differentiation has been difficult to control, but methods are improving, and the process, to a certain extent, can be manipulated and directed. The stem cell-derived cardiomyocytes described to date exhibit rather immature functional and structural characteristics compared to adult cardiomyocytes. Thus, a future challenge will be to develop strategies to reach a higher degree of cardiomyocyte maturation in vitro, to isolate cardiomyocytes from the heterogeneous pool of differentiating cells, as well as to guide the differentiation into the desired subtype, that is, ventricular, atrial, and pacemaker cells. In this paper, we will discuss the strategies for the generation of cardiomyocytes from pluripotent stem cells and their characteristics, as well as highlight some applications for the cells.
Collapse
Affiliation(s)
- Kristiina Rajala
- Regea - Institute for Regenerative Medicine, University of Tampere, Tampere University Hospital, 33520 Tampere, Finland
| | | | | |
Collapse
|
31
|
Yuan SH, Martin J, Elia J, Flippin J, Paramban RI, Hefferan MP, Vidal JG, Mu Y, Killian RL, Israel MA, Emre N, Marsala S, Marsala M, Gage FH, Goldstein LSB, Carson CT. Cell-surface marker signatures for the isolation of neural stem cells, glia and neurons derived from human pluripotent stem cells. PLoS One 2011; 6:e17540. [PMID: 21407814 PMCID: PMC3047583 DOI: 10.1371/journal.pone.0017540] [Citation(s) in RCA: 281] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Accepted: 02/08/2011] [Indexed: 12/22/2022] Open
Abstract
Background Neural induction of human pluripotent stem cells often yields heterogeneous cell populations that can hamper quantitative and comparative analyses. There is a need for improved differentiation and enrichment procedures that generate highly pure populations of neural stem cells (NSC), glia and neurons. One way to address this problem is to identify cell-surface signatures that enable the isolation of these cell types from heterogeneous cell populations by fluorescence activated cell sorting (FACS). Methodology/Principal Findings We performed an unbiased FACS- and image-based immunophenotyping analysis using 190 antibodies to cell surface markers on naïve human embryonic stem cells (hESC) and cell derivatives from neural differentiation cultures. From this analysis we identified prospective cell surface signatures for the isolation of NSC, glia and neurons. We isolated a population of NSC that was CD184+/CD271−/CD44−/CD24+ from neural induction cultures of hESC and human induced pluripotent stem cells (hiPSC). Sorted NSC could be propagated for many passages and could differentiate to mixed cultures of neurons and glia in vitro and in vivo. A population of neurons that was CD184−/CD44−/CD15LOW/CD24+ and a population of glia that was CD184+/CD44+ were subsequently purified from cultures of differentiating NSC. Purified neurons were viable, expressed mature and subtype-specific neuronal markers, and could fire action potentials. Purified glia were mitotic and could mature to GFAP-expressing astrocytes in vitro and in vivo. Conclusions/Significance These findings illustrate the utility of immunophenotyping screens for the identification of cell surface signatures of neural cells derived from human pluripotent stem cells. These signatures can be used for isolating highly pure populations of viable NSC, glia and neurons by FACS. The methods described here will enable downstream studies that require consistent and defined neural cell populations.
Collapse
Affiliation(s)
- Shauna H. Yuan
- Howard Hughes Medical Institute and Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Jody Martin
- BD Biosciences, La Jolla, California, United States of America
| | - Jeanne Elia
- BD Biosciences, La Jolla, California, United States of America
| | - Jessica Flippin
- Howard Hughes Medical Institute and Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | | | - Mike P. Hefferan
- Anesthesiology Research Laboratory, Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
| | - Jason G. Vidal
- BD Biosciences, La Jolla, California, United States of America
| | - Yangling Mu
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Rhiannon L. Killian
- Howard Hughes Medical Institute and Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, California, United States of America
| | - Mason A. Israel
- Howard Hughes Medical Institute and Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, California, United States of America
| | - Nil Emre
- BD Biosciences, La Jolla, California, United States of America
| | - Silvia Marsala
- Anesthesiology Research Laboratory, Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
| | - Martin Marsala
- Anesthesiology Research Laboratory, Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
- Institute of Neurobiology, Slovak Academy of Sciences, Košice, Slovakia
| | - Fred H. Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Lawrence S. B. Goldstein
- Howard Hughes Medical Institute and Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | | |
Collapse
|
32
|
Cardiac regeneration: still a 21st century challenge in search for cardiac progenitors from stem cells and embryos. J Cardiovasc Pharmacol 2010; 56:16-21. [PMID: 20631550 DOI: 10.1097/fjc.0b013e3181d8bc6d] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Regeneration of the heart after a stroke would be the best biologic response to restore its function. However, although this phenomenon occurs in primitive organisms, the regenerative potential is lost in mammals. Thus, the search for an appropriate cardiac progenitor with the potential to differentiate into a functional cardiomyocyte in vitro and in vivo has been the subject of intensive investigation. We summarize the cardiogenic transcriptional pathway that constitutes the molecular scaffold to drive pluripotent stem cells toward a cardiac progenitor fate. Then we overview the literature on derivation of cardiac progenitors from both embryos and stem cells.
Collapse
|
33
|
Kamata M, Liu S, Liang M, Nagaoka Y, Chen ISY. Generation of human induced pluripotent stem cells bearing an anti-HIV transgene by a lentiviral vector carrying an internal murine leukemia virus promoter. Hum Gene Ther 2010; 21:1555-67. [PMID: 20524893 DOI: 10.1089/hum.2010.050] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The recent development of induced pluripotent stem cells (iPSCs) by ectopic expression of defined reprogramming factors offers enormous therapeutic opportunity. To deliver these factors, murine leukemia virus (MLV)-based vectors have been broadly used in the setting of hematopoietic stem cell transplantation. However, MLV vectors have been implicated in malignancy induced by insertional mutagenesis, whereas lentiviral vectors have not. Furthermore, the infectivity of MLV vectors is limited to dividing cells, whereas lentiviral vectors can also transduce nondividing cells. One important characteristic of MLV vectors is a self-silencing property of the promoter element in pluripotent stem cells, allowing temporal transgene expression in a nonpluripotent state before iPSC derivation. Here we test iPSC generation using a novel chimeric vector carrying a mutant MLV promoter internal to a lentiviral vector backbone, thereby containing the useful properties of both types of vectors. Transgene expression of this chimeric vector was highly efficient compared with that of MLV vectors and was silenced specifically in human embryonic stem cells. Human fetal fibroblasts transduced with the vector encoding each factor were efficiently reprogrammed into a pluripotent state, and these iPSCs had potential to differentiate into a variety of cell types. To explore the possibility of iPSCs for gene therapy, we established iPSC clones expressing a short hairpin RNA (shRNA) targeting chemokine receptor 5 (CCR5), the main coreceptor for HIV-1. Using a reporter construct for CCR5 expression, we confirmed that CCR5 shRNA was expressed and specifically knocked down the reporter expression in iPSCs. These data indicate that our chimeric lentiviral vector is a valuable tool for generation of iPSCs and the combination with vectors encoding transgenes allows for rapid establishment of desired genetically engineered iPSC lines.
Collapse
Affiliation(s)
- Masakazu Kamata
- Department of Microbiology, Immunology and Molecular Genetics, University of California at Los Angeles, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | | | | | | | | |
Collapse
|
34
|
Bel A, Planat-Bernard V, Saito A, Bonnevie L, Bellamy V, Sabbah L, Bellabas L, Brinon B, Vanneaux V, Pradeau P, Peyrard S, Larghero J, Pouly J, Binder P, Garcia S, Shimizu T, Sawa Y, Okano T, Bruneval P, Desnos M, Hagège AA, Casteilla L, Pucéat M, Menasché P. Composite cell sheets: a further step toward safe and effective myocardial regeneration by cardiac progenitors derived from embryonic stem cells. Circulation 2010; 122:S118-23. [PMID: 20837902 DOI: 10.1161/circulationaha.109.927293] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND The safety and efficacy of myocardial regeneration using embryonic stem cells are limited by the risk of teratoma and the high rate of cell death. METHODS AND RESULTS To address these issues, we developed a composite construct made of a sheet of adipose tissue-derived stroma cells and embryonic stem cell-derived cardiac progenitors. Ten Rhesus monkeys underwent a transient coronary artery occlusion followed, 2 weeks later, by the open-chest delivery of the composite cell sheet over the infarcted area or a sham operation. The sheet was made of adipose tissue-derived stroma cells grown from a biopsy of autologous adipose tissue and cultured onto temperature-responsive dishes. Allogeneic Rhesus embryonic stem cells were committed to a cardiac lineage and immunomagnetically sorted to yield SSEA-1(+) cardiac progenitors, which were then deposited onto the cell sheet. Cyclosporine was given for 2 months until the animals were euthanized. Preimplantation studies showed that the SSEA-1(+) progenitors expressed cardiac markers and had lost pluripotency. After 2 months, there was no teratoma in any of the 5 cell-treated monkeys. Analysis of >1500 histological sections showed that the SSEA-1(+) cardiac progenitors had differentiated into cardiomyocytes, as evidenced by immunofluorescence and real-time polymerase chain reaction. There were also a robust engraftment of autologous adipose tissue-derived stroma cells and increased angiogenesis compared with the sham animals. CONCLUSIONS These data collected in a clinically relevant nonhuman primate model show that developmentally restricted SSEA-1(+) cardiac progenitors appear to be safe and highlight the benefit of the epicardial delivery of a construct harboring cells with a cardiomyogenic differentiation potential and cells providing them the necessary trophic support.
Collapse
Affiliation(s)
- Alain Bel
- Assistance Publique-Hôpitaux de Paris, Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, University Paris Descartes, INSERM U633, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
DeQuach JA, Mezzano V, Miglani A, Lange S, Keller GM, Sheikh F, Christman KL. Simple and high yielding method for preparing tissue specific extracellular matrix coatings for cell culture. PLoS One 2010; 5:e13039. [PMID: 20885963 PMCID: PMC2946408 DOI: 10.1371/journal.pone.0013039] [Citation(s) in RCA: 192] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2010] [Accepted: 09/02/2010] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The native extracellular matrix (ECM) consists of a highly complex, tissue-specific network of proteins and polysaccharides, which help regulate many cellular functions. Despite the complex nature of the ECM, in vitro cell-based studies traditionally assess cell behavior on single ECM component substrates, which do not adequately mimic the in vivo extracellular milieu. METHODOLOGY/PRINCIPAL FINDINGS We present a simple approach for developing naturally derived ECM coatings for cell culture that provide important tissue-specific cues unlike traditional cell culture coatings, thereby enabling the maturation of committed C2C12 skeletal myoblast progenitors and human embryonic stem cells differentiated into cardiomyocytes. Here we show that natural muscle-specific coatings can (i) be derived from decellularized, solubilized adult porcine muscle, (ii) contain a complex mixture of ECM components including polysaccharides, (iii) adsorb onto tissue culture plastic and (iv) promote cell maturation of committed muscle progenitor and stem cells. CONCLUSIONS This versatile method can create tissue-specific ECM coatings, which offer a promising platform for cell culture to more closely mimic the mature in vivo ECM microenvironment.
Collapse
Affiliation(s)
- Jessica A. DeQuach
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - Valeria Mezzano
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Amar Miglani
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - Stephan Lange
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Gordon M. Keller
- McEwen Centre for Regenerative Medicine, University Health Network, Toronto, Ontario, Canada
| | - Farah Sheikh
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Karen L. Christman
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
36
|
Dick E, Rajamohan D, Ronksley J, Denning C. Evaluating the utility of cardiomyocytes from human pluripotent stem cells for drug screening. Biochem Soc Trans 2010; 38:1037-45. [PMID: 20659000 DOI: 10.1042/bst0381037] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Functional cardiomyocytes can now be derived routinely from hPSCs (human pluripotent stem cells), which collectively include embryonic and induced pluripotent stem cells. This technology presents new opportunities to develop pharmacologically relevant in vitro screens to detect cardiotoxicity, with a view to improving patient safety while reducing the economic burden to industry arising from high drug attrition rates. In the present article, we consider the need for human cardiomyocytes in drug-screening campaigns and review the strategies used to differentiate hPSCs towards the cardiac lineage. During early stages of differentiation, hPSC-cardiomyocytes display gene expression profiles, ultra-structures, ion channel functionality and pharmacological responses reminiscent of an embryonic phenotype, but maturation during extended time in culture has been demonstrated convincingly. Notably, hPSC-cardiomyocytes have been shown to respond in a highly predictable manner to over 40 compounds that have a known pharmacological effect on the human heart. This suggests that further development and validation of the hPSC-cardiomyocyte model as a tool for assessing cardiotoxicity is warranted.
Collapse
Affiliation(s)
- Emily Dick
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | | | | | | |
Collapse
|
37
|
Schussler O, Chachques JC, Mesana TG, Suuronen EJ, Lecarpentier Y, Ruel M. 3-dimensional structures to enhance cell therapy and engineer contractile tissue. Asian Cardiovasc Thorac Ann 2010; 18:188-98. [PMID: 20304859 DOI: 10.1177/0218492310361531] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Experimental studies in animals and recent human clinical trials have revealed the current limitations of cellular transplantation, which include poor cell survival, lack of cell engraftment, and poor differentiation. Evidence in animals suggests that use of a 3-dimensional scaffold may enhance cell therapy and engineer myocardial tissue by improving initial cell retention, survival, differentiation, and integration. Several scaffolds of synthetic or natural origin are under development. Until now, contractility has been demonstrated in vitro only in biological scaffolds prepared from decellularized organs or tissue, or in collagenic porous scaffold obtained by crosslinking collagen fibers. While contractility of a cellularized collagen construct is poor, it can be greatly enhanced by tumor basement membrane extract. Recent advances in biochemistry have shown improved cell-matrix interactions by coupling adhesion molecules to achieve an efficient and safe bioartificial myocardium with no tumoral component. Fixation of adhesion molecules may also be a way to enhance cell homing and/or differentiation to increase local angiogenesis. Whatever the clinically successful combination ultimately proves to be, it is likely that cell therapy will require providing a supportive biochemical, physical, and spatial environment that will allow the cells to optimally differentiate and integrate within the target myocardial tissue.
Collapse
Affiliation(s)
- Olivier Schussler
- Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Suite 3403, Ottawa, ON, K1Y 4W7, Canada.
| | | | | | | | | | | |
Collapse
|
38
|
Chiriac A, Nelson TJ, Faustino RS, Behfar A, Terzic A. Cardiogenic induction of pluripotent stem cells streamlined through a conserved SDF-1/VEGF/BMP2 integrated network. PLoS One 2010; 5:e9943. [PMID: 20376342 PMCID: PMC2848581 DOI: 10.1371/journal.pone.0009943] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2009] [Accepted: 02/22/2010] [Indexed: 12/28/2022] Open
Abstract
Background Pluripotent stem cells produce tissue-specific lineages through programmed acquisition of sequential gene expression patterns that function as a blueprint for organ formation. As embryonic stem cells respond concomitantly to diverse signaling pathways during differentiation, extraction of a pro-cardiogenic network would offer a roadmap to streamline cardiac progenitor output. Methods and Results To resolve gene ontology priorities within precursor transcriptomes, cardiogenic subpopulations were here generated according to either growth factor guidance or stage-specific biomarker sorting. Innate expression profiles were independently delineated through unbiased systems biology mapping, and cross-referenced to filter transcriptional noise unmasking a conserved progenitor motif (55 up- and 233 down-regulated genes). The streamlined pool of 288 genes organized into a core biological network that prioritized the “Cardiovascular Development” function. Recursive in silico deconvolution of the cardiogenic neighborhood and associated canonical signaling pathways identified a combination of integrated axes, CXCR4/SDF-1, Flk-1/VEGF and BMP2r/BMP2, predicted to synchronize cardiac specification. In vitro targeting of the resolved triad in embryoid bodies accelerated expression of Nkx2.5, Mef2C and cardiac-MHC, enhanced beating activity, and augmented cardiogenic yield. Conclusions Transcriptome-wide dissection of a conserved progenitor profile thus revealed functional highways that coordinate cardiogenic maturation from a pluripotent ground state. Validating the bioinformatics algorithm established a strategy to rationally modulate cell fate, and optimize stem cell-derived cardiogenesis.
Collapse
Affiliation(s)
- Anca Chiriac
- Marriott Heart Disease Research Program, Division of Cardiovascular Diseases, Departments of Medicine, Molecular Pharmacology and Experimental Therapeutics, and Medical Genetics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Timothy J. Nelson
- Marriott Heart Disease Research Program, Division of Cardiovascular Diseases, Departments of Medicine, Molecular Pharmacology and Experimental Therapeutics, and Medical Genetics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Randolph S. Faustino
- Marriott Heart Disease Research Program, Division of Cardiovascular Diseases, Departments of Medicine, Molecular Pharmacology and Experimental Therapeutics, and Medical Genetics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Atta Behfar
- Marriott Heart Disease Research Program, Division of Cardiovascular Diseases, Departments of Medicine, Molecular Pharmacology and Experimental Therapeutics, and Medical Genetics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Andre Terzic
- Marriott Heart Disease Research Program, Division of Cardiovascular Diseases, Departments of Medicine, Molecular Pharmacology and Experimental Therapeutics, and Medical Genetics, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
39
|
Blin G, Nury D, Stefanovic S, Neri T, Guillevic O, Brinon B, Bellamy V, Rücker-Martin C, Barbry P, Bel A, Bruneval P, Cowan C, Pouly J, Mitalipov S, Gouadon E, Binder P, Hagège A, Desnos M, Renaud JF, Menasché P, Pucéat M. A purified population of multipotent cardiovascular progenitors derived from primate pluripotent stem cells engrafts in postmyocardial infarcted nonhuman primates. J Clin Invest 2010; 120:1125-39. [PMID: 20335662 PMCID: PMC2846046 DOI: 10.1172/jci40120] [Citation(s) in RCA: 233] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2009] [Accepted: 01/13/2010] [Indexed: 12/21/2022] Open
Abstract
Cell therapy holds promise for tissue regeneration, including in individuals with advanced heart failure. However, treatment of heart disease with bone marrow cells and skeletal muscle progenitors has had only marginal positive benefits in clinical trials, perhaps because adult stem cells have limited plasticity. The identification, among human pluripotent stem cells, of early cardiovascular cell progenitors required for the development of the first cardiac lineage would shed light on human cardiogenesis and might pave the way for cell therapy for cardiac degenerative diseases. Here, we report the isolation of an early population of cardiovascular progenitors, characterized by expression of OCT4, stage-specific embryonic antigen 1 (SSEA-1), and mesoderm posterior 1 (MESP1), derived from human pluripotent stem cells treated with the cardiogenic morphogen BMP2. This progenitor population was multipotential and able to generate cardiomyocytes as well as smooth muscle and endothelial cells. When transplanted into the infarcted myocardium of immunosuppressed nonhuman primates, an SSEA-1+ progenitor population derived from Rhesus embryonic stem cells differentiated into ventricular myocytes and reconstituted 20% of the scar tissue. Notably, primates transplanted with an unpurified population of cardiac-committed cells, which included SSEA-1- cells, developed teratomas in the scar tissue, whereas those transplanted with purified SSEA-1+ cells did not. We therefore believe that the SSEA-1+ progenitors that we have described here have the potential to be used in cardiac regenerative medicine.
Collapse
Affiliation(s)
- Guillaume Blin
- INSERM U633, Avenir Program, Embryonic Stem Cells and Cardiogenesis, Evry, France.
University Montpellier II, France.
INSERM U633, University Paris Descartes, France.
CNRS-UMR 8162, Université Paris-Sud, and Hôpital Marie Lannelongue, Le Plessis Robinson, France.
CNRS, University of Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, Nice, France.
Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, Department of Cardiovascular Surgery, Paris, France.
AP-HP, Hôpital Européen Georges-Pompidou, Department of Pathology, and INSERM U970, Paris, France.
Stowers Medical Institute, Center for Regenerative Medicine and Technology, Cardiovascular Research Center, Boston, Massachusetts.
Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton.
Institut de Médecine Aérospatiale du Service de Santé des Armées (IMASSA), Brétigny sur Orge, France.
Department of Cardiology, AP-HP, Hôpital Européen Georges-Pompidou
| | - David Nury
- INSERM U633, Avenir Program, Embryonic Stem Cells and Cardiogenesis, Evry, France.
University Montpellier II, France.
INSERM U633, University Paris Descartes, France.
CNRS-UMR 8162, Université Paris-Sud, and Hôpital Marie Lannelongue, Le Plessis Robinson, France.
CNRS, University of Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, Nice, France.
Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, Department of Cardiovascular Surgery, Paris, France.
AP-HP, Hôpital Européen Georges-Pompidou, Department of Pathology, and INSERM U970, Paris, France.
Stowers Medical Institute, Center for Regenerative Medicine and Technology, Cardiovascular Research Center, Boston, Massachusetts.
Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton.
Institut de Médecine Aérospatiale du Service de Santé des Armées (IMASSA), Brétigny sur Orge, France.
Department of Cardiology, AP-HP, Hôpital Européen Georges-Pompidou
| | - Sonia Stefanovic
- INSERM U633, Avenir Program, Embryonic Stem Cells and Cardiogenesis, Evry, France.
University Montpellier II, France.
INSERM U633, University Paris Descartes, France.
CNRS-UMR 8162, Université Paris-Sud, and Hôpital Marie Lannelongue, Le Plessis Robinson, France.
CNRS, University of Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, Nice, France.
Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, Department of Cardiovascular Surgery, Paris, France.
AP-HP, Hôpital Européen Georges-Pompidou, Department of Pathology, and INSERM U970, Paris, France.
Stowers Medical Institute, Center for Regenerative Medicine and Technology, Cardiovascular Research Center, Boston, Massachusetts.
Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton.
Institut de Médecine Aérospatiale du Service de Santé des Armées (IMASSA), Brétigny sur Orge, France.
Department of Cardiology, AP-HP, Hôpital Européen Georges-Pompidou
| | - Tui Neri
- INSERM U633, Avenir Program, Embryonic Stem Cells and Cardiogenesis, Evry, France.
University Montpellier II, France.
INSERM U633, University Paris Descartes, France.
CNRS-UMR 8162, Université Paris-Sud, and Hôpital Marie Lannelongue, Le Plessis Robinson, France.
CNRS, University of Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, Nice, France.
Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, Department of Cardiovascular Surgery, Paris, France.
AP-HP, Hôpital Européen Georges-Pompidou, Department of Pathology, and INSERM U970, Paris, France.
Stowers Medical Institute, Center for Regenerative Medicine and Technology, Cardiovascular Research Center, Boston, Massachusetts.
Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton.
Institut de Médecine Aérospatiale du Service de Santé des Armées (IMASSA), Brétigny sur Orge, France.
Department of Cardiology, AP-HP, Hôpital Européen Georges-Pompidou
| | - Oriane Guillevic
- INSERM U633, Avenir Program, Embryonic Stem Cells and Cardiogenesis, Evry, France.
University Montpellier II, France.
INSERM U633, University Paris Descartes, France.
CNRS-UMR 8162, Université Paris-Sud, and Hôpital Marie Lannelongue, Le Plessis Robinson, France.
CNRS, University of Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, Nice, France.
Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, Department of Cardiovascular Surgery, Paris, France.
AP-HP, Hôpital Européen Georges-Pompidou, Department of Pathology, and INSERM U970, Paris, France.
Stowers Medical Institute, Center for Regenerative Medicine and Technology, Cardiovascular Research Center, Boston, Massachusetts.
Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton.
Institut de Médecine Aérospatiale du Service de Santé des Armées (IMASSA), Brétigny sur Orge, France.
Department of Cardiology, AP-HP, Hôpital Européen Georges-Pompidou
| | - Benjamin Brinon
- INSERM U633, Avenir Program, Embryonic Stem Cells and Cardiogenesis, Evry, France.
University Montpellier II, France.
INSERM U633, University Paris Descartes, France.
CNRS-UMR 8162, Université Paris-Sud, and Hôpital Marie Lannelongue, Le Plessis Robinson, France.
CNRS, University of Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, Nice, France.
Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, Department of Cardiovascular Surgery, Paris, France.
AP-HP, Hôpital Européen Georges-Pompidou, Department of Pathology, and INSERM U970, Paris, France.
Stowers Medical Institute, Center for Regenerative Medicine and Technology, Cardiovascular Research Center, Boston, Massachusetts.
Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton.
Institut de Médecine Aérospatiale du Service de Santé des Armées (IMASSA), Brétigny sur Orge, France.
Department of Cardiology, AP-HP, Hôpital Européen Georges-Pompidou
| | - Valérie Bellamy
- INSERM U633, Avenir Program, Embryonic Stem Cells and Cardiogenesis, Evry, France.
University Montpellier II, France.
INSERM U633, University Paris Descartes, France.
CNRS-UMR 8162, Université Paris-Sud, and Hôpital Marie Lannelongue, Le Plessis Robinson, France.
CNRS, University of Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, Nice, France.
Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, Department of Cardiovascular Surgery, Paris, France.
AP-HP, Hôpital Européen Georges-Pompidou, Department of Pathology, and INSERM U970, Paris, France.
Stowers Medical Institute, Center for Regenerative Medicine and Technology, Cardiovascular Research Center, Boston, Massachusetts.
Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton.
Institut de Médecine Aérospatiale du Service de Santé des Armées (IMASSA), Brétigny sur Orge, France.
Department of Cardiology, AP-HP, Hôpital Européen Georges-Pompidou
| | - Catherine Rücker-Martin
- INSERM U633, Avenir Program, Embryonic Stem Cells and Cardiogenesis, Evry, France.
University Montpellier II, France.
INSERM U633, University Paris Descartes, France.
CNRS-UMR 8162, Université Paris-Sud, and Hôpital Marie Lannelongue, Le Plessis Robinson, France.
CNRS, University of Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, Nice, France.
Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, Department of Cardiovascular Surgery, Paris, France.
AP-HP, Hôpital Européen Georges-Pompidou, Department of Pathology, and INSERM U970, Paris, France.
Stowers Medical Institute, Center for Regenerative Medicine and Technology, Cardiovascular Research Center, Boston, Massachusetts.
Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton.
Institut de Médecine Aérospatiale du Service de Santé des Armées (IMASSA), Brétigny sur Orge, France.
Department of Cardiology, AP-HP, Hôpital Européen Georges-Pompidou
| | - Pascal Barbry
- INSERM U633, Avenir Program, Embryonic Stem Cells and Cardiogenesis, Evry, France.
University Montpellier II, France.
INSERM U633, University Paris Descartes, France.
CNRS-UMR 8162, Université Paris-Sud, and Hôpital Marie Lannelongue, Le Plessis Robinson, France.
CNRS, University of Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, Nice, France.
Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, Department of Cardiovascular Surgery, Paris, France.
AP-HP, Hôpital Européen Georges-Pompidou, Department of Pathology, and INSERM U970, Paris, France.
Stowers Medical Institute, Center for Regenerative Medicine and Technology, Cardiovascular Research Center, Boston, Massachusetts.
Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton.
Institut de Médecine Aérospatiale du Service de Santé des Armées (IMASSA), Brétigny sur Orge, France.
Department of Cardiology, AP-HP, Hôpital Européen Georges-Pompidou
| | - Alain Bel
- INSERM U633, Avenir Program, Embryonic Stem Cells and Cardiogenesis, Evry, France.
University Montpellier II, France.
INSERM U633, University Paris Descartes, France.
CNRS-UMR 8162, Université Paris-Sud, and Hôpital Marie Lannelongue, Le Plessis Robinson, France.
CNRS, University of Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, Nice, France.
Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, Department of Cardiovascular Surgery, Paris, France.
AP-HP, Hôpital Européen Georges-Pompidou, Department of Pathology, and INSERM U970, Paris, France.
Stowers Medical Institute, Center for Regenerative Medicine and Technology, Cardiovascular Research Center, Boston, Massachusetts.
Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton.
Institut de Médecine Aérospatiale du Service de Santé des Armées (IMASSA), Brétigny sur Orge, France.
Department of Cardiology, AP-HP, Hôpital Européen Georges-Pompidou
| | - Patrick Bruneval
- INSERM U633, Avenir Program, Embryonic Stem Cells and Cardiogenesis, Evry, France.
University Montpellier II, France.
INSERM U633, University Paris Descartes, France.
CNRS-UMR 8162, Université Paris-Sud, and Hôpital Marie Lannelongue, Le Plessis Robinson, France.
CNRS, University of Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, Nice, France.
Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, Department of Cardiovascular Surgery, Paris, France.
AP-HP, Hôpital Européen Georges-Pompidou, Department of Pathology, and INSERM U970, Paris, France.
Stowers Medical Institute, Center for Regenerative Medicine and Technology, Cardiovascular Research Center, Boston, Massachusetts.
Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton.
Institut de Médecine Aérospatiale du Service de Santé des Armées (IMASSA), Brétigny sur Orge, France.
Department of Cardiology, AP-HP, Hôpital Européen Georges-Pompidou
| | - Chad Cowan
- INSERM U633, Avenir Program, Embryonic Stem Cells and Cardiogenesis, Evry, France.
University Montpellier II, France.
INSERM U633, University Paris Descartes, France.
CNRS-UMR 8162, Université Paris-Sud, and Hôpital Marie Lannelongue, Le Plessis Robinson, France.
CNRS, University of Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, Nice, France.
Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, Department of Cardiovascular Surgery, Paris, France.
AP-HP, Hôpital Européen Georges-Pompidou, Department of Pathology, and INSERM U970, Paris, France.
Stowers Medical Institute, Center for Regenerative Medicine and Technology, Cardiovascular Research Center, Boston, Massachusetts.
Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton.
Institut de Médecine Aérospatiale du Service de Santé des Armées (IMASSA), Brétigny sur Orge, France.
Department of Cardiology, AP-HP, Hôpital Européen Georges-Pompidou
| | - Julia Pouly
- INSERM U633, Avenir Program, Embryonic Stem Cells and Cardiogenesis, Evry, France.
University Montpellier II, France.
INSERM U633, University Paris Descartes, France.
CNRS-UMR 8162, Université Paris-Sud, and Hôpital Marie Lannelongue, Le Plessis Robinson, France.
CNRS, University of Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, Nice, France.
Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, Department of Cardiovascular Surgery, Paris, France.
AP-HP, Hôpital Européen Georges-Pompidou, Department of Pathology, and INSERM U970, Paris, France.
Stowers Medical Institute, Center for Regenerative Medicine and Technology, Cardiovascular Research Center, Boston, Massachusetts.
Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton.
Institut de Médecine Aérospatiale du Service de Santé des Armées (IMASSA), Brétigny sur Orge, France.
Department of Cardiology, AP-HP, Hôpital Européen Georges-Pompidou
| | - Shoukhrat Mitalipov
- INSERM U633, Avenir Program, Embryonic Stem Cells and Cardiogenesis, Evry, France.
University Montpellier II, France.
INSERM U633, University Paris Descartes, France.
CNRS-UMR 8162, Université Paris-Sud, and Hôpital Marie Lannelongue, Le Plessis Robinson, France.
CNRS, University of Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, Nice, France.
Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, Department of Cardiovascular Surgery, Paris, France.
AP-HP, Hôpital Européen Georges-Pompidou, Department of Pathology, and INSERM U970, Paris, France.
Stowers Medical Institute, Center for Regenerative Medicine and Technology, Cardiovascular Research Center, Boston, Massachusetts.
Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton.
Institut de Médecine Aérospatiale du Service de Santé des Armées (IMASSA), Brétigny sur Orge, France.
Department of Cardiology, AP-HP, Hôpital Européen Georges-Pompidou
| | - Elodie Gouadon
- INSERM U633, Avenir Program, Embryonic Stem Cells and Cardiogenesis, Evry, France.
University Montpellier II, France.
INSERM U633, University Paris Descartes, France.
CNRS-UMR 8162, Université Paris-Sud, and Hôpital Marie Lannelongue, Le Plessis Robinson, France.
CNRS, University of Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, Nice, France.
Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, Department of Cardiovascular Surgery, Paris, France.
AP-HP, Hôpital Européen Georges-Pompidou, Department of Pathology, and INSERM U970, Paris, France.
Stowers Medical Institute, Center for Regenerative Medicine and Technology, Cardiovascular Research Center, Boston, Massachusetts.
Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton.
Institut de Médecine Aérospatiale du Service de Santé des Armées (IMASSA), Brétigny sur Orge, France.
Department of Cardiology, AP-HP, Hôpital Européen Georges-Pompidou
| | - Patrice Binder
- INSERM U633, Avenir Program, Embryonic Stem Cells and Cardiogenesis, Evry, France.
University Montpellier II, France.
INSERM U633, University Paris Descartes, France.
CNRS-UMR 8162, Université Paris-Sud, and Hôpital Marie Lannelongue, Le Plessis Robinson, France.
CNRS, University of Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, Nice, France.
Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, Department of Cardiovascular Surgery, Paris, France.
AP-HP, Hôpital Européen Georges-Pompidou, Department of Pathology, and INSERM U970, Paris, France.
Stowers Medical Institute, Center for Regenerative Medicine and Technology, Cardiovascular Research Center, Boston, Massachusetts.
Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton.
Institut de Médecine Aérospatiale du Service de Santé des Armées (IMASSA), Brétigny sur Orge, France.
Department of Cardiology, AP-HP, Hôpital Européen Georges-Pompidou
| | - Albert Hagège
- INSERM U633, Avenir Program, Embryonic Stem Cells and Cardiogenesis, Evry, France.
University Montpellier II, France.
INSERM U633, University Paris Descartes, France.
CNRS-UMR 8162, Université Paris-Sud, and Hôpital Marie Lannelongue, Le Plessis Robinson, France.
CNRS, University of Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, Nice, France.
Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, Department of Cardiovascular Surgery, Paris, France.
AP-HP, Hôpital Européen Georges-Pompidou, Department of Pathology, and INSERM U970, Paris, France.
Stowers Medical Institute, Center for Regenerative Medicine and Technology, Cardiovascular Research Center, Boston, Massachusetts.
Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton.
Institut de Médecine Aérospatiale du Service de Santé des Armées (IMASSA), Brétigny sur Orge, France.
Department of Cardiology, AP-HP, Hôpital Européen Georges-Pompidou
| | - Michel Desnos
- INSERM U633, Avenir Program, Embryonic Stem Cells and Cardiogenesis, Evry, France.
University Montpellier II, France.
INSERM U633, University Paris Descartes, France.
CNRS-UMR 8162, Université Paris-Sud, and Hôpital Marie Lannelongue, Le Plessis Robinson, France.
CNRS, University of Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, Nice, France.
Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, Department of Cardiovascular Surgery, Paris, France.
AP-HP, Hôpital Européen Georges-Pompidou, Department of Pathology, and INSERM U970, Paris, France.
Stowers Medical Institute, Center for Regenerative Medicine and Technology, Cardiovascular Research Center, Boston, Massachusetts.
Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton.
Institut de Médecine Aérospatiale du Service de Santé des Armées (IMASSA), Brétigny sur Orge, France.
Department of Cardiology, AP-HP, Hôpital Européen Georges-Pompidou
| | - Jean-François Renaud
- INSERM U633, Avenir Program, Embryonic Stem Cells and Cardiogenesis, Evry, France.
University Montpellier II, France.
INSERM U633, University Paris Descartes, France.
CNRS-UMR 8162, Université Paris-Sud, and Hôpital Marie Lannelongue, Le Plessis Robinson, France.
CNRS, University of Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, Nice, France.
Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, Department of Cardiovascular Surgery, Paris, France.
AP-HP, Hôpital Européen Georges-Pompidou, Department of Pathology, and INSERM U970, Paris, France.
Stowers Medical Institute, Center for Regenerative Medicine and Technology, Cardiovascular Research Center, Boston, Massachusetts.
Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton.
Institut de Médecine Aérospatiale du Service de Santé des Armées (IMASSA), Brétigny sur Orge, France.
Department of Cardiology, AP-HP, Hôpital Européen Georges-Pompidou
| | - Philippe Menasché
- INSERM U633, Avenir Program, Embryonic Stem Cells and Cardiogenesis, Evry, France.
University Montpellier II, France.
INSERM U633, University Paris Descartes, France.
CNRS-UMR 8162, Université Paris-Sud, and Hôpital Marie Lannelongue, Le Plessis Robinson, France.
CNRS, University of Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, Nice, France.
Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, Department of Cardiovascular Surgery, Paris, France.
AP-HP, Hôpital Européen Georges-Pompidou, Department of Pathology, and INSERM U970, Paris, France.
Stowers Medical Institute, Center for Regenerative Medicine and Technology, Cardiovascular Research Center, Boston, Massachusetts.
Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton.
Institut de Médecine Aérospatiale du Service de Santé des Armées (IMASSA), Brétigny sur Orge, France.
Department of Cardiology, AP-HP, Hôpital Européen Georges-Pompidou
| | - Michel Pucéat
- INSERM U633, Avenir Program, Embryonic Stem Cells and Cardiogenesis, Evry, France.
University Montpellier II, France.
INSERM U633, University Paris Descartes, France.
CNRS-UMR 8162, Université Paris-Sud, and Hôpital Marie Lannelongue, Le Plessis Robinson, France.
CNRS, University of Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, Nice, France.
Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, Department of Cardiovascular Surgery, Paris, France.
AP-HP, Hôpital Européen Georges-Pompidou, Department of Pathology, and INSERM U970, Paris, France.
Stowers Medical Institute, Center for Regenerative Medicine and Technology, Cardiovascular Research Center, Boston, Massachusetts.
Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton.
Institut de Médecine Aérospatiale du Service de Santé des Armées (IMASSA), Brétigny sur Orge, France.
Department of Cardiology, AP-HP, Hôpital Européen Georges-Pompidou
| |
Collapse
|
40
|
Otsuji TG, Minami I, Kurose Y, Yamauchi K, Tada M, Nakatsuji N. Progressive maturation in contracting cardiomyocytes derived from human embryonic stem cells: Qualitative effects on electrophysiological responses to drugs. Stem Cell Res 2010; 4:201-13. [PMID: 20199896 DOI: 10.1016/j.scr.2010.01.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 01/27/2010] [Accepted: 01/28/2010] [Indexed: 11/15/2022] Open
Abstract
The field of drug testing currently needs a new integrated assay system, as accurate as systems using native tissues, that will allow us to predict arrhythmia risks of candidate drugs and the relationship between genetic mutations and acquired electrophysiological phenotypes. This could be accomplished by combining the microelectrode array (MEA) system with cardiomyocytes (CMs) derived from human embryonic stem cells (hESC) and induced pluripotential stem cells. CMs have been successfully induced from both types, but their maturation process is not systematically controlled; this results in loss of beating potency and insufficient ion channel function. We generated a transgenic hESC line that facilitates maintenance of hESC-CM clusters every 2 weeks by expressing GFP driven by a cardiac-specific alphaMHC promoter, thereby producing a compact pacemaker lineage within a ventricular population over a year. Further analyses, including quantitative RT-PCR, patch-clamp, and MEA-mediated QT tests, demonstrated that replating culturing continuously enhanced gene expression, ionic current amplitudes, and resistance to K(+) channel blockades in hESC-CMs. Moreover, temporal three-dimensional (3D) culturing accelerated maturation by restoring the global gene repressive status established in the adhesive status. Replating/3D culturing thus produces hESC-CMs that act as functional syncytia suitable for use in regenerative medicine and accurate drug tests.
Collapse
|
41
|
Moretti A, Bellin M, Jung CB, Thies TM, Takashima Y, Bernshausen A, Schiemann M, Fischer S, Moosmang S, Smith AG, Lam JT, Laugwitz KL. Mouse and human induced pluripotent stem cells as a source for multipotent Isl1+ cardiovascular progenitors. FASEB J 2009; 24:700-11. [PMID: 19850773 DOI: 10.1096/fj.09-139477] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Ectopic expression of defined sets of genetic factors can reprogram somatic cells to create induced pluripotent stem (iPS) cells. The capacity to direct human iPS cells to specific differentiated lineages and to their progenitor populations can be used for disease modeling, drug discovery, and eventually autologous cell replacement therapies. During mouse cardiogenesis, the major lineages of the mature heart, cardiomyocytes, smooth muscle cells, and endothelial cells arise from a common, multipotent cardiovascular progenitor expressing the transcription factors Isl1 and Nkx2.5. Here we show, using genetic fate-mapping, that Isl1(+) multipotent cardiovascular progenitors can be generated from mouse iPS cells and spontaneously differentiate in all 3 cardiovascular lineages in vivo without teratoma. Moreover, we report the identification of human iPS-derived ISL1(+) progenitors with similar developmental potential. These results support the possibility to use patient-specific iPS-generated cardiovascular progenitors as a model to elucidate the pathogenesis of congenital and acquired forms of heart diseases.-Moretti, A., Bellin, M., Jung, C. B., Thies, T.-M., Takashima, Y., Bernshausen, A., Schiemann, M., Fischer, S., Moosmang, S., Smith, A. G., Lam, J. T., Laugwitz, K.-L. Mouse and human induced pluripotent stem cells as a source for multipotent Isl1(+) cardiovascular progenitors.
Collapse
Affiliation(s)
- Alessandra Moretti
- Klinikum rechts der Isar and Deutsches Herzzentrum, Technical University of Munich, I. Medical Department, Molecular Cardiology, Ismaninger Strasse 22, 81675 Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Csiszar A, Lehoux S, Ungvari Z. Hemodynamic forces, vascular oxidative stress, and regulation of BMP-2/4 expression. Antioxid Redox Signal 2009; 11:1683-97. [PMID: 19320562 PMCID: PMC2842584 DOI: 10.1089/ars.2008.2401] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Changes in the hemodynamic environment (e.g., hypertension, disturbed-flow conditions) are known to promote atherogenesis by inducing proinflammatory phenotypic alterations in endothelial and smooth muscle cells; however, the mechanisms underlying mechanosensitive induction of inflammatory gene expression are not completely understood. Bone morphogenetic protein-2 and -4 (BMP-2/4) are TGF-beta superfamily cytokines that are expressed by both endothelial and smooth muscle cells and regulate a number of cellular processes involved in atherogenesis, including vascular calcification and endothelial activation. This review considers how hemodynamic forces regulate BMP-2/4 expression and explores the role of mechanosensitive generation of reactive oxygen species by NAD(P)H oxidases in the control of BMP signaling.
Collapse
Affiliation(s)
- Anna Csiszar
- Department of Physiology, New York Medical College, Valhalla, New York, USA
| | | | | |
Collapse
|
43
|
Menasche P. Cell-based therapy for heart disease: a clinically oriented perspective. Mol Ther 2009; 17:758-66. [PMID: 19277020 DOI: 10.1038/mt.2009.40] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Over the past decade, cell therapy has emerged as a potential new treatment of a variety of cardiac diseases, including acute myocardial infarction, refractory angina, and chronic heart failure. A myriad of cell types have been tested experimentally, each of them being usually credited by its advocates of a high "regeneration" potential. This has led to a flurry of clinical trials entailing the use of skeletal myoblasts or bone marrow-derived cells either unfractionated or enriched in progenitor subpopulations. As often in medicine, the hype generated by the early uncontrolled and small-sized studies has been dampened by the marginally successful outcomes of the subsequent, more rigorously conducted randomized trials. Although they may have failed to achieve their primary end points, these trials have been positive in the sense that they have allowed to identify some key issues and it is reasonable to speculate that if these issues can now be addressed by appropriately focused benchwork, the outcomes of the second generation of cell-transplantation studies would likely be upgraded. It, thus, appears that not "one cell fits all" but that the selection of the cell type should be tailored to the primary clinical indication. On the one hand, it does not make sense to develop an "ideal" cell in a culture dish, if we remain unable to deliver it appropriately and to keep it alive, at least for a while, which requires to improve on the delivery techniques and to provide cells along with the vascular and extracellular matrix type of support necessary for their survival and patterning. On the other hand, the persisting mechanistic uncertainties about cell therapy should not preclude continuing clinical trials, which often provide the unique opportunity of identifying issues missed by our suboptimal preclinical models. Finally, regardless of whether cells are expected to act paracrinally or by physically replacing lost cardiomyocytes and, thus, effecting a true myocardial regeneration, safety remains a primary concern. It is, thus, important that clinical development programs be shaped in a way that allows the final cell-therapy product to be manufactured from fully traceable materials, phenotypically well characterized, consistent, scalable, sterile, and genetically stable as these characteristics are those that will be required by the ultimate gatekeeper, i.e., the regulator, and are thus unbypassable prerequisites for an effective and streamlined leap from bench to bedside.
Collapse
Affiliation(s)
- Philippe Menasche
- Department of Cardiovascular Surgery, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France.
| |
Collapse
|
44
|
Abstract
Human embryonic stem (HES) cells are pluripotent and give rise to any cell lineage. More specifically, how the first embryonic lineage (i.e., cardiac lineage) is acquired remains in many aspects questionable. Herein, we summarize the protocols that have been used to direct the fate of HES cells toward the cardiomyocytic lineage. We further discuss the regulation of transcriptional pathways underlying this process of differentiation. Finally, we propose perspectives of this research in the near future.
Collapse
|