1
|
Macrae RGC, Colzani MT, Williams TL, Bayraktar S, Kuc RE, Pullinger AL, Bernard WG, Robinson EL, Davenport EE, Maguire JJ, Sinha S, Davenport AP. Inducible apelin receptor knockdown reduces differentiation efficiency and contractility of hESC-derived cardiomyocytes. Cardiovasc Res 2023; 119:587-598. [PMID: 36239923 PMCID: PMC10064845 DOI: 10.1093/cvr/cvac065] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 04/04/2022] [Accepted: 04/12/2022] [Indexed: 11/14/2022] Open
Abstract
AIMS The apelin receptor, a G protein-coupled receptor, has emerged as a key regulator of cardiovascular development, physiology, and disease. However, there is a lack of suitable human in vitro models to investigate the apelinergic system in cardiovascular cell types. For the first time we have used human embryonic stem cell-derived cardiomyocytes (hESC-CMs) and a novel inducible knockdown system to examine the role of the apelin receptor in both cardiomyocyte development and to determine the consequences of loss of apelin receptor function as a model of disease. METHODS AND RESULTS Expression of the apelin receptor and its ligands in hESCs and hESC-CMs was determined. hESCs carrying a tetracycline-inducible short hairpin RNA targeting the apelin receptor were generated using the sOPTiKD system. Phenotypic assays characterized the consequences of either apelin receptor knockdown before hESC-CM differentiation (early knockdown) or in 3D engineered heart tissues as a disease model (late knockdown). hESC-CMs expressed the apelin signalling system at a similar level to the adult heart. Early apelin receptor knockdown decreased cardiomyocyte differentiation efficiency and prolonged voltage sensing, associated with asynchronous contraction. Late apelin receptor knockdown had detrimental consequences on 3D engineered heart tissue contractile properties, decreasing contractility and increasing stiffness. CONCLUSIONS We have successfully knocked down the apelin receptor, using an inducible system, to demonstrate a key role in hESC-CM differentiation. Knockdown in 3D engineered heart tissues recapitulated the phenotype of apelin receptor down-regulation in a failing heart, providing a potential platform for modelling heart failure and testing novel therapeutic strategies.
Collapse
Affiliation(s)
- Robyn G C Macrae
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Level 6, Addenbrooke’s Centre for Clinical Investigation, Box 110, Cambridge CB2 0QQ, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Maria T Colzani
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Thomas L Williams
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Level 6, Addenbrooke’s Centre for Clinical Investigation, Box 110, Cambridge CB2 0QQ, UK
| | - Semih Bayraktar
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Rhoda E Kuc
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Level 6, Addenbrooke’s Centre for Clinical Investigation, Box 110, Cambridge CB2 0QQ, UK
| | - Anna L Pullinger
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Level 6, Addenbrooke’s Centre for Clinical Investigation, Box 110, Cambridge CB2 0QQ, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - William G Bernard
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Emma L Robinson
- School of Medicine, Division of Cardiology, University of Colorado Denver, Aurora, CO, USA
| | | | - Janet J Maguire
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Level 6, Addenbrooke’s Centre for Clinical Investigation, Box 110, Cambridge CB2 0QQ, UK
| | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Anthony P Davenport
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Level 6, Addenbrooke’s Centre for Clinical Investigation, Box 110, Cambridge CB2 0QQ, UK
| |
Collapse
|
2
|
Apelin Receptor Signaling During Mesoderm Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020. [PMID: 32648246 DOI: 10.1007/5584_2020_567] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
The Apelin receptor (Aplnr) is a G-protein coupled receptor which has a wide body distribution and various physiological roles including homeostasis, angiogenesis, cardiovascular and neuroendocrine function. Apelin and Elabela are two peptide components of the Aplnr signaling and are cleaved to give different isoforms which are active in different tissues and organisms.Aplnr signaling is related to several pathologies including obesity, heart disases and cancer in the adult body. However, the developmental role in mammalian embryogenesis is crucial for migration of early cardiac progenitors and cardiac function. Aplnr and peptide components have a role in proliferation, differentiation and movement of endodermal precursors. Although expression of Aplnr signaling is observed in endodermal lineages, the main function is the control of mesoderm cell movement and cardiac development. Mutant of the Aplnr signaling components results in the malformations, defects and lethality mainly due to the deformed heart function. This developmental role share similarity with the cardiovascular functions in the adult body.Determination of Aplnr signaling and underlying mechanisms during mammalian development might enable understanding of regulatory molecular mechanisms which not only control embryonic development process but also control tissue function and disease pathology in the adult body.
Collapse
|
3
|
Kamaraj US, Chen J, Katwadi K, Ouyang JF, Yang Sun YB, Lim YM, Liu X, Handoko L, Polo JM, Petretto E, Rackham OJ. EpiMogrify Models H3K4me3 Data to Identify Signaling Molecules that Improve Cell Fate Control and Maintenance. Cell Syst 2020; 11:509-522.e10. [DOI: 10.1016/j.cels.2020.09.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 04/30/2020] [Accepted: 09/14/2020] [Indexed: 12/14/2022]
|
4
|
Chen Y, Qiao X, Zhang L, Li X, Liu Q. Apelin-13 regulates angiotensin ii-induced Cx43 downregulation and autophagy via the AMPK/mTOR signaling pathway in HL-1 cells. Physiol Res 2020; 69:813-822. [PMID: 32901500 DOI: 10.33549/physiolres.934488] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Atrial fibrillation is associated with atrial remodeling, in which connexin 43 (Cx43) and cell hypertrophy play important roles. In this study, apelin-13, an aliphatic peptide, was used to explore the protective effects of the adenosine monophosphate-activated protein kinase (AMPK)/mTOR signaling pathway on Cx43 expression and autophagy, using murine atrial HL-1 cells. The expression of Cx43, AMPK, B-type natriuretic peptide (BNP) and pathway-related proteins was detected by Western blot analysis. Cellular fluorescence imaging was used to visualize Cx43 distribution and the cytoskeleton. Our results showed that the Cx43 expression was significantly decreased in HL-1 cells treated with angiotensin II but increased in cells additionally treated with apelin-13. Meanwhile, apelin-13 decreased BNP expression and increased AMPK expression. However, the expression of Cx43 and LC3 increased by apelin-13 was inhibited by treatment with compound C, an AMPK inhibitor. In addition, rapamycin, an mTOR inhibitor, promoted the development of autophagy, further inhibited the protective effect on Cx43 expression and increased cell hypertrophy. Thus, apelin-13 enhances Cx43 expression and autophagy via the AMPK/mTOR signaling pathway, and serving as a potential therapeutic target for atrial fibrillation.
Collapse
Affiliation(s)
- Y Chen
- Shanxi Bethune Hospital Affiliated to Shanxi Medical University, Taiyuan, China. , Department of Pathophysiology, Shanxi Medical University, Taiyuan, China.
| | | | | | | | | |
Collapse
|
5
|
Pan Y, Li Q, Yan H, Huang J, Wang Z. Apela improves cardiac and renal function in mice with acute myocardial infarction. J Cell Mol Med 2020; 24:10382-10390. [PMID: 32686917 PMCID: PMC7521152 DOI: 10.1111/jcmm.15651] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/18/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022] Open
Abstract
Apela was recently identified as a new ligand of the apelin peptide jejunum (APJ) receptor. The purpose of this study was to investigate the role of apela in post-myocardial infarction (post-MI) recovery from cardiorenal damage. A murine MI model was established, and apela was then infused subcutaneously for two weeks. Echocardiographs were performed before and after infarction at the indicated times. Renal function was evaluated by serum and urine biochemistry. Immunohistochemistry of heart and kidney tissue was performed by in situ terminal deoxynucleotidyl transferase-mediated dUPT nick end-labelling reaction. Compared to the control group (MI/vehicle), the average value of the left ventricular ejection fraction in apela-treated mice increased by 32% and 39% at 2- and 4-week post-MI, respectively. The mean levels of serum blood urea nitrogen,creatinine, N-terminal pro-brain natriuretic peptide and 24-hour urine protein were significantly decreased at 4-week post-MI in apela-treated mice relative to that of control animals. At the cellular level, we found that apela treatment significantly reduced myocardial fibrosis and cellular apoptosis in heart and kidney tissue. These data suggest that apela improves cardiac and renal function in mice with acute MI. The peptide may be potential therapeutic agent for heart failure.
Collapse
Affiliation(s)
- Yang Pan
- Department of Cardiovascular Medicine, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China.,Department of Cardiovascular Medicine, Nanjing Chest Hospital, Nanjing, China
| | - Quanyi Li
- Department of Cardiovascular Medicine, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China.,Department of Cardiovascular Medicine, Nanjing Chest Hospital, Nanjing, China
| | - Hong Yan
- Department of Clinical Laboratory, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China.,Department of Clinical Laboratory, Nanjing Chest Hospital, Nanjing, China
| | - Jin Huang
- Department of Cardiovascular Medicine, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China.,Department of Cardiovascular Medicine, Nanjing Chest Hospital, Nanjing, China
| | - Zhi Wang
- Department of Cardiovascular Medicine, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China.,Department of Cardiovascular Medicine, Nanjing Chest Hospital, Nanjing, China
| |
Collapse
|
6
|
Esmaeili S, Bandarian F, Esmaeili B, Nasli-Esfahani E. Apelin and stem cells: the role played in the cardiovascular system and energy metabolism. Cell Biol Int 2019; 43:1332-1345. [PMID: 31166051 DOI: 10.1002/cbin.11191] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 06/02/2019] [Indexed: 01/24/2023]
Abstract
Apelin, a member of the adipokine family, is widely distributed in the body and exerts cytoprotective effects on many organs. Apelin isoforms are involved in different physiological processes, including regulation of the cardiovascular system, cardiac contractility, angiogenesis, and energy metabolism. Several investigations have been performed to study the effect of apelin on stem cell therapy. This review aims to summarize the literature representing the effects of apelin on stem cell properties. Furthermore, this review discusses the therapeutic potential of apelin-treated stem cells for cardiovascular diseases and demonstrates the effect of stem cells overexpressing apelin on energy metabolism. Stem cells with their unique characteristics play a crucial role in the maintenance of tissue integrity. These cells participate in tissue regeneration via multiple mechanisms. Although preclinical and clinical studies have demonstrated the therapeutic potential of stem cells in various diseases, their application in regenerative medicine has not been efficient. A number of strategies such as genetic modification or treatment of stem cells with different factors have been used to improve the efficacy of cell therapy and to increase their survival after transplantation. This article reviews the effect of apelin treatment on the efficacy of cell therapy.
Collapse
Affiliation(s)
- Shahnaz Esmaeili
- Diabetic Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, 1411713137, Iran
| | - Fatemeh Bandarian
- Diabetic Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, 1411713137, Iran
| | - Behnaz Esmaeili
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, 14194, Iran
| | - Ensieh Nasli-Esfahani
- Diabetic Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, 1411713137, Iran
| |
Collapse
|
7
|
Srivastava AC, Thompson YG, Singhal J, Stellern J, Srivastava A, Du J, O'Connor TR, Riggs AD. Elimination of human folypolyglutamate synthetase alters programming and plasticity of somatic cells. FASEB J 2019; 33:13747-13761. [PMID: 31585510 DOI: 10.1096/fj.201901721r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Folates are vital cofactors for the regeneration of S-adenosyl methionine, which is the methyl source for DNA methylation, protein methylation, and other aspects of one-carbon (C1) metabolism. Thus, folates are critical for establishing and preserving epigenetic programming. Folypolyglutamate synthetase (FPGS) is known to play a crucial role in the maintenance of intracellular folate levels. Therefore, any modulation in FPGS is expected to alter DNA methylation and numerous other metabolic pathways. To explore the role of polyglutamylation of folate, we eliminated both isoforms of FPGS in human cells (293T), producing FPGS knockout (FPGSko) cells. The elimination of FPGS significantly decreased cell proliferation, with a major effect on oxidative phosphorylation and a lesser effect on glycolysis. We found a substantial reduction in global DNA methylation and noteworthy changes in gene expression related to C1 metabolism, cell division, DNA methylation, pluripotency, Glu metabolism, neurogenesis, and cardiogenesis. The expression levels of NANOG, octamer-binding transcription factor 4, and sex-determining region Y-box 2 levels were increased in the mutant, consistent with the transition to a stem cell-like state. Gene expression and metabolite data also indicate a major change in Glu and GABA metabolism. In the appropriate medium, FPGSko cells can differentiate to produce mainly cells with characteristics of either neural stem cells or cardiomyocytes.-Srivastava, A. C., Thompson, Y. G., Singhal, J., Stellern, J., Srivastava, A., Du, J., O'Connor, T. R., Riggs, A. D. Elimination of human folypolyglutamate synthetase alters programming and plasticity of somatic cells.
Collapse
Affiliation(s)
- Avinash C Srivastava
- Department of Diabetes Complications and Metabolism, City of Hope National Medical Center, Duarte, California, USA
| | | | - Jyotsana Singhal
- Department of Diabetes Complications and Metabolism, City of Hope National Medical Center, Duarte, California, USA
| | - Jordan Stellern
- Department of Cancer Biology, City of Hope National Medical Center, Duarte, California, USA
| | - Anviksha Srivastava
- Department of Cancer Biology, City of Hope National Medical Center, Duarte, California, USA
| | - Juan Du
- Integrative Genomics Core Facility, City of Hope National Medical Center, Duarte, California, USA
| | - Timothy R O'Connor
- Department of Cancer Biology, City of Hope National Medical Center, Duarte, California, USA
| | - Arthur D Riggs
- Department of Diabetes Complications and Metabolism, City of Hope National Medical Center, Duarte, California, USA
| |
Collapse
|
8
|
Wang Z, Huang J. Apela Promotes Cardiomyocyte Differentiation from Transgenic Human Embryonic Stem Cell Lines. Appl Biochem Biotechnol 2019; 189:396-410. [PMID: 31025171 DOI: 10.1007/s12010-019-03012-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/27/2019] [Indexed: 12/22/2022]
Abstract
Although embryonic stem (ES) cells (ESCs) may be a promising donor source for the repair of infarcted or ischemic heart tissues, their successful application in regenerative medicine has been hampered by difficulties in enriching, identifying, and selecting cardiomyocytes from the differentiating cells. We established transgenic human ES cell lines by transcriptional control of the α-cardiac myosin heavy chain (α-MHC) promoter driving green fluorescent protein (GFP) expression. Differentiated GFP-expressing cells display the characteristics of cardiomyocytes (CMs). Apela, a recently identified short peptide, up-regulated the expression of the cardiac-restricted transcription factors Tbx5 and GATA4 as well as differentiated the cardiomyocyte markers α-MHC and β-MHC. Flow cytometric analysis showed that apela increased the percentage of GFP-expressing cells in the beating foci of the embryoid bodies. The percentage of cardiac troponin T (TNT)-positive cells and the protein expression of TNT were increased in the ES cell-derived CMs with apela treatment. Functionally, the contractile frequency of the ES-derived CMs responded appropriately to the vasoactive drugs isoprenaline and carbachol. Our work presented a protocol for specially labelling and enriching CMs by combining transgenic human ES cell lines and exogenous growth factor treatment.
Collapse
Affiliation(s)
- Zhi Wang
- Department of Cardiovascular Medicine, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, People's Republic of China. .,Department of Cardiovascular Medicine, Nanjing Chest Hospital, Nanjing, People's Republic of China. .,Department of Cardiology, Nanjing Brain Hospital, Nanjing Medical University, No. 264 Guangzhou Road, Nanjing, 210029, People's Republic of China.
| | - Jin Huang
- Department of Cardiovascular Medicine, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, People's Republic of China. .,Department of Cardiovascular Medicine, Nanjing Chest Hospital, Nanjing, People's Republic of China.
| |
Collapse
|
9
|
Kim YH, Kim BJ, Kim SM, Kim SU, Ryu BY. Induction of cardiomyocyte‑like cells from hair follicle cells in mice. Int J Mol Med 2019; 43:2230-2240. [PMID: 30864673 DOI: 10.3892/ijmm.2019.4133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 03/08/2019] [Indexed: 11/05/2022] Open
Abstract
Hair follicles (HFs) are a well‑characterized niche for adult stem cells (SCs), and include epithelial and melanocytic SCs. HF cells are an accessible source of multipotent adult SCs for the generation of the interfollicular epidermis, HF structures and sebaceous glands in addition to the reconstitution of novel HFs in vivo. In the present study, it was demonstrated that HF cells are able to be induced to differentiate into cardiomyocyte‑like cells in vitro under specific conditions. It was determined that HF cells cultured on OP9 feeder cells in KnockOut‑Dulbecco's modified Eagle's medium/B27 in the presence of vascular endothelial growth factors differentiated into cardiomyocyte‑like cells that express markers specific to cardiac lineage, but do not express non‑cardiac lineage markers including neural stem/progenitor cell, HF bulge cells or undifferentiated spermatogonia markers. These cardiomyocyte‑like cells exhibited a spindle‑ and filament‑shaped morphology similar to that presented by cardiac muscles and exhibited spontaneous beating that persisted for over 3 months. These results demonstrate that SC reprogramming and differentiation may be induced without resulting in any genetic modification, which is important for the clinical applications of SCs including tissue and organ regeneration.
Collapse
Affiliation(s)
- Yong-Hee Kim
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung‑Ang University, Anseong, Gyeonggi‑do 17546, Republic of Korea
| | - Bang-Jin Kim
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Seok-Man Kim
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung‑Ang University, Anseong, Gyeonggi‑do 17546, Republic of Korea
| | - Sun-Uk Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungcheongbuk‑do 28116, Republic of Korea
| | - Buom-Yong Ryu
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung‑Ang University, Anseong, Gyeonggi‑do 17546, Republic of Korea
| |
Collapse
|
10
|
Ma R, Liang J, Huang W, Guo L, Cai W, Wang L, Paul C, Yang HT, Kim HW, Wang Y. Electrical Stimulation Enhances Cardiac Differentiation of Human Induced Pluripotent Stem Cells for Myocardial Infarction Therapy. Antioxid Redox Signal 2018; 28:371-384. [PMID: 27903111 PMCID: PMC5770128 DOI: 10.1089/ars.2016.6766] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AIMS Electrical stimulation (EleS) can promote cardiac differentiation, but the underlying mechanism is not well known. This study investigated the effect of EleS on cardiomyocyte (CM) differentiation of human induced pluripotent stem cells (hiPSCs) and evaluated the therapeutic effects for the treatment of myocardial infarction (MI). RESULTS Cardiac differentiation of hiPSCs was induced with EleS after embryoid body formation. Spontaneously beating hiPSCs were observed as early at 2 days when treated with EleS compared with control treatment. The cardiac differentiation efficiency of hiPSCs was significantly enhanced by EleS. In addition, the functional maturation of hiPSC-CMs under EleS was confirmed by calcium indicators, intracellular Ca2+ levels, and expression of structural genes. Mechanistically, EleS mediated cardiac differentiation of hiPSCs through activation of Ca2+/PKC/ERK pathways, as revealed by RNA sequencing, quantitative polymerase chain reaction, and Western blotting. After transplantation in immunodeficient MI mice, EleS-preconditioned hiPSC-derived cells significantly improved cardiac function and attenuated expansion of infarct size. The preconditioned hiPSC-derived CMs were functionally integrated with the host heart. INNOVATION We show EleS as an efficacious time-saving approach for CM generation. The global RNA profiling shows that EleS can accelerate cardiac differentiation of hiPSCs through activation of multiple pathways. The cardiac-mimetic electrical signals will provide a novel approach to generate functional CMs and facilitate cardiac tissue engineering for successful heart regeneration. CONCLUSION EleS can enhance efficiency of cardiac differentiation in hiPSCs and promote CM maturation. The EleS-preconditioned CMs emerge as a promising approach for clinical application in MI treatment. Antioxid. Redox Signal. 28, 371-384.
Collapse
Affiliation(s)
- Ruilian Ma
- 1 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Jialiang Liang
- 1 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Wei Huang
- 1 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Linlin Guo
- 1 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Wenfeng Cai
- 1 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Lei Wang
- 1 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Christian Paul
- 1 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Huang-Tian Yang
- 2 Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) and Shanghai Jiao Tong University School of Medicine (SJTUSM) , Shanghai, China
| | - Ha Won Kim
- 1 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Yigang Wang
- 1 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati , Cincinnati, Ohio
| |
Collapse
|
11
|
Li X, Yu L, Gao J, Bi X, Zhang J, Xu S, Wang M, Chen M, Qiu F, Fu G. Apelin Ameliorates High Glucose-Induced Downregulation of Connexin 43 via AMPK-Dependent Pathway in Neonatal Rat Cardiomyocytes. Aging Dis 2018; 9:66-76. [PMID: 29392082 PMCID: PMC5772859 DOI: 10.14336/ad.2017.0426] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 04/26/2017] [Indexed: 11/10/2022] Open
Abstract
Diabetes Mellitus is a common disorder, with increasing risk of cardiac arrhythmias. Studies have shown that altered connexin expression and gap junction remodeling under hyperglycemia contribute to the high prevalence of cardiac arrhythmias and even sudden death. Connexin 43 (Cx43), a major protein that assembles to form cardiac gap junctions, has been found to be downregulated under high glucose conditions, along with inhibition of gap junctional intercellular communication (GJIC). While, apelin, a beneficial adipokine, increases Cx43 protein expression in mouse and human embryonic stem cells during cardiac differentiation. However, it remains unknown whether apelin influences GJIC capacity in cardiomyocytes. Here, using Western blotting and dye transfer assays, we found that Cx43 protein expression was reduced and GJIC was impaired after treatment with high glucose, which, however, could be abrogated after apelin treatment for 48 h. We also found that apelin increased Cx43 expression under normal glucose. Real-time PCR showed that the Cx43 mRNA was not significantly affected under high glucose conditions in the presence of apelin or high glucose and apelin. High glucose decreased the phosphorylation of AMPKα; however, apelin activated AMPKα. Interestingly, we found that Cx43 expression was increased after treatment with AICAR, an activator of AMPK signaling. AMPKα inhibition mediated with transfection of siRNA-AMPKα1 and siRNA-AMPKα2 abolished the protective effect of apelin on Cx43 expression. Our data suggest that apelin attenuates high glucose-induced Cx43 downregulation and improves the loss of functional gap junctions partly through the AMPK pathway.
Collapse
Affiliation(s)
- Xiaoting Li
- 1Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lu Yu
- 1Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jing Gao
- 1Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xukun Bi
- 1Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Juhong Zhang
- 2Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shiming Xu
- 3Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Meihui Wang
- 4Biomedical Research Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Mengmeng Chen
- 1Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Fuyu Qiu
- 1Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Guosheng Fu
- 1Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
12
|
Hou J, Wang L, Long H, Wu H, Wu Q, Zhong T, Chen X, Zhou C, Guo T, Wang T. Hypoxia preconditioning promotes cardiac stem cell survival and cardiogenic differentiation in vitro involving activation of the HIF-1α/apelin/APJ axis. Stem Cell Res Ther 2017; 8:215. [PMID: 28962638 PMCID: PMC5622481 DOI: 10.1186/s13287-017-0673-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/03/2017] [Accepted: 09/13/2017] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Cardiac stem cells (CSCs) transplantation has been regarded as an optimal therapeutic approach for cardiovascular disease. However, inferior survival and low differentiation efficiency of these cells in the local infarct site reduce their therapeutic efficacy. In this study, we investigated the influence of hypoxia preconditioning (HP) on CSCs survival and cardiogenic differentiation in vitro and explored the relevant mechanism. METHODS CSCs were obtained from Sprague-Dawley rats and cells of the third passage were cultured in vitro and exposed to hypoxia (1% O2). Cells survival and apoptosis were evaluated by MTS assay and flow cytometry respectively. Cardiogenic differentiation was induced by using 5-azacytidine for another 24 h after the cells experienced HP. Normoxia (20% O2) was used as a negative control during the whole process. Cardiogenic differentiation was assessed 2 weeks after the induction. Relevant molecules were examined after HP and during the differentiation process. Anti-hypoxia-inducible factor-1α (HIF-1α) small interfering RNA (siRNA), anti-apelin siRNA, and anti-putative receptor protein related to the angiotensin receptor AT1 (APJ) siRNA were transfected in order to block their expression, and relevant downstream molecules were detected. RESULTS Compared with the normoxia group, the hypoxia group presented more rapid growth at time points of 12 and 24 h (p < 0.01). Cells exhibited the highest proliferation rate at the time point of 24 h (p < 0.01). The cell apoptosis rate significantly declined after 24 h of hypoxia exposure (p < 0.01). Expression levels of HIF-1α, apelin, and APJ were all enhanced after HP. The percentage of apelin, α-SA, and cTnT positive cells was greatly increased in the HP group after 2 weeks of induction. The protein level of α-SA and cTnT was also significantly elevated at 7 and 14 days (p < 0.01). HIF-1α, apelin, and APJ were all increased at different time points during the cardiogenic differentiation process (p < 0.01). Knockdown of HIF-1α, apelin or APJ by siRNAs resulted in a significant reduction of α-SA and cTnT. HIF-1α blockage caused a remarkable decrease of apelin and APJ (p < 0.01). Expression levels of apelin and APJ were depressed after the inhibition of apelin (p < 0.01). CONCLUSION HP could effectively promote CSCs survival and cardiogenic differentiation in vitro, and this procedure involved activation of the HIF-1α/apelin/APJ axis. This study provided a new perspective for exploring novel strategies to enhance CSCs transplantation efficiency.
Collapse
Affiliation(s)
- Jingying Hou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong, 510120, China.,Department of Emergency, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong, 510120, China
| | - Lei Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong, 510120, China.,Department of Emergency, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong, 510120, China
| | - Huibao Long
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong, 510120, China.,Department of Emergency, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong, 510120, China
| | - Hao Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong, 510120, China.,Department of Emergency, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong, 510120, China
| | - Quanhua Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong, 510120, China.,Department of Emergency, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong, 510120, China
| | - Tingting Zhong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong, 510120, China.,Department of Emergency, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong, 510120, China
| | - Xuxiang Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong, 510120, China.,Department of Emergency, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong, 510120, China
| | - Changqing Zhou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong, 510120, China.,Department of Emergency, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong, 510120, China
| | - Tianzhu Guo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong, 510120, China.,Department of Emergency, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong, 510120, China
| | - Tong Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong, 510120, China. .,Department of Emergency, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong, 510120, China. .,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, 107 Yanjiang Xi Road, Guangzhou, Guangdong, China.
| |
Collapse
|
13
|
Fu J, Chuah YJ, Ang WT, Zheng N, Wang DA. Optimization of a polydopamine (PD)-based coating method and polydimethylsiloxane (PDMS) substrates for improved mouse embryonic stem cell (ESC) pluripotency maintenance and cardiac differentiation. Biomater Sci 2017; 5:1156-1173. [DOI: 10.1039/c7bm00266a] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Optimization of a polydopamine (PD)-based coating method and PDMS substrates for improved ESC pluripotency maintenance and cardiac differentiation.
Collapse
Affiliation(s)
- Jiayin Fu
- Division of Bioengineering
- School of Chemical and Biomedical Engineering
- Nanyang Technological University
- Singapore 637457
| | - Yon Jin Chuah
- Division of Bioengineering
- School of Chemical and Biomedical Engineering
- Nanyang Technological University
- Singapore 637457
| | - Wee Tong Ang
- Division of Bioengineering
- School of Chemical and Biomedical Engineering
- Nanyang Technological University
- Singapore 637457
| | - Nan Zheng
- Division of Bioengineering
- School of Chemical and Biomedical Engineering
- Nanyang Technological University
- Singapore 637457
| | - Dong-An Wang
- Division of Bioengineering
- School of Chemical and Biomedical Engineering
- Nanyang Technological University
- Singapore 637457
| |
Collapse
|
14
|
Chung WJ, Cho A, Byun K, Moon J, Ge X, Seo HS, Moon E, Dash R, Yang PC. Apelin-13 infusion salvages the peri-infarct region to preserve cardiac function after severe myocardial injury. Int J Cardiol 2016; 222:361-367. [PMID: 27500765 DOI: 10.1016/j.ijcard.2016.07.263] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 07/22/2016] [Accepted: 07/30/2016] [Indexed: 11/30/2022]
Abstract
BACKGROUND Apelin-13 (A13) regulates cardiac homeostasis. However, the effects and mechanism of A13 infusion after an acute myocardial injury (AMI) have not been elucidated. This study assesses the restorative effects and mechanism of A13 on the peri-infarct region in murine AMI model. METHODS 51 FVB/N mice (12weeks, 30g) underwent AMI. A week following injury, continuous micro-pump infusion of A13 (0.5μg/g/day) and saline was initiated for 4-week duration. Dual contrast MRI was conducted on weeks 1, 2, 3, and 5, consisting of delayed-enhanced and manganese-enhanced MRI. Four mice in each group were followed for an extended period of 4weeks without further infusion and underwent MRI scans on weeks 7 and 9. RESULTS A13 infusion demonstrated preserved LVEF compared to saline from weeks 1 to 4 (21.9±3.2% to 23.1±1.7%* vs. 23.5±1.7% to 16.9±2.8%, *p=0.02), which persisted up to 9weeks post-MI (+1.4%* vs. -9.4%, *p=0.03). Mechanistically, dual contrast MRI demonstrated significant decrease in the peri-infarct and scar % volume in A13 group from weeks 1 to 4 (15.1 to 7.4% and 34.3 to 25.1%, p=0.02, respectively). This was corroborated by significant increase in 5-ethynyl-2'-deoxyuridine (EdU(+)) cells by A13 vs. saline groups in the peri-infarct region (16.5±3.1% vs. 8.1±1.6%; p=0.04), suggesting active cell mitosis. Finally, significantly enhanced mobilization of CD34(+) cells in the peripheral blood and up-regulation of APJ, fibrotic, and apoptotic genes in the peri-infarct region were found. CONCLUSIONS A13 preserves cardiac performance by salvaging the peri-infarct region and may contribute to permanent restoration of the severely injured myocardium.
Collapse
Affiliation(s)
- Wook-Jin Chung
- Department of Cardiovascular Medicine, Stanford University, Stanford, CA, USA; Department of Cardiovascular Medicine, Gachon University, Incheon, Republic of Korea; Gachon Cardiovascular Research Institute, Gachon University, Incheon, Republic of Korea
| | - Ahryon Cho
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Kyunghee Byun
- Gachon Cardiovascular Research Institute, Gachon University, Incheon, Republic of Korea; Department of Anatomy and Cell Biology, Gachon University, Incheon, Republic of Korea; Center for Genomics and Proteomics & Stem Cell Core Facility, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Jeongsik Moon
- Gachon Cardiovascular Research Institute, Gachon University, Incheon, Republic of Korea; Center for Genomics and Proteomics & Stem Cell Core Facility, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Xiaohu Ge
- Department of Cardiovascular Medicine, Stanford University, Stanford, CA, USA
| | - Hye-Sun Seo
- Division of Cardiology, Soon Chun Hyang University College of Medicine, Bucheon, Republic of Korea
| | - Ejung Moon
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Rajesh Dash
- Department of Cardiovascular Medicine, Stanford University, Stanford, CA, USA
| | - Phillip C Yang
- Department of Cardiovascular Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
15
|
Wei W, Huang W, Yue J. Requirement of IP3 receptor 3 (IP3R3) in nitric oxide induced cardiomyocyte differentiation of mouse embryonic stem cells. Exp Cell Res 2016; 346:9-16. [PMID: 27349290 DOI: 10.1016/j.yexcr.2016.06.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 06/21/2016] [Accepted: 06/23/2016] [Indexed: 01/29/2023]
Abstract
Nitric oxide (NO) markedly induces cardiomyocyte (CM) differentiation of embryonic stem (ES) cells. Here we examined the role of the Ca(2+) signaling in the NO-induced CM differentiation of mouse ES cells. We found that NO induced intracellular Ca(2+) increases in ES cells in a dose-dependent manner, and application of IP3 pathway antagonists not only significantly inhibited this induced Ca(2+) increase but also abolished NO-induced CM differentiation of ES cells. Subsequently, all 3 types of inositol 1, 4, 5-trisphosphate (IP3) receptors (IP3Rs) in mouse ES cells were individually or triply knocked down. Interestingly, only knockdown of type 3 IP3R (IP3R3) or triple-knockdown of three types of IP3Rs significantly inhibited the NO-induced Ca(2+) increases. Consistently, IP3R3 knockdown blocked the NO-induced CM differentiation of ES cells. CMs derived from IP3R3 knockdown ES cells also showed both structural and functional defects. In summary, our results indicate that the IP3R3-Ca(2+) pathway is required for NO-induced CM differentiation of ES cells.
Collapse
Affiliation(s)
- Wenjie Wei
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China; Department of Biology, South University of Science and Technology of China, Shenzhen 518052, China
| | - Wei Huang
- Department of Biology, South University of Science and Technology of China, Shenzhen 518052, China
| | - Jianbo Yue
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China.
| |
Collapse
|
16
|
Wang L, Zhu ZM, Zhang NK, Fang ZR, Xu XH, Zheng N, Gao LR. Apelin: an endogenous peptide essential for cardiomyogenic differentiation of mesenchymal stem cells via activating extracellular signal-regulated kinase 1/2 and 5. Cell Biol Int 2016; 40:501-14. [PMID: 26787000 DOI: 10.1002/cbin.10581] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 01/12/2016] [Indexed: 01/25/2023]
Abstract
Growing evidence has shown that apelin/APJ system functions as a critical mediator of cardiac development as well as cardiovascular function. Here, we investigated the role of apelin in the cardiomyogenic differentiation of mesenchymal stem cells derived from Wharton's jelly of human umbilical cord in vitro. In this research, we used RNA interference methodology and gene transfection technique to regulate the expression of apelin in Wharton's jelly-derived mesenchymal stem cells and induced cells with a effective cardiac differentiation protocol including 5-azacytidine and bFGF. Four weeks after induction, induced cells assumed a stick-like morphology and myotube-like structures except apelin-silenced cells and the control group. The silencing expression of apelin in Wharton's jelly-derived mesenchymal stem cells decreased the expression of several critical cardiac progenitor transcription factors (Mesp1, Mef2c, NKX2.5) and cardiac phenotypes (cardiac α-actin, β-MHC, cTnT, and connexin-43). Meanwhile, endogenous compensation of apelin contributed to differentiating into cells with characteristics of cardiomyocytes in vitro. Further experiment showed that exogenous apelin peptide rescued the cardiomyogenic differentiation of apelin-silenced mesenchymal stem cells in the early stage (1-4 days) of induction. Remarkably, our experiment indicated that apelin up-regulated cardiac specific genes in Wharton's jelly-derived mesenchymal stem cells via activating extracellular signal-regulated kinase (ERK) 1/2 and 5.
Collapse
Affiliation(s)
- Li Wang
- Cardiovascular Center, Navy General Hospital, Beijing, 100048, China
- Department of Internal Medicine, The 413th Hospital of P. L. A., Zhoushan, Zhejiang, 316000, China
| | - Zhi-Ming Zhu
- Cardiovascular Center, Navy General Hospital, Beijing, 100048, China
| | - Ning-Kun Zhang
- Cardiovascular Center, Navy General Hospital, Beijing, 100048, China
| | - Zhi-Rong Fang
- Department of Internal Medicine, The 413th Hospital of P. L. A., Zhoushan, Zhejiang, 316000, China
| | - Xiao-Hong Xu
- Cardiovascular Center, Navy General Hospital, Beijing, 100048, China
| | - Nan Zheng
- Cardiovascular Center, Navy General Hospital, Beijing, 100048, China
| | - Lian-Ru Gao
- Cardiovascular Center, Navy General Hospital, Beijing, 100048, China
| |
Collapse
|
17
|
Cardiotrophin-1 promotes cardiomyocyte differentiation from mouse induced pluripotent stem cells via JAK2/STAT3/Pim-1 signaling pathway. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2016; 12:591-9. [PMID: 26788034 PMCID: PMC4712363 DOI: 10.11909/j.issn.1671-5411.2015.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
BACKGROUND The induced pluripotent stem cell (iPSC) has shown great potential in cellular therapy of myocardial infarction (MI), while its application is hampered by the low efficiency of cardiomyocyte differentiation. The present study was designed to investigate the effects of cardiotrophin-1 (CT-1) on cardiomyocyte differentiation from mouse induced pluripotent stem cells (miPSCs) and the underlying mechanisms involved. METHODS The optimal treatment condition for cardiomyocyte differentiation from miPSCs was established with ideal concentration (10 ng/mL) and duration (from day 3 to day 14) of CT-1 administration. Up-regulated expression of cardiac specific genes that accounted for embryonic cardiogenesis was observed by quantitative RT-PCR. Elevated amount of α-myosin heavy chain (α-MHC) and cardiac troponin I (cTn I) positive cells were detected by immunofluorescence staining and flow cytometry analysis in CT-1 group. RESULTS Transmission electron microscopic analysis revealed that cells treated with CT-1 showed better organized sacromeric structure and more mitochondria, which are morphological characteristic of matured cardiomyocytes. Western blot demonstrated that CT-1 promotes cardiomyocyte differentiation from miPSCs partly via JAK2/STAT3/Pim-1 pathway as compared with control group. CONCLUSIONS These findings suggested that CT-1 could enhance the cardiomyocyte differentiation as well as the maturation of mouse induced pluripotent stem cell derived cardiomyocytes by regulating JAK2/STAT3/Pim-1signaling pathway.
Collapse
|
18
|
Dolatshad NF, Hellen N, Jabbour RJ, Harding SE, Földes G. G-protein Coupled Receptor Signaling in Pluripotent Stem Cell-derived Cardiovascular Cells: Implications for Disease Modeling. Front Cell Dev Biol 2015; 3:76. [PMID: 26697426 PMCID: PMC4673467 DOI: 10.3389/fcell.2015.00076] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 11/09/2015] [Indexed: 12/13/2022] Open
Abstract
Human pluripotent stem cell derivatives show promise as an in vitro platform to study a range of human cardiovascular diseases. A better understanding of the biology of stem cells and their cardiovascular derivatives will help to understand the strengths and limitations of this new model system. G-protein coupled receptors (GPCRs) are key regulators of stem cell maintenance and differentiation and have an important role in cardiovascular cell signaling. In this review, we will therefore describe the state of knowledge concerning the regulatory role of GPCRs in both the generation and function of pluripotent stem cell derived-cardiomyocytes, -endothelial, and -vascular smooth muscle cells. We will consider how far the in vitro disease models recapitulate authentic GPCR signaling and provide a useful basis for discovery of disease mechanisms or design of therapeutic strategies.
Collapse
Affiliation(s)
- Nazanin F Dolatshad
- Myocardial Function, National Heart and Lung Institute, Imperial College London London, UK
| | - Nicola Hellen
- Myocardial Function, National Heart and Lung Institute, Imperial College London London, UK
| | - Richard J Jabbour
- Myocardial Function, National Heart and Lung Institute, Imperial College London London, UK
| | - Sian E Harding
- Myocardial Function, National Heart and Lung Institute, Imperial College London London, UK
| | - Gabor Földes
- Myocardial Function, National Heart and Lung Institute, Imperial College London London, UK ; The Heart and Vascular Center of Semmelweis University, Semmelweis University Budapest, Hungary
| |
Collapse
|
19
|
Ye L, Ding F, Zhang L, Shen A, Yao H, Deng L, Ding Y. Serum apelin is associated with left ventricular hypertrophy in untreated hypertension patients. J Transl Med 2015; 13:290. [PMID: 26342945 PMCID: PMC4560865 DOI: 10.1186/s12967-015-0635-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 08/11/2015] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Apelin is an endogenous ligand for the G protein-coupled receptor APJ. The association between apelin and cardiac modeling has been reported. However, if serum apelin affect the left ventricular hypertrophy (LVH) prevalence in hypertensive patients remains unknown. METHODS We enrolled 344 untreated hypertensive patients. The presence of LVH was determined by echocardiography. The blood was drawn from these patients and serum apelin level was detected. To study the direct effect of apelin on cardiac hypertrophy, cardiomyocytes were cultured and were transfected with apelin gene. Morphometric analysis and measurement of protein contain per cell were then performed. RESULTS We observed a significantly lower serum apelin level in hypertensive patients with LVH compared with those without LVH. Receiver operating characteristic analyses shows that serum apelin level is robust in discriminating patients with LVH from those without. Our in vitro study showed that cellular protein content and cellular size was increased by Ang II treatment, which can be markedly inhibited by the apelin over-expression in cultured cardiomyocytes. CONCLUSION Our clinical date established a link between apelin and LVH, suggesting serum apelin may be used as a predicator for LVH prevalence in hypertensive patients. The direct evidence in vitro suggest apelin pathway is involved in the cardiomyocyte adaption to hypertrophic stimuli.
Collapse
Affiliation(s)
- Lijun Ye
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical College, No. 57 Southern Renmin Avenue, 524023, Zhanjiang, Guangdong, China.
| | - Fenghua Ding
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical College, No. 57 Southern Renmin Avenue, 524023, Zhanjiang, Guangdong, China.
| | - Liang Zhang
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical College, No. 57 Southern Renmin Avenue, 524023, Zhanjiang, Guangdong, China.
| | - Anna Shen
- Department of Cardiology, The Third Affiliated Hospital of Southern Medical University, No.183, West Zhongshan Ave, Guangzhou, Tianhe District, China.
| | - Huaguo Yao
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical College, No. 57 Southern Renmin Avenue, 524023, Zhanjiang, Guangdong, China.
| | - Liehua Deng
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical College, No. 57 Southern Renmin Avenue, 524023, Zhanjiang, Guangdong, China.
| | - Yuanlin Ding
- The Institute of Medical System Biology, School of Public Health, Guangdong Medical College, Dongguan, China.
| |
Collapse
|
20
|
Han S, Englander EW, Gomez GA, Rastellini C, Quertermous T, Kundu RK, Greeley GH. Pancreatic Islet APJ Deletion Reduces Islet Density and Glucose Tolerance in Mice. Endocrinology 2015; 156:2451-60. [PMID: 25965959 DOI: 10.1210/en.2014-1631] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Protection and replenishment of a functional pancreatic β-cell mass (BCM) are key goals of all diabetes therapies. Apelin, a small regulatory peptide, is the endogenous ligand for the apelin receptor (APJ) receptor. The apelin-APJ signaling system is expressed in rodent and human islet cells. Apelin exposure has been shown to inhibit and to stimulate insulin secretion. Our aim was to assess the influence of a selective APJ deletion in pancreatic islet cells on islet homeostasis and glucose tolerance in mice. Cre-LoxP strategy was utilized to mediate islet APJ deletion. APJ deletion in islet cells (APJ(Δislet)) resulted in a significantly reduced islet size, density and BCM. An ip glucose tolerance test showed significantly impaired glucose clearance in APJ(Δislet) mice. APJ(Δislet) mice were not insulin resistant and in vivo glucose-stimulated insulin secretion was reduced modestly. In vitro glucose-stimulated insulin secretion showed a significantly reduced insulin secretion by islets from APJ(Δislet) mice. Glucose clearance in response to ip glucose tolerance test in obese APJ(Δislet) mice fed a chronic high-fat (HF) diet, but not pregnant APJ(Δislet) mice, was impaired significantly. In addition, the obesity-induced adaptive elevations in mean islet size and fractional islet area were reduced significantly in obese APJ(Δislet) mice when compared with wild-type mice. Together, these findings demonstrate a stimulatory role for the islet cell apelin-APJ signaling axis in regulation of pancreatic islet homeostasis and in metabolic induced β-cell hyperplasia. The results indicate the apelin-APJ system can be exploited for replenishment of BCM.
Collapse
Affiliation(s)
- Song Han
- Department of Surgery (S.H., E.W.E., G.A.G., C.R., G.H.G.), University of Texas Medical Branch, Galveston, Texas 77555; and School of Medicine (T.Q., R.K.K.), Division of Cardiovascular Medicine, Stanford University, Stanford, California 94305
| | - Ella W Englander
- Department of Surgery (S.H., E.W.E., G.A.G., C.R., G.H.G.), University of Texas Medical Branch, Galveston, Texas 77555; and School of Medicine (T.Q., R.K.K.), Division of Cardiovascular Medicine, Stanford University, Stanford, California 94305
| | - Guillermo A Gomez
- Department of Surgery (S.H., E.W.E., G.A.G., C.R., G.H.G.), University of Texas Medical Branch, Galveston, Texas 77555; and School of Medicine (T.Q., R.K.K.), Division of Cardiovascular Medicine, Stanford University, Stanford, California 94305
| | - Cristiana Rastellini
- Department of Surgery (S.H., E.W.E., G.A.G., C.R., G.H.G.), University of Texas Medical Branch, Galveston, Texas 77555; and School of Medicine (T.Q., R.K.K.), Division of Cardiovascular Medicine, Stanford University, Stanford, California 94305
| | - Thomas Quertermous
- Department of Surgery (S.H., E.W.E., G.A.G., C.R., G.H.G.), University of Texas Medical Branch, Galveston, Texas 77555; and School of Medicine (T.Q., R.K.K.), Division of Cardiovascular Medicine, Stanford University, Stanford, California 94305
| | - Ramendra K Kundu
- Department of Surgery (S.H., E.W.E., G.A.G., C.R., G.H.G.), University of Texas Medical Branch, Galveston, Texas 77555; and School of Medicine (T.Q., R.K.K.), Division of Cardiovascular Medicine, Stanford University, Stanford, California 94305
| | - George H Greeley
- Department of Surgery (S.H., E.W.E., G.A.G., C.R., G.H.G.), University of Texas Medical Branch, Galveston, Texas 77555; and School of Medicine (T.Q., R.K.K.), Division of Cardiovascular Medicine, Stanford University, Stanford, California 94305
| |
Collapse
|
21
|
Activin-A and Bmp4 levels modulate cell type specification during CHIR-induced cardiomyogenesis. PLoS One 2015; 10:e0118670. [PMID: 25706534 PMCID: PMC4338295 DOI: 10.1371/journal.pone.0118670] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 01/22/2015] [Indexed: 11/19/2022] Open
Abstract
The use of human pluripotent cell progeny for cardiac disease modeling, drug testing and therapeutics requires the ability to efficiently induce pluripotent cells into the cardiomyogenic lineage. Although direct activation of the Activin-A and/or Bmp pathways with growth factors yields context-dependent success, recent studies have shown that induction of Wnt signaling using low molecular weight molecules such as CHIR, which in turn induces the Activin-A and Bmp pathways, is widely effective. To further enhance the reproducibility of CHIR-induced cardiomyogenesis, and to ultimately promote myocyte maturation, we are using exogenous growth factors to optimize cardiomyogenic signaling downstream of CHIR induction. As indicated by RNA-seq, induction with CHIR during Day 1 (Days 0-1) was followed by immediate expression of Nodal ligands and receptors, followed later by Bmp ligands and receptors. Co-induction with CHIR and high levels of the Nodal mimetic Activin-A (50-100 ng/ml) during Day 0-1 efficiently induced definitive endoderm, whereas CHIR supplemented with Activin-A at low levels (10 ng/ml) consistently improved cardiomyogenic efficiency, even when CHIR alone was ineffective. Moreover, co-induction using CHIR and low levels of Activin-A apparently increased the rate of cardiomyogenesis, as indicated by the initial appearance of rhythmically beating cells by Day 6 instead of Day 8. By contrast, co-induction with CHIR plus low levels (3-10 ng/ml) of Bmp4 during Day 0-1 consistently and strongly inhibited cardiomyogenesis. These findings, which demonstrate that cardiomyogenic efficacy is improved by optimizing levels of CHIR-induced growth factors when applied in accord with their sequence of endogenous expression, are consistent with the idea that Nodal (Activin-A) levels toggle the entry of cells into the endodermal or mesodermal lineages, while Bmp levels regulate subsequent allocation into mesodermal cell types.
Collapse
|
22
|
Kim PJ, Mahmoudi M, Ge X, Matsuura Y, Toma I, Metzler S, Kooreman NG, Ramunas J, Holbrook C, McConnell MV, Blau H, Harnish P, Rulifson E, Yang PC. Direct evaluation of myocardial viability and stem cell engraftment demonstrates salvage of the injured myocardium. Circ Res 2015; 116:e40-50. [PMID: 25654979 DOI: 10.1161/circresaha.116.304668] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
RATIONALE The mechanism of functional restoration by stem cell therapy remains poorly understood. Novel manganese-enhanced MRI and bioluminescence reporter gene imaging were applied to follow myocardial viability and cell engraftment, respectively. Human-placenta-derived amniotic mesenchymal stem cells (AMCs) demonstrate unique immunoregulatory and precardiac properties. In this study, the restorative effects of 3 AMC-derived subpopulations were examined in a murine myocardial injury model: (1) unselected AMCs, (2) ckit(+)AMCs, and (3) AMC-derived induced pluripotent stem cells (MiPSCs). OBJECTIVE To determine the differential restorative effects of the AMC-derived subpopulations in the murine myocardial injury model using multimodality imaging. METHODS AND RESULTS SCID (severe combined immunodeficiency) mice underwent left anterior descending artery ligation and were divided into 4 treatment arms: (1) normal saline control (n=14), (2) unselected AMCs (n=10), (3) ckit(+)AMCs (n=13), and (4) MiPSCs (n=11). Cardiac MRI assessed myocardial viability and left ventricular function, whereas bioluminescence imaging assessed stem cell engraftment during a 4-week period. Immunohistological labeling and reverse transcriptase polymerase chain reaction of the explanted myocardium were performed. The unselected AMC and ckit(+)AMC-treated mice demonstrated transient left ventricular functional improvement. However, the MiPSCs exhibited a significantly greater increase in left ventricular function compared with all the other groups during the entire 4-week period. Left ventricular functional improvement correlated with increased myocardial viability and sustained stem cell engraftment. The MiPSC-treated animals lacked any evidence of de novo cardiac differentiation. CONCLUSION The functional restoration seen in MiPSCs was characterized by increased myocardial viability and sustained engraftment without de novo cardiac differentiation, indicating salvage of the injured myocardium.
Collapse
Affiliation(s)
- Paul J Kim
- From the Division of Cardiovascular Medicine, Department of Medicine, Stanford University Medical Center, CA (P.J.K., M.M., X.G., Y.M., I.T., S.M., N.G.K., M.V.M., E.R., P.C.Y.); Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (J.R., C.H., H.B.); and Eagle Vision Pharmaceutical Corporation, Exton, PA (P.H.)
| | - Morteza Mahmoudi
- From the Division of Cardiovascular Medicine, Department of Medicine, Stanford University Medical Center, CA (P.J.K., M.M., X.G., Y.M., I.T., S.M., N.G.K., M.V.M., E.R., P.C.Y.); Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (J.R., C.H., H.B.); and Eagle Vision Pharmaceutical Corporation, Exton, PA (P.H.)
| | - Xiaohu Ge
- From the Division of Cardiovascular Medicine, Department of Medicine, Stanford University Medical Center, CA (P.J.K., M.M., X.G., Y.M., I.T., S.M., N.G.K., M.V.M., E.R., P.C.Y.); Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (J.R., C.H., H.B.); and Eagle Vision Pharmaceutical Corporation, Exton, PA (P.H.)
| | - Yuka Matsuura
- From the Division of Cardiovascular Medicine, Department of Medicine, Stanford University Medical Center, CA (P.J.K., M.M., X.G., Y.M., I.T., S.M., N.G.K., M.V.M., E.R., P.C.Y.); Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (J.R., C.H., H.B.); and Eagle Vision Pharmaceutical Corporation, Exton, PA (P.H.)
| | - Ildiko Toma
- From the Division of Cardiovascular Medicine, Department of Medicine, Stanford University Medical Center, CA (P.J.K., M.M., X.G., Y.M., I.T., S.M., N.G.K., M.V.M., E.R., P.C.Y.); Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (J.R., C.H., H.B.); and Eagle Vision Pharmaceutical Corporation, Exton, PA (P.H.)
| | - Scott Metzler
- From the Division of Cardiovascular Medicine, Department of Medicine, Stanford University Medical Center, CA (P.J.K., M.M., X.G., Y.M., I.T., S.M., N.G.K., M.V.M., E.R., P.C.Y.); Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (J.R., C.H., H.B.); and Eagle Vision Pharmaceutical Corporation, Exton, PA (P.H.)
| | - Nigel G Kooreman
- From the Division of Cardiovascular Medicine, Department of Medicine, Stanford University Medical Center, CA (P.J.K., M.M., X.G., Y.M., I.T., S.M., N.G.K., M.V.M., E.R., P.C.Y.); Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (J.R., C.H., H.B.); and Eagle Vision Pharmaceutical Corporation, Exton, PA (P.H.)
| | - John Ramunas
- From the Division of Cardiovascular Medicine, Department of Medicine, Stanford University Medical Center, CA (P.J.K., M.M., X.G., Y.M., I.T., S.M., N.G.K., M.V.M., E.R., P.C.Y.); Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (J.R., C.H., H.B.); and Eagle Vision Pharmaceutical Corporation, Exton, PA (P.H.)
| | - Colin Holbrook
- From the Division of Cardiovascular Medicine, Department of Medicine, Stanford University Medical Center, CA (P.J.K., M.M., X.G., Y.M., I.T., S.M., N.G.K., M.V.M., E.R., P.C.Y.); Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (J.R., C.H., H.B.); and Eagle Vision Pharmaceutical Corporation, Exton, PA (P.H.)
| | - Michael V McConnell
- From the Division of Cardiovascular Medicine, Department of Medicine, Stanford University Medical Center, CA (P.J.K., M.M., X.G., Y.M., I.T., S.M., N.G.K., M.V.M., E.R., P.C.Y.); Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (J.R., C.H., H.B.); and Eagle Vision Pharmaceutical Corporation, Exton, PA (P.H.)
| | - Helen Blau
- From the Division of Cardiovascular Medicine, Department of Medicine, Stanford University Medical Center, CA (P.J.K., M.M., X.G., Y.M., I.T., S.M., N.G.K., M.V.M., E.R., P.C.Y.); Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (J.R., C.H., H.B.); and Eagle Vision Pharmaceutical Corporation, Exton, PA (P.H.)
| | - Phillip Harnish
- From the Division of Cardiovascular Medicine, Department of Medicine, Stanford University Medical Center, CA (P.J.K., M.M., X.G., Y.M., I.T., S.M., N.G.K., M.V.M., E.R., P.C.Y.); Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (J.R., C.H., H.B.); and Eagle Vision Pharmaceutical Corporation, Exton, PA (P.H.)
| | - Eric Rulifson
- From the Division of Cardiovascular Medicine, Department of Medicine, Stanford University Medical Center, CA (P.J.K., M.M., X.G., Y.M., I.T., S.M., N.G.K., M.V.M., E.R., P.C.Y.); Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (J.R., C.H., H.B.); and Eagle Vision Pharmaceutical Corporation, Exton, PA (P.H.)
| | - Phillip C Yang
- From the Division of Cardiovascular Medicine, Department of Medicine, Stanford University Medical Center, CA (P.J.K., M.M., X.G., Y.M., I.T., S.M., N.G.K., M.V.M., E.R., P.C.Y.); Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (J.R., C.H., H.B.); and Eagle Vision Pharmaceutical Corporation, Exton, PA (P.H.).
| |
Collapse
|
23
|
|
24
|
Rodriguez ML, Graham BT, Pabon LM, Han SJ, Murry CE, Sniadecki NJ. Measuring the contractile forces of human induced pluripotent stem cell-derived cardiomyocytes with arrays of microposts. J Biomech Eng 2015; 136:051005. [PMID: 24615475 DOI: 10.1115/1.4027145] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 03/10/2014] [Indexed: 12/31/2022]
Abstract
Human stem cell-derived cardiomyocytes hold promise for heart repair, disease modeling, drug screening, and for studies of developmental biology. All of these applications can be improved by assessing the contractility of cardiomyocytes at the single cell level. We have developed an in vitro platform for assessing the contractile performance of stem cell-derived cardiomyocytes that is compatible with other common endpoints such as microscopy and molecular biology. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) were seeded onto elastomeric micropost arrays in order to characterize the contractile force, velocity, and power produced by these cells. We assessed contractile function by tracking the deflection of microposts beneath an individual hiPSC-CM with optical microscopy. Immunofluorescent staining of these cells was employed to assess their spread area, nucleation, and sarcomeric structure on the microposts. Following seeding of hiPSC-CMs onto microposts coated with fibronectin, laminin, and collagen IV, we found that hiPSC-CMs on laminin coatings demonstrated higher attachment, spread area, and contractile velocity than those seeded on fibronectin or collagen IV coatings. Under optimized conditions, hiPSC-CMs spread to an area of approximately 420 μm2, generated systolic forces of approximately 15 nN/cell, showed contraction and relaxation rates of 1.74 μm/s and 1.46 μm/s, respectively, and had a peak contraction power of 29 fW. Thus, elastomeric micropost arrays can be used to study the contractile strength and kinetics of hiPSC-CMs. This system should facilitate studies of hiPSC-CM maturation, disease modeling, and drug screens as well as fundamental studies of human cardiac contraction.
Collapse
|
25
|
Lundy SD, Gantz JA, Pagan CM, Filice D, Laflamme MA. Pluripotent stem cell derived cardiomyocytes for cardiac repair. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2014; 16:319. [PMID: 24838687 DOI: 10.1007/s11936-014-0319-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OPINION STATEMENT The adult mammalian heart has limited capacity for regeneration, and any major injury such as a myocardial infarction results in the permanent loss of up to 1 billion cardiomyocytes. The field of cardiac cell therapy aims to replace these lost contractile units with de novo cardiomyocytes to restore lost systolic function and prevent progression to heart failure. Arguably, the ideal cell for this application is the human cardiomyocyte itself, which can electromechanically couple with host myocardium and contribute active systolic force. Pluripotent stem cells from human embryonic or induced pluripotent lineages are attractive sources for cardiomyocytes, and preclinical investigation of these cells is in progress. Recent work has focused on the efficient generation and purification of cardiomyocytes, tissue engineering efforts, and examining the consequences of cell transplantation from mechanical, vascular, and electrical standpoints. Here we discuss historical and contemporary aspects of pluripotent stem cell-based cardiac cell therapy, with an emphasis on recent preclinical studies with translational goals.
Collapse
Affiliation(s)
- Scott D Lundy
- Department of Bioengineering, University of Washington, Box 358050, 850 Republican St., Seattle, WA, 98195, USA
| | | | | | | | | |
Collapse
|
26
|
Yang X, Rodriguez M, Pabon L, Fischer KA, Reinecke H, Regnier M, Sniadecki NJ, Ruohola-Baker H, Murry CE. Tri-iodo-l-thyronine promotes the maturation of human cardiomyocytes-derived from induced pluripotent stem cells. J Mol Cell Cardiol 2014; 72:296-304. [PMID: 24735830 DOI: 10.1016/j.yjmcc.2014.04.005] [Citation(s) in RCA: 339] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 03/15/2014] [Accepted: 04/05/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) have great potential as a cell source for therapeutic applications such as regenerative medicine, disease modeling, drug screening, and toxicity testing. This potential is limited, however, by the immature state of the cardiomyocytes acquired using current protocols. Tri-iodo-l-thyronine (T3) is a growth hormone that is essential for optimal heart growth. In this study, we investigated the effect of T3 on hiPSC-CM maturation. METHODS AND RESULTS A one-week treatment with T3 increased cardiomyocyte size, anisotropy, and sarcomere length. T3 treatment was associated with reduced cell cycle activity, manifest as reduced DNA synthesis and increased expression of the cyclin-dependent kinase inhibitor p21. Contractile force analyses were performed on individual cardiomyocytes using arrays of microposts, revealing an almost two-fold higher force per-beat after T3 treatment and also an enhancement in contractile kinetics. This improvement in force generation was accompanied by an increase in rates of calcium release and reuptake, along with a significant increase in sarcoendoplasmic reticulum ATPase expression. Finally, although mitochondrial genomes were not numerically increased, extracellular flux analysis showed a significant increase in maximal mitochondrial respiratory capacity and respiratory reserve capability after T3 treatment. CONCLUSIONS Using a broad spectrum of morphological, molecular, and functional parameters, we conclude that T3 is a driver for hiPSC-CM maturation. T3 treatment may enhance the utility of hiPSC-CMs for therapy, disease modeling, or drug/toxicity screens.
Collapse
Affiliation(s)
- Xiulan Yang
- Department of Pathology, University of Washington, Seattle, WA 98109, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Marita Rodriguez
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98109, USA
| | - Lil Pabon
- Department of Pathology, University of Washington, Seattle, WA 98109, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Karin A Fischer
- Department of Biochemistry, University of Washington, Seattle, WA 98109, USA
| | - Hans Reinecke
- Department of Pathology, University of Washington, Seattle, WA 98109, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Nathan J Sniadecki
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98109, USA; Department of Bioengineering, University of Washington, Seattle, WA 98109, USA
| | | | - Charles E Murry
- Department of Pathology, University of Washington, Seattle, WA 98109, USA; Department of Bioengineering, University of Washington, Seattle, WA 98109, USA; Department of Medicine/Cardiology, University of Washington, Seattle, WA 98109, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
27
|
Pauli A, Norris ML, Valen E, Chew GL, Gagnon JA, Zimmerman S, Mitchell A, Ma J, Dubrulle J, Reyon D, Tsai SQ, Joung JK, Saghatelian A, Schier AF. Toddler: an embryonic signal that promotes cell movement via Apelin receptors. Science 2014; 343:1248636. [PMID: 24407481 DOI: 10.1126/science.1248636] [Citation(s) in RCA: 486] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
It has been assumed that most, if not all, signals regulating early development have been identified. Contrary to this expectation, we identified 28 candidate signaling proteins expressed during zebrafish embryogenesis, including Toddler, a short, conserved, and secreted peptide. Both absence and overproduction of Toddler reduce the movement of mesendodermal cells during zebrafish gastrulation. Local and ubiquitous production of Toddler promote cell movement, suggesting that Toddler is neither an attractant nor a repellent but acts globally as a motogen. Toddler drives internalization of G protein-coupled APJ/Apelin receptors, and activation of APJ/Apelin signaling rescues toddler mutants. These results indicate that Toddler is an activator of APJ/Apelin receptor signaling, promotes gastrulation movements, and might be the first in a series of uncharacterized developmental signals.
Collapse
Affiliation(s)
- Andrea Pauli
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Yu XH, Tang ZB, Liu LJ, Qian H, Tang SL, Zhang DW, Tian GP, Tang CK. Apelin and its receptor APJ in cardiovascular diseases. Clin Chim Acta 2014; 428:1-8. [DOI: 10.1016/j.cca.2013.09.001] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Revised: 08/31/2013] [Accepted: 09/01/2013] [Indexed: 12/29/2022]
|
29
|
Parameswaran S, Kumar S, Verma RS, Sharma RK. Cardiomyocyte culture - an update on the in vitro cardiovascular model and future challenges. Can J Physiol Pharmacol 2013; 91:985-98. [PMID: 24289068 DOI: 10.1139/cjpp-2013-0161] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The success of any work with isolated cardiomyocytes depends on the reproducibility of cell isolation, because the cells do not divide. To date, there is no suitable in vitro model to study human adult cardiac cell biology. Although embryonic stem cells and induced pluripotent stem cells are able to differentiate into cardiomyocytes in vitro, the efficiency of this process is low. Isolation and expansion of human cardiomyocyte progenitor cells from cardiac surgical waste or, alternatively, from fetal heart tissue is another option. However, to overcome various issues related to human tissue usage, especially ethical concerns, researchers use large- and small-animal models to study cardiac pathophysiology. A simple model to study the changes at the cellular level is cultures of cardiomyocytes. Although primary murine cardiomyocyte cultures have their own advantages and drawbacks, alternative strategies have been developed in the last two decades to minimise animal usage and interspecies differences. This review discusses the use of freshly isolated murine cardiomyocytes and cardiomyocyte alternatives for use in cardiac disease models and other related studies.
Collapse
Affiliation(s)
- Sreejit Parameswaran
- a Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
| | | | | | | |
Collapse
|
30
|
Wang W, McKinnie SMK, Patel VB, Haddad G, Wang Z, Zhabyeyev P, Das SK, Basu R, McLean B, Kandalam V, Penninger JM, Kassiri Z, Vederas JC, Murray AG, Oudit GY. Loss of Apelin exacerbates myocardial infarction adverse remodeling and ischemia-reperfusion injury: therapeutic potential of synthetic Apelin analogues. J Am Heart Assoc 2013; 2:e000249. [PMID: 23817469 PMCID: PMC3828798 DOI: 10.1161/jaha.113.000249] [Citation(s) in RCA: 171] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Coronary artery disease leading to myocardial ischemia is the most common cause of heart failure. Apelin (APLN), the endogenous peptide ligand of the APJ receptor, has emerged as a novel regulator of the cardiovascular system. METHODS AND RESULTS Here we show a critical role of APLN in myocardial infarction (MI) and ischemia-reperfusion (IR) injury in patients and animal models. Myocardial APLN levels were reduced in patients with ischemic heart failure. Loss of APLN increased MI-related mortality, infarct size, and inflammation with drastic reductions in prosurvival pathways resulting in greater systolic dysfunction and heart failure. APLN deficiency decreased vascular sprouting, impaired sprouting of human endothelial progenitor cells, and compromised in vivo myocardial angiogenesis. Lack of APLN enhanced susceptibility to ischemic injury and compromised functional recovery following ex vivo and in vivo IR injury. We designed and synthesized two novel APLN analogues resistant to angiotensin converting enzyme 2 cleavage and identified one analogue, which mimicked the function of APLN, to be markedly protective against ex vivo and in vivo myocardial IR injury linked to greater activation of survival pathways and promotion of angiogenesis. CONCLUSIONS APLN is a critical regulator of the myocardial response to infarction and ischemia and pharmacologically targeting this pathway is feasible and represents a new class of potential therapeutic agents.
Collapse
Affiliation(s)
- Wang Wang
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Affiliation(s)
- Annemarieke E. Loot
- From the Institute for Vascular Signalling, Centre for Molecular Medicine and German Centre for Cardiovascular Research (DZHK) partner site Rhine-Main, Goethe University, Theodor-Stern-Kai, Frankfurt am Main, Germany
| | - Ingrid Fleming
- From the Institute for Vascular Signalling, Centre for Molecular Medicine and German Centre for Cardiovascular Research (DZHK) partner site Rhine-Main, Goethe University, Theodor-Stern-Kai, Frankfurt am Main, Germany
| |
Collapse
|
32
|
Kasai A, Ishimaru Y, Higashino K, Kobayashi K, Yamamuro A, Yoshioka Y, Maeda S. Inhibition of apelin expression switches endothelial cells from proliferative to mature state in pathological retinal angiogenesis. Angiogenesis 2013; 16:723-34. [PMID: 23640575 PMCID: PMC3682100 DOI: 10.1007/s10456-013-9349-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 04/24/2013] [Indexed: 11/25/2022]
Abstract
The recruitment of mural cells such as pericytes to patent vessels with an endothelial lumen is a key factor for the maturation of blood vessels and the prevention of hemorrhage in pathological angiogenesis. To date, our understanding of the specific trigger underlying the transition from cell growth to the maturation phase remains incomplete. Since rapid endothelial cell growth causes pericyte loss, we hypothesized that suppression of endothelial growth factors would both promote pericyte recruitment, in addition to inhibiting pathological angiogenesis. Here, we demonstrate that targeted knockdown of apelin in endothelial cells using siRNA induced the expression of monocyte chemoattractant protein-1 (MCP-1) through activation of Smad3, via suppression of the PI3K/Akt pathway. The conditioned medium of endothelial cells treated with apelin siRNA enhanced the migration of vascular smooth muscle cells, through MCP-1 and its receptor pathway. Moreover, in vivo delivery of siRNA targeting apelin, which causes exuberant endothelial cell proliferation and pathological angiogenesis through its receptor APJ, led to increased pericyte coverage and suppressed pathological angiogenesis in an oxygen-induced retinopathy model. These data demonstrate that apelin is not only a potent endothelial growth factor, but also restricts pericyte recruitment, establishing a new connection between endothelial cell proliferation signaling and a trigger of mural recruitment.
Collapse
Affiliation(s)
- Atsushi Kasai
- Interdisciplinary Program for Biomedical Sciences, Institute for Academic Initiatives, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | | | | | | | | | | | | |
Collapse
|