scholarly journals 422Enrichment of cardiac differentiation of mouse pluripotent stem cells by beta 3 adrenoceptor stimulation

2018 ◽  
Vol 114 (suppl_1) ◽  
pp. S103-S103
Author(s):  
V Spinelli ◽  
L Sartiani ◽  
A Laurino ◽  
L Raimondi ◽  
M Calvani ◽  
...  
2019 ◽  
Vol 68 (1) ◽  
pp. 35-47 ◽  
Author(s):  
Praopilas Phakdeedindan ◽  
Piyathip Setthawong ◽  
Narong Tiptanavattana ◽  
Sasitorn Rungarunlert ◽  
Praewphan Ingrungruanglert ◽  
...  

2018 ◽  
Vol 54 (7) ◽  
pp. 513-522 ◽  
Author(s):  
Minh Nguyen Tuyet Le ◽  
Mika Takahi ◽  
Kenshiro Maruyama ◽  
Akira Kurisaki ◽  
Kiyoshi Ohnuma

2016 ◽  
Vol 119 (suppl_1) ◽  
Author(s):  
Hyun-Jai Cho ◽  
Choon-Soo Lee ◽  
Jin-Woo Lee ◽  
Jung-Kyu Han ◽  
Han-Mo Yang ◽  
...  

Backgrounds: The identification of a lineage-specific marker plays a pivotal role in understanding developmental process and is utilized to isolate a certain cell type with high purity for the therapeutic purpose. We here report a new cardiac-specific marker, and demonstrate its functional significance in the cardiac development. Methods and Results: When mouse pluripotent stem cells (ES and iPS cells) were stimulated with BMP4, Activin A, bFGF and VEGF, they differentiated into cardiac cells. To screen cell-surface expressing molecules on cardiac progenitor cells compared to undifferentiated mouse iPS and ES cells, we isolated Flk1+/PDGFRa+ cells at differentiation day 4 and performed microarray analysis. Among candidates, we identified a new G protein-coupled receptor, Latrophilin-2 (LPHN2) whose signaling pathway and its effect on cardiac differentiation is unknown. In sorting experiments under cardiac differentiation condition, LPHN2+ cells derived from pluripotent stem cells strongly expressed cardiac-related genes (Mesp1, Nkx2.5, aMHC and cTnT) and exclusively gave rise to beating cardiomyocytes, as compared with LPHN2- cells. LPHN2-/- mice revealed embryonically lethal and huge defects in cardiac development. Interestingly, LPHN2+/- heterozygotes were alive and fertile. For the purpose of cardiac regeneration, we transplanted iPS-derived LPHN2+ cells into the infarcted heart of adult mice. LPHN2+ cells differentiated into cardiomyocytes, and systolic function of left ventricle was improved and infarct size was reduced. We confirmed LPHN2 expression on human iPS and ES cell-derived cardiac progenitor cells and human heart. Conclusions: We demonstrate that LPHN2 is a functionally significant and cell-surface expressing marker for both mouse and human cardiac progenitor and cardiomyocytes. Our findings provide a valuable tool for isolating cardiac lineage cells from pluripotent stem cells and an insight into cardiac development and regeneration.


2021 ◽  
Vol 129 (Suppl_1) ◽  
Author(s):  
Morteza Mahmoudi ◽  
Phillip C Yang ◽  
Vahid Serpooshan ◽  
Parisa Abadi ◽  
Mahyar Heydarpour

Introduction: Patient-specific human induced pluripotent stem cells (hiPSC)-derived cardiomyocytes (CMs) are increasingly used for in vitro disease modeling and drug screening, as well in vivo regenerative therapies. The cardiac differentiation efficacy of hiPSCs, together with the maturation level of generated CMs, are critical factors in achieving the required numbers of functional patient-specific cardiac muscle cells for clinical applications. Although extensive studies have improved the efficacy of differentiation and maturation processes, the role of cell sex in these processes has not been fully investigated. Hypothesis: Cell sex affects i) the cardiogenic differentiation efficacy of hiPSCs; and ii) maturation processes of hiPSC-CMs. Methods and Results: We have successfully and reproducibly fabricated patterned substrates recapitulating the 3D shape of mature CMs, using photolithography approaches, and demonstrated that the substrate could i) accelerate the differentiation of hiPSCs to CMs, and ii) facilitate maturation and functionality of immature hiPSC-CMs. Male and female hiPSCs, derived from human amniotic mesenchymal stem cells of male and female fetuses, were cultured onto flat (control) vs. patterned substrates. A total of 400 differentiation assays were conducted, 200 per each cell sex, on the flat ( n = 100) and patterned ( n = 100) substrates. A chemically defined approach was used to differentiate the cells toward CMs. On the flat (conventional) substrates, 59% of batches of male and 87% of batches of female hiPSCs differentiated into beating CMs (> 80%). On the patterned substrates, these numbers changed to 83% and 94% of successful differentiations for male and female hiPSCs, respectively. These results indicate the significant effect of substrate-mediated topographical cues on the cardiac differentiation yield of stem cells and the batch-to-batch variation. On both substrate types, female cells demonstrated significantly higher success rates of cardiac differentiation compared to the male cells. In addition, the CMs produced on the patterned substrates demonstrated higher purity than those created on the flat substrates both for male and female cells. Quantitative polymerase chain reaction (qPCR) was used to probe the male and female cell differences in expression of genes related to cardiac maturity, contractility, and Ca 2+ transport (TNNT2, MYH6, MYH7, and CACNA1c) and the outcomes revealed substantially greater expression levels of the maturation genes in differentiated female CMs cultured on the patterned substrates compared to the male cells. Conclusions: These results indicate that male and female hiPSCs and hiPSC-CMs respond differently to the identical substrates in terms of their differentiation and maturation efficacies.


2019 ◽  
Vol 2019 ◽  
pp. 1-12 ◽  
Author(s):  
Jarmon G. Lees ◽  
Anne M. Kong ◽  
Yi C. Chen ◽  
Priyadharshini Sivakumaran ◽  
Damián Hernández ◽  
...  

Human induced pluripotent stem cells (iPSCs) can be differentiated in vitro into bona fide cardiomyocytes for disease modelling and personalized medicine. Mitochondrial morphology and metabolism change dramatically as iPSCs differentiate into mesodermal cardiac lineages. Inhibiting mitochondrial fission has been shown to promote cardiac differentiation of iPSCs. However, the effect of hydrazone M1, a small molecule that promotes mitochondrial fusion, on cardiac mesodermal commitment of human iPSCs is unknown. Here, we demonstrate that treatment with M1 promoted mitochondrial fusion in human iPSCs. Treatment of iPSCs with M1 during embryoid body formation significantly increased the percentage of beating embryoid bodies and expression of cardiac-specific genes. The pro-fusion and pro-cardiogenic effects of M1 were not associated with changes in expression of the α and β subunits of adenosine triphosphate (ATP) synthase. Our findings demonstrate for the first time that hydrazone M1 is capable of promoting cardiac differentiation of human iPSCs, highlighting the important role of mitochondrial dynamics in cardiac mesoderm lineage specification and cardiac development. M1 and other mitochondrial fusion promoters emerge as promising molecular targets to generate lineages of the heart from human iPSCs for patient-specific regenerative medicine.


2016 ◽  
Vol 2016 ◽  
pp. 1-12 ◽  
Author(s):  
Damián Hernández ◽  
Rodney Millard ◽  
Priyadharshini Sivakumaran ◽  
Raymond C. B. Wong ◽  
Duncan E. Crombie ◽  
...  

Background.Human induced pluripotent stem cells (iPSCs) are an attractive source of cardiomyocytes for cardiac repair and regeneration. In this study, we aim to determine whether acute electrical stimulation of human iPSCs can promote their differentiation to cardiomyocytes.Methods. Human iPSCs were differentiated to cardiac cells by forming embryoid bodies (EBs) for 5 days. EBs were then subjected to brief electrical stimulation and plated down for 14 days.Results. In iPS(Foreskin)-2 cell line, brief electrical stimulation at 65 mV/mm or 200 mV/mm for 5 min significantly increased the percentage of beating EBs present by day 14 after plating. Acute electrical stimulation also significantly increased the cardiac gene expression ofACTC1,TNNT2,MYH7, andMYL7. However, the cardiogenic effect of electrical stimulation was not reproducible in another iPS cell line, CERA007c6. Beating EBs from control and electrically stimulated groups expressed various cardiac-specific transcription factors and contractile muscle markers. Beating EBs were also shown to cycle calcium and were responsive to the chronotropic agents, isoproterenol and carbamylcholine, in a concentration-dependent manner.Conclusions. Our results demonstrate that brief electrical stimulation can promote cardiac differentiation of human iPS cells. The cardiogenic effect of brief electrical stimulation is dependent on the cell line used.


Sign in / Sign up

Export Citation Format

Share Document