Abstract 154: Generation of Purified Cardiomyocytes from Pluripotent Stem Cells Using Molecular Beacons

2012 ◽  
Vol 111 (suppl_1) ◽  
Author(s):  
Kiwon Ban ◽  
Brian Wile ◽  
Sangsung Kim ◽  
Jaemin Byun ◽  
Talib Saafir ◽  
...  

Background: While various methods for generating cardiomyocytes (CMs) from pluripotent stem cells (PSCs) including both embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have been reported, all available methods are only allowed to produce heterogeneous population of CM mixed with non-CM cells. Therefore, strategies to enrich pure CMs for scientific and clinical applications have been highly required. Hence, we developed a novel system in which CMs can be purified by cardiac specific molecular beacons (MBs). MBs are dual-labeled antisense nano-scale probes that emit a fluorescence signal when hybridized to target mRNAs. We hypothesized that MBs targeted to CM specific mRNAs can identify CMs and allow isolation of purified CMs by fluorescence-activated cell sorting (FACS). Methods and results: Five MBs targeting distinct sites on either cardiac troponin T (cTNT) or α/β myosin heavy chain (α/βMHC) were designed and characterized in various cell types. To find the optimal MB that can selectively identifying CMs, each MB was delivered into HL-1 CMs, an immortalized mouse CM cell line, smooth muscle cells, endothelial cells, mouse ESCs and fibroblasts and its specificity was determined by flow cytometry. As a result, two MBs identified MB+ cells up to 98% from HL-1 CMs but lower than 10% of the non-CM cells suggesting these MBs are CM specific. Subsequently, the selected MBs were delivered into both mouse and human PSCs derived CMs and 41 to 49% of the cells were identified as an MB+ population. Interestingly, the rate of MB+ cells was similar to CM quantification determined by cTNT intracellular flow cytometry. Finally, we determined whether cell sorting with cardiac-specific MBs can enrich CMs from the heterogeneous mouse and human PSC cultures and found that ∼97% of MB-based sorted CMs expressed cTNT. These enriched cells were further cultured and their CM identity was verified by immunocytochemistry and qRT-PCR analysis. Ca2+ transient analysis further confirmed that these purified CMs displayed functional CM characteristics Conclusion: Using cardiac specific MBs, we were able to obtain highly purified CMs. These purified CMs and the system can be highly useful for clinical applications as well as drug discovery.

Circulation ◽  
2014 ◽  
Vol 130 (suppl_2) ◽  
Author(s):  
Shayne Boucher ◽  
Stacy Jones ◽  
David Kuninger ◽  
Mohan Vemuri

Objective: Current protocols for differentiating pluripotent stem cells (PSCs) have led to heterogeneous results, varying purity levels, and long lead times for generation of cardiomyocytes. We hypothesized that a simplified and rapid cardiomyocyte differentiation media system can be developed in a scalable workflow to enable generation of large numbers of consistent, spontaneously active cardiomyocytes that could be used in basic and translational research. Methods: High quality PSCs were maintained under xenofree, feeder-free culture conditions. At time of passaging, PSC were dissociated with 0.5 mM EDTA, seeded on 1:100 Geltrex © -coated surface as small clusters at ~0.5 to 1 x 10 5 /well of a 12-well plate and maintained for four days under serum-free condition. After reaching target confluence of ~60 to 80%, an induction media was added for two days followed by addition of a second induction media for two days. After the induction step, the media was replaced with maintenance media and re-fed every other day for up to five weeks. PSC-derived cardiomyocytes were analyzed by morphology, gene expression, flow cytometry, immunocytochemistry and multi-electrode array (MEA). Results: We observed individual beating cells by Day 7 and contracting syncytia by Day 10. An over 100 fold increase in cell number was noted from the time of plating to generation of contracting syncytia of cardiomyocytes. Quantitative flow cytometry detected populations of troponin T type 2 (TNNT2)-immunoreactive cells that reached as high as 96.6%. Number of TNNT2-positive cells dropped by 20% when induced at 90% versus 60% confluency. PCR studies confirmed expression of mesoderm (T, MIXL1, MESP1), cardiac mesoderm (ISL1, GATA4, MEF2C) and mature cardiomyocyte genes (NKX2.5, TNNT2, MYH6). Immunocytochemistry studies verified expression of cardiac markers NKX2.5,GATA4, MEF2C, TNNT2 and MYH6. Initial MEA studies corroborated the presence of electrically active cells. Conclusions: We conclude that a simplified complete differentiation media system could serve as a standardized culture system for generating large numbers of consistent, spontaneously active cardiomyocytes for basic and translational research studies.


Circulation ◽  
2014 ◽  
Vol 130 (suppl_2) ◽  
Author(s):  
Kiwon Ban ◽  
Brian Wile ◽  
Kyu-Won Cho ◽  
Sangsung Kim ◽  
Jason Singerd ◽  
...  

Background: Ventricular cardiomyocytes (CMs) are an ideal cell type for cardiac cell therapy since they are the main cells generating cardiac forces. However, isolating them from differentiating pluripotent stem cells (PSCs) has been challenging due to the lack of specific surface markers. Here we show that ventricular CMs can be purified from differentiating mouse embryonic stem cells (mESCs) using molecular beacons (MBs) targeting specific intracellular mRNAs. MBs are dual-labeled oligonucleotide hairpin probes that emit a fluorescence signal when hybridized to target mRNAs, allowing isolation of specific target cells by fluorescence activated cell sorting (FACS) with high specificity and sensitivity. Methods and Results: We generated three different MBs (IRX4-1, -2, -3) designed to target specific regions of mRNAs of iroquois homeobox protein 4 (Irx4), a specific transcription factor for ventricular CMs. Among three IRX4 MBs, IRX4-2 MB demonstrated the highest sensitivity and specificity, thus IRX4-2 MB was selected to purify mESC-derived ventricular CMs. Subsequently, IRX4-2 MBs were delivered into cardiomyogenically differentiating mESC cultures and cells showing strong signals from IRX4-2 MBs were FACS-sorted. Flow cytometry demonstrated that 92~97% of IRX4-2 MB-positive cells expressed a marker for ventricular CMs myosin light chain 2 ventricular isoform (Myl2) as well as cardiac troponin 2 (Tnnt2). Importantly, higher than 98% of IRX4-2 MB-positive cells displayed ventricular CM-like action potentials during electrophysiological analyses. These IRX4-2 MB-based purified ventricular CMs continuously maintained their CM characteristics verified by synchronous beating, Ca2+ transient, and expression of ventricular CM-specific proteins. Conclusions: We established a novel MB-based cell sorting system targeting a transcription factor that is specific for ventricular CM to generate homogeneous and functional ventricular CMs. This is the first report to show the feasibility of isolating pure ventricular CMs without modifying host genes, and this platform will be useful for therapeutic applications, disease modeling, and drug discovery.


Science ◽  
2013 ◽  
Vol 341 (6146) ◽  
pp. 651-654 ◽  
Author(s):  
Pingping Hou ◽  
Yanqin Li ◽  
Xu Zhang ◽  
Chun Liu ◽  
Jingyang Guan ◽  
...  

Pluripotent stem cells can be induced from somatic cells, providing an unlimited cell resource, with potential for studying disease and use in regenerative medicine. However, genetic manipulation and technically challenging strategies such as nuclear transfer used in reprogramming limit their clinical applications. Here, we show that pluripotent stem cells can be generated from mouse somatic cells at a frequency up to 0.2% using a combination of seven small-molecule compounds. The chemically induced pluripotent stem cells resemble embryonic stem cells in terms of their gene expression profiles, epigenetic status, and potential for differentiation and germline transmission. By using small molecules, exogenous “master genes” are dispensable for cell fate reprogramming. This chemical reprogramming strategy has potential use in generating functional desirable cell types for clinical applications.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 347-347
Author(s):  
Dan Ran ◽  
Wei-Jong Shia ◽  
Miao-Chia Lo ◽  
Junbao Fan ◽  
David A. Knorr ◽  
...  

Abstract Abstract 347 Both human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs) are pluripotent stem cells (hPSCs) with potential to differentiate into all types of somatic cells. Patients suffering from blood disorders can be cured with hematopoietic cell transplantations (HCT). Technical advancements in hPSC production and handling have revolutionized their potential applications in regenerative medicine and provided enormous hope for patients who may need HCT. hiPSCs derived from autologous cells could provide unlimited leukocyte antigen matched blood cells on a patient-specific basis. A remaining hurdle in this process remains the need for efficient and effective generation of specific blood cells from hPSCs for therapeutic use. Transcription factors play key roles in regulating maintenance, expansion, and differentiation of blood cells from hPSCs. Studies have shown that transcription factor RUNX1 is required for the formation of definitive blood cells. There are several alternatively spliced isoforms of the RUNX1 protein, including the shortest form RUNX1a and two longer forms RUNX1b and RUNX1c. Based on known properties of RUNX1 proteins, we hypothesized that RUNX1a promotes the production of therapeutic hematopoietic stem cells from hPSCs. By employing ectopic expression of RUNX1a on different human ESC and iPSC lines (H9, BC1, iCB5) under a defined hematopoietic differentiation system, we aimed to identify function of RUNX1a on lineage commitment and molecular mechanisms of RUNX1 activity in differentiation of PSCs to hematopoietic cells. We demonstrated that expression of endogenous RUNX1a parallels lineage commitment and hematopoietic emergence from hPSCs. During differentiation process RUNX1a enhanced the expression of several mesoderm and hematopoietic differentiation related factors, including KDR, SCL, GATA2, and PU.1. In addition, over-expression of RUNX1a in embryoid bodies (EBs) showed more efficient and earlier emergence of typical sac structures, which predicts cell lineage commitment and germ layer development at the early stage of EB differentiation. At day 7, EBs derived from hPSCs was dissociated into single cells for flow cytometry analysis. The mean frequency of CD31+CD34+CD45− and total CD34+ cells with hemato-endothelial cell features are 35.1% and 67.1% from RUNX1a-overexpressing EBs, and 8.7% and 24.1% from vector control EBs. Immunohistochemistry analysis of EBs at day 9 of differentiation confirmed that expression of RUNX1a accelerated mesoderm commitment and emergence of hemato-endothelial precursors. Flow cytometry analysis on EBs collected at days 9, 11, 13 showed that ectopic RUNX1a induced a robust increase in the frequency of hematopoietic progenitor cells in all hPSC lines examined. At Day 9, RUNX1a-overexpressing EBs generated 48.5% CD43+CD45+ cells, 45.1% CD34+CD45+ cells, and 8.5 folds higher CD43+ cells than vector EBs. Later at Day 13, 80% CD45+ and 75% CD43+/CD34+CD45+ hematopoietic stem/progenitor cells (HSPCs) achieved from dissociated EBs. In liquid culture, RUNX1a HSPC showed strong expansion and high percentage of CD235a+CD45− (20%) and CD71+CD235a+ (16%), markers for erythroid populations. Flow cytometry and western blots on RUNX1a-EB formed colonies showed significantly higher β-globin production than that of the vector, suggesting expression of RUNX1a in HSPC enhanced definitive hematopoiesis. RUNX1a-hPSCs derived HSPCs possess self-renewal capability and are capable of differentiating into multi-lineages ex vivo. Furthermore HSPCs generated from RUNX1a-EBs possessed the capacity of interacting with surrogate niche and showed long-term repopulation ability under LTC-IC (Long-Term Culture-Initiating Cell Assay) condition. Colonies generated from HSPC of RUNX1a-EBs after 3 week bulk LTC-IC culture showed 300 folds higher than vector control. RUNX1a-hPSCs derived CD34+CD45+ cells could maintain a non-adherent population in ouldCD45+ sEBsND THIS SENTENCE5 week culture on stromal cell M210. In summary we identified that RUNX1a enhances derivation of definitive hematopoietic cells from human PSCs. Our study provides an important and useful system to enhance specificity and efficiency of generating functional blood cells and further differentiated cells from human PSCs, which may provide valuable source for future clinical applications in patients with hematologic disorders. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 7 (1) ◽  
Author(s):  
Jeong-Eun Yoo ◽  
Dongjin R. Lee ◽  
Sanghyun Park ◽  
Hye-Rim Shin ◽  
Kun Gu Lee ◽  
...  

AbstractSuccessful cell therapy for Parkinson’s disease (PD) requires large numbers of homogeneous ventral mesencephalic dopaminergic (vmDA) precursors. Enrichment of vmDA precursors via cell sorting is required to ensure high safety and efficacy of the cell therapy. Here, using LMX1A-eGFP knock-in reporter human embryonic stem cells, we discovered a novel surface antigen, trophoblast glycoprotein (TPBG), which was preferentially expressed in vmDA precursors. TPBG-targeted cell sorting enriched FOXA2+LMX1A+ vmDA precursors and helped attain efficient behavioral recovery of rodent PD models with increased numbers of TH+, NURR1+, and PITX3+ vmDA neurons in the grafts. Additionally, fewer proliferating cells were detected in TPBG+ cell-derived grafts than in TPBG− cell-derived grafts. Our approach is an efficient way to obtain enriched bona fide vmDA precursors, which could open a new avenue for effective PD treatment.


2016 ◽  
Vol 119 (suppl_1) ◽  
Author(s):  
Myong-Ho Jeong ◽  
Young-Eun Leem ◽  
Hyun-Ji Kim ◽  
Kyungjin Kang ◽  
Hana Cho ◽  
...  

Sonic hedgehog (Shh) signaling plays an important role for early heart development, such as heart looping and cardiomyogenesis of pluripotent stem cells. A multifunctional receptor Cdo functions as a Shh coreceptor together with Boc and Gas1 to activate Shh signaling and these coreceptors seem to play compensatory roles in early heart development. Thus in this study, we examined the role of Cdo in cardiomyogenesis by utilizing an in vitro differentiation of pluripotent stem cells. Here we show that Cdo is required for efficient cardiomyogenesis of pluripotent stem cells by activation of Shh signaling. Cdo is induced concurrently with Shh signaling activation upon induction of cardiomyogenesis of P19 embryonal carcinoma (EC) cells. Cdo -depleted P19 EC and Cdo –/– mouse embryonic stem (ES) cells display decreased expression of key cardiac regulators, including Gata4, Nkx2.5 and Mef2c and this decrease coincides with reduced Shh signaling activities. Furthermore Cdo deficiency causes a stark reduction in formation of mature contractile cardiomyocytes. This defect in cardiomyogenesis is overcome by reactivation of Shh signaling at the early specification stage of cardiomyogenesis. The Shh agonist treatment restores differentiation capacities of Cdo -deficient ES cells into contractile cardiomyocytes by recovering both the expression of early cardiac regulators and structural genes such as cardiac troponin T and Connexin 43. Therefore Cdo is required for efficient cardiomyogenesis of pluripotent stem cells and an excellent target to improve the differentiation potential of stem cells for generation of transplantable cells to treat cardiomyopathies.


2020 ◽  
Vol 15 (4) ◽  
pp. 301-307 ◽  
Author(s):  
Gaifang Wang ◽  
Maryam Farzaneh

Primary Ovarian Insufficiency (POI) is one of the main diseases causing female infertility that occurs in about 1% of women between 30-40 years of age. There are few effective methods for the treatment of women with POI. In the past few years, stem cell-based therapy as one of the most highly investigated new therapies has emerged as a promising strategy for the treatment of POI. Human pluripotent stem cells (hPSCs) can self-renew indefinitely and differentiate into any type of cell. Human Embryonic Stem Cells (hESCs) as a type of pluripotent stem cells are the most powerful candidate for the treatment of POI. Human-induced Pluripotent Stem Cells (hiPSCs) are derived from adult somatic cells by the treatment with exogenous defined factors to create an embryonic-like pluripotent state. Both hiPSCs and hESCs can proliferate and give rise to ectodermal, mesodermal, endodermal, and germ cell lineages. After ovarian stimulation, the number of available oocytes is limited and the yield of total oocytes with high quality is low. Therefore, a robust and reproducible in-vitro culture system that supports the differentiation of human oocytes from PSCs is necessary. Very few studies have focused on the derivation of oocyte-like cells from hiPSCs and the details of hPSCs differentiation into oocytes have not been fully investigated. Therefore, in this review, we focus on the differentiation potential of hPSCs into human oocyte-like cells.


2009 ◽  
Vol 1 (1) ◽  
pp. 76-82 ◽  
Author(s):  
Mark Denham ◽  
Jessie Leung ◽  
Cheryl Tay ◽  
Raymond C.B. Wong ◽  
Peter Donovan ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document