Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
ACS Appl Bio Mater ; 3(8): 4974-4986, 2020 Aug 17.
Article in English | MEDLINE | ID: mdl-35021675

ABSTRACT

The basic requirement of any engineered scaffold is to mimic the native tissue extracellular matrix (ECM). Despite substantial strides in understanding the ECM, scaffold fabrication processes of sufficient product robustness and bioactivity require further investigation, owing to the complexity of the natural ECM. A promising bioacive platform for cardiac tissue engineering is that of decellularized porcine cardiac ECM (pcECM, used here as a soft tissue representative model). However, this platform's complexity and batch-to-batch variability serve as processing limitations in attaining a robust and tunable cardiac tissue-specific bioactive scaffold. To address these issues, we fabricated 3D composite scaffolds (3DCSs) that demonstrate comparable physical and biochemical properties to the natural pcECM using wet electrospinning and functionalization with a pcECM hydrogel. The fabricated 3DCSs are non-immunogenic in vitro and support human mesenchymal stem cells' proliferation. Most importantly, the 3DCSs demonstrate tissue-specific bioactivity in inducing spontaneous cardiac lineage differentiation in human induced pluripotent stem cells (hiPSC) and further support the viability, functionality, and maturation of hiPSC-derived cardiomyocytes. Overall, this work illustrates the technology to fabricate robust yet tunable 3D scaffolds of tissue-specific bioactivity (with a proof of concept provided for cardiac tissues) as a platform for basic materials science studies and possible future R&D application in regenerative medicine.

2.
Stem Cell Res Ther ; 10(1): 78, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30845990

ABSTRACT

BACKGROUND: Optimal cell type as cell-based therapies for heart failure (HF) remains unclear. We sought to compare the safety and efficacy of direct intramyocardial transplantation of human embryonic stem cell-derived cardiomyocytes (hESC-CMs) and human induced pluripotent stem cell-derived mesenchymal stem cells (hiPSC-MSCs) in a porcine model of HF. METHODS: Eight weeks after induction of HF with myocardial infarction (MI) and rapid pacing, animals with impaired left ventricular ejection fraction (LVEF) were randomly assigned to receive direct intramyocardial injection of saline (MI group), 2 × 108 hESC-CMs (hESC-CM group), or 2 × 108 hiPSC-MSCs (hiPSC-MSC group). The hearts were harvested for immunohistochemical evaluation after serial echocardiography and hemodynamic evaluation and ventricular tachyarrhythmia (VT) induction by in vivo programmed electrical stimulation. RESULTS: At 8 weeks post-transplantation, LVEF, left ventricular maximal positive pressure derivative, and end systolic pressure-volume relationship were significantly higher in the hiPSC-MSC group but not in the hESC-CM group compared with the MI group. The incidence of early spontaneous ventricular tachyarrhythmia (VT) episodes was higher in the hESC-CM group but the incidence of inducible VT was similar among the different groups. Histological examination showed no tumor formation but hiPSC-MSCs exhibited a stronger survival capacity by activating regulatory T cells and reducing the inflammatory cells. In vitro study showed that hiPSC-MSCs were insensitive to pro-inflammatory interferon-gamma-induced human leukocyte antigen class II expression compared with hESC-CMs. Moreover, hiPSC-MSCs also significantly enhanced angiogenesis compared with other groups via increasing expression of distinct angiogenic factors. CONCLUSIONS: Our results demonstrate that transplantation of hiPSC-MSCs is safe and does not increase proarrhythmia or tumor formation and superior to hESC-CMs for the improvement of cardiac function in HF. This is due to their immunomodulation that improves in vivo survival and enhanced angiogenesis via paracrine effects.


Subject(s)
Heart Failure , Immunomodulation , Induced Pluripotent Stem Cells/immunology , Mesenchymal Stem Cells/immunology , Myocytes, Cardiac , Neovascularization, Physiologic/immunology , Animals , Cell Line , Disease Models, Animal , Female , Heart Failure/immunology , Heart Failure/pathology , Heart Failure/therapy , Humans , Induced Pluripotent Stem Cells/pathology , Mesenchymal Stem Cells/pathology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , Myocytes, Cardiac/transplantation , Swine
3.
Stem Cell Res ; 31: 161-173, 2018 08.
Article in English | MEDLINE | ID: mdl-30098478

ABSTRACT

Human pluripotent stem cells (hPSCs) can be a renewable source for generating cardiomyocyte (CM) for treating myocardial infraction. In our previous publication, we described an integrated microcarrier-based wave reactor process for the expansion and differentiation of hPSCs to CMs on a rocker based platform. However, this platform is limited in terms of linear scalability and CMs purity. The present study describes ways to overcome these limitations by the use of a stirred scalable platform and incorporation of an additional lactate based purification step which increases CM purity. Efficient CM differentiation in stirred spinners was achieved by (1) Addition of ascorbic acid (AS) during the differentiation phase which resulted in an increase of 38.42% in CM yield (0.84 ±â€¯0.03 × 106vs 1.17 ±â€¯0.07 × 106 CM/mL for cultures without AS and with AS respectively) and (2) Change of agitation regime to a shorter static intervals one (from 66 min off/6 min on (66/6) to 8 min off/1 min on (8/1)) during the first 3 days of differentiation which resulted in 22% increase in CM yield (1.50 ±â€¯0.10 × 106vs 1.23 ±â€¯0.07 × 106 CM/mL). The combination of AS addition and change in agitation regime resulted in a production yield of 1.50 ±â€¯0.10 × 106 CM/mL which is comparable to that achieved in the rocker platform as described before (1.61 ±â€¯0.36 × 106 CM/mL). Increase in CM purity was achieved by changing of culture medium to RPMI1640 (without glucose) + 5 mM lactate +0.6 mM AS at day 10 of differentiation which resulted in 44.5% increase in CM purity at day 15. The increase in purity of CMs was due to the death of the non-CM cells (~76% of cell death). It is important to note that in the absence of glucose, lactate was consumed at a rate of 0.01 mmol/106 cells/h. Addition of glucose, even in small amounts, during the purification step prevents the process of CM purification, due to the growth of the non-CM cell population. In summary, hPSC (hESC-HES3 and hiPSC-IMR90) can be efficiently differentiated to CMs in a scalable spinner process which integrates 7 days of expansion (3.01 ±â€¯0.51 × 106 to 3.50 ±â€¯0.65 × 106 cells/mL) followed by 10 days of WNT modulated CM differentiation and 5 days of lactate based purification. CM yield of 1.38 ±â€¯0.22 × 106 to 1.29 ±â€¯0.42 × 106 CM/mL with 72.5 ±â€¯8.35% to 83.12 ±â€¯8.73% cardiac troponin-T positive cells were obtained from these cultures.


Subject(s)
Cell Culture Techniques/methods , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/metabolism , Cells, Cultured , Humans
4.
Stem Cell Reports ; 10(6): 1851-1866, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29706502

ABSTRACT

Cardiac differentiation efficiency is hampered by inconsistencies and low reproducibility. We analyzed the differentiation process of multiple human pluripotent stem cell (hPSC) lines in response to dynamic GSK3ß inhibition under varying cell culture conditions. hPSCs showed strong differences in cell-cycle profiles with varying culture confluency. hPSCs with a higher percentage of cells in the G1 phase of the cell cycle exhibited cell death and required lower doses of GSK3ß inhibitors to induce cardiac differentiation. GSK3ß inhibition initiated cell-cycle progression via cyclin D1 and modulated both Wnt signaling and the transcription factor (TCF) levels, resulting in accelerated or delayed mesoderm differentiation. The TCF levels were key regulators during hPSC differentiation with CHIR99021. Our results explain how differences in hPSC lines and culture conditions impact cell death and cardiac differentiation. By analyzing the cell cycle, we were able to select for highly cardiogenic hPSC lines and increase the experimental reproducibility by predicting differentiation outcomes.


Subject(s)
Cell Differentiation/drug effects , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology , Cell Cycle/drug effects , Cell Death/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Pluripotent Stem Cells/metabolism , Reproducibility of Results
5.
J Tissue Eng Regen Med ; 12(4): e2029-e2038, 2018 04.
Article in English | MEDLINE | ID: mdl-29266858

ABSTRACT

Replacing cardiac tissues lost to myocardial infarction remains a therapeutic goal for regenerative therapy in recovering cardiac function. We assessed the feasibility of constructing a macrosized human cardiac tissue construct using pluripotent stem cell-derived cardiomyocytes or control fibroblasts infused fibrin/collagen hydrogel and performed ectopic implantation in peripheral vascular system of a porcine model for 3 weeks. Finally, an optimized vascularized cardiac construct was explanted and grafted onto porcine myocardium for 2 weeks. Myocardial-grafted human cardiac constructs showed a nascent tissue-like organization with aligned cardiomyocytes within the remodelled collagen matrix. Nevertheless, no significant changes in intraconstruct density of cardiomyocytes were observed in the myocardial-grafted constructs (human embryonic stem cell [hESC]-derived cardiomyocyte [n = 4]: 70.5 ± 22.8 troponin I+ cardiomyocytes/high power field [HPF]) as compared to peripherally implanted constructs (hESC-derived cardiomyocyte [n = 4]: 59.0 ± 19.6 troponin I+ cardiomyocytes/HPF; human induced pluripotent stem cell-derived cardiomyocyte [n = 3]: 50.9 ± 8.5 troponin I+ cardiomyocytes/HPF, p = ns). However, the myocardial-grafted constructs showed an increased in neovascularization (194.4 ± 24.7 microvessels/mm2 tissue, p < .05), microvascular maturation (82.8 ± 24.7 mature microvessels/mm2 , p < .05), and tissue-like formation whereas the peripherally implanted constructs of hESC-derived cardiomyocyte (168.3 ± 98.2 microvessels/mm2 tissue and 68.1 ± 33.4 mature microvessels/mm2 ) and human induced pluripotent stem cell-derived cardiomyocyte (86.8 ± 57.4 microvessels/mm2 tissue and 22.0 ± 32.7 mature microvessels/mm2 ) were not significantly different in vascularized response when compared to the control human fibroblasts (n = 3) constructs (65.6 ± 34.1 microvessels/mm2 tissue and 30.7 ± 20.7 mature microvessels/mm2 ). We presented results on technical feasibility and challenges of grafting vascularized centimetre-sized human cardiac construct that may spur novel approaches in cardiac tissue replacement strategy.


Subject(s)
Cell Differentiation , Human Embryonic Stem Cells/metabolism , Hydrogels/chemistry , Induced Pluripotent Stem Cells/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Animals , Cell Line , Collagen/chemistry , Fibrin/chemistry , Human Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Myocardium/cytology , Myocytes, Cardiac/cytology , Swine
6.
Sci Rep ; 7(1): 11498, 2017 09 13.
Article in English | MEDLINE | ID: mdl-28904341

ABSTRACT

A correction to this article has been published and is linked from the HTML version of this paper. The error has been fixed in the paper.

7.
Sci Rep ; 7(1): 2465, 2017 05 26.
Article in English | MEDLINE | ID: mdl-28550304

ABSTRACT

The current study explored the Na+/K+-ATPase (NKA) inhibition-independent proarrhythmic mechanisms of cardiac glycosides (CGs) which are well-known NKA inhibitors. With the cytosolic Ca2+ chelated by EGTA and BAPTA or extracellular Ca2+ replaced by Ba2+, effects of bufadienolides (bufalin (BF) and cinobufagin (CBG)) and cardenolides (ouabain (Oua) and pecilocerin A (PEA)) on the L-type calcium current (I Ca,L) were recorded in heterologous expression Cav1.2-CHO cells and human embryonic stem cell-derived cardiomyocytes (hESC-CMs). BF and CBG demonstrated a concentration-dependent (0.1 to100 µM) I Ca,L inhibition (maximal ≥50%) without and with the NKA activity blocked by 10 µM Oua. BF significantly shortened the action potential duration at 1.0 µM and shortened the extracellular field potential duration at 0.01~1.0 µM. On the other hand, BF and CBG at 100 µM demonstrated a strong inhibition (≥40%) of the rapidly activating component of the delayed rectifier K+ current (I Kr) in heterologous expression HEK293 cells and prolonged the APD of the heart of day-3 Zebrafish larva with disrupted rhythmic contractions. Moreover, hESC-CMs treated with BF (10 nM) for 24 hours showed moderate yet significant prolongation in APD90. In conclusion, our data indicate that CGs particularly bufadienolides possess cytosolic [Ca2+]i- and NKA inhibition- independent proarrhythmic potential through I Ca,L and I Kr inhibitions.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Bufanolides/pharmacology , Calcium/metabolism , Cardiac Glycosides/pharmacology , Myocytes, Cardiac/metabolism , Animals , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , CHO Cells , Calcium Channels, L-Type/metabolism , Cardenolides/pharmacology , Cell Differentiation/drug effects , Cell Line , Cricetulus , ERG1 Potassium Channel/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , HEK293 Cells , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/metabolism , Humans , Larva , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Ouabain/pharmacology , Voltage-Gated Sodium Channel beta-1 Subunit/metabolism , Zebrafish
8.
Sci Rep ; 6: 31068, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27501774

ABSTRACT

The differentiation efficiency of human embryonic stem cells (hESCs) into heart muscle cells (cardiomyocytes) is highly sensitive to culture conditions. To elucidate the regulatory mechanisms involved, we investigated hESCs grown on three distinct culture platforms: feeder-free Matrigel, mouse embryonic fibroblast feeders, and Matrigel replated on feeders. At the outset, we profiled and quantified their differentiation efficiency, transcriptome, transcription factor binding sites and DNA-methylation. Subsequent genome-wide analyses allowed us to reconstruct the relevant interactome, thereby forming the regulatory basis for implicating the contrasting differentiation efficiency of the culture conditions. We hypothesized that the parental expressions of FOXC1, FOXD1 and FOXQ1 transcription factors (TFs) are correlative with eventual cardiomyogenic outcome. Through WNT induction of the FOX TFs, we observed the co-activation of WNT3 and EOMES which are potent inducers of mesoderm differentiation. The result strengthened our hypothesis on the regulatory role of the FOX TFs in enhancing mesoderm differentiation capacity of hESCs. Importantly, the final proportions of cells expressing cardiac markers were directly correlated to the strength of FOX inductions within 72 hours after initiation of differentiation across different cell lines and protocols. Thus, we affirmed the relationship between early FOX TF expressions and cardiomyogenesis efficiency.


Subject(s)
Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Animals , Binding Sites , Cell Culture Techniques/methods , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Collagen , Drug Combinations , Epigenesis, Genetic , Feeder Cells/metabolism , Gene Expression Profiling , Genome-Wide Association Study , Humans , Laminin , Mesoderm/cytology , Mesoderm/metabolism , Mice , Models, Cardiovascular , Proteoglycans , Signal Transduction , Wnt Proteins/metabolism
9.
Biochem Biophys Res Commun ; 473(3): 764-8, 2016 05 06.
Article in English | MEDLINE | ID: mdl-26385176

ABSTRACT

Current methods for human pluripotent stem cells (hPSC) expansion and differentiation can be limited in scalability and costly (due to their labor intensive nature). This can limit their use in cell therapy, drug screening and toxicity assays. One of the approaches that can overcome these limitations is microcarrier (MC) based cultures in which cells are expanded as cell/MC aggregates and then directly differentiated as embryoid bodies (EBs) in the same agitated reactor. This integrated process can be scaled up and eliminate the need for some culture manipulation used in common monolayer and EBs cultures. This review describes the principles of such microcarriers based integrated hPSC expansion and differentiation process, and parameters that can affect its efficiency (such as MC type and extracellular matrix proteins coatings, cell/MC aggregates size, and agitation). Finally examples of integrated process for generation cardiomyocytes (CM) and neural progenitor cells (NPC) as well as challenges to be solved are described.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Neurons/metabolism , Pluripotent Stem Cells/cytology , Cell Culture Techniques , Cell Proliferation , Cell- and Tissue-Based Therapy/methods , Cells, Cultured , Humans , Myocytes, Cardiac/cytology , Stem Cells/cytology
10.
Tissue Eng Part A ; 21(9-10): 1507-19, 2015 May.
Article in English | MEDLINE | ID: mdl-25602926

ABSTRACT

Functional vascularization is a prerequisite for cardiac tissue engineering of constructs with physiological thicknesses. We previously reported the successful preservation of main vascular conduits in isolated thick acellular porcine cardiac ventricular ECM (pcECM). We now unveil this scaffold's potential in supporting human cardiomyocytes and promoting new blood vessel development ex vivo, providing long-term cell support in the construct bulk. A custom-designed perfusion bioreactor was developed to remodel such vascularization ex vivo, demonstrating, for the first time, functional angiogenesis in vitro with various stages of vessel maturation supporting up to 1.7 mm thick constructs. A robust methodology was developed to assess the pcECM maximal cell capacity, which resembled the human heart cell density. Taken together these results demonstrate feasibility of producing physiological-like constructs such as the thick pcECM suggested here as a prospective treatment for end-stage heart failure. Methodologies reported herein may also benefit other tissues, offering a valuable in vitro setting for "thick-tissue" engineering strategies toward large animal in vivo studies.


Subject(s)
Extracellular Matrix/metabolism , Myocardium/metabolism , Neovascularization, Physiologic , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Bioreactors , Coculture Techniques , Feasibility Studies , Human Umbilical Vein Endothelial Cells/cytology , Humans , Mesenchymal Stem Cells/cytology , Myocardium/cytology , Sus scrofa
11.
Stem Cell Res ; 13(2): 202-13, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25043964

ABSTRACT

The development of novel platforms for large scale production of human embryonic stem cells (hESC) derived cardiomyocytes (CM) becomes more crucial as the demand for CMs in preclinical trials, high throughput cardio toxicity assays and future regenerative therapeutics rises. To this end, we have designed a microcarrier (MC) suspension agitated platform that integrates pluripotent hESC expansion followed by CM differentiation in a continuous, homogenous process. Hydrodynamic shear stresses applied during the hESC expansion and CM differentiation steps drastically reduced the capability of the cells to differentiate into CMs. Applying vigorous stirring during pluripotent hESC expansion on Cytodex 1 MC in spinner cultures resulted in low CM yields in the following differentiation step (cardiac troponin-T (cTnT): 22.83±2.56%; myosin heavy chain (MHC): 19.30±5.31%). Whereas the lower shear experienced in side to side rocker (wave type) platform resulted in higher CM yields (cTNT: 47.50±7.35%; MHC: 42.85±2.64%). The efficiency of CM differentiation is also affected by the hydrodynamic shear stress applied during the first 3days of the differentiation stage. Even low shear applied continuously by side to side rocker agitation resulted in very low CM differentiation efficiency (cTnT<5%; MHC<2%). Simply by applying intermittent agitation during these 3days followed by continuous agitation for the subsequent 9days, CM differentiation efficiency can be substantially increased (cTNT: 65.73±10.73%; MHC: 59.73±9.17%). These yields are 38.3% and 39.3% higher (for cTnT and MHC respectively) than static culture control. During the hESC expansion phase, cells grew on continuously agitated rocker platform as pluripotent cell/MC aggregates (166±88×10(5)µm(2)) achieving a cell concentration of 3.74±0.55×10(6)cells/mL (18.89±2.82 fold expansion) in 7days. These aggregates were further differentiated into CMs using a WNT modulation differentiation protocol for the subsequent 12days on a rocking platform with an intermittent agitation regime during the first 3days. Collectively, the integrated MC rocker platform produced 190.5±58.8×10(6) CMs per run (31.75±9.74 CM/hESC seeded). The robustness of the system was demonstrated by using 2 cells lines, hESC (HES-3) and human induced pluripotent stem cell (hiPSC) IMR-90. The CM/MC aggregates formed extensive sarcomeres that exhibited cross-striations confirming cardiac ontogeny. Functionality of the CMs was demonstrated by monitoring the effect of inotropic drug, Isoproterenol on beating frequency. In conclusion, we have developed a simple robust and scalable platform that integrates both hESC expansion and CM differentiation in one unit process which is capable of meeting the need for large amounts of CMs.


Subject(s)
Cell Culture Techniques/instrumentation , Cell Differentiation , Cell Proliferation , Embryonic Stem Cells/physiology , Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/physiology , Biomarkers/metabolism , Cardiotonic Agents/pharmacology , Cells, Cultured , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Equipment Design , Glucose/metabolism , Glutamine/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Isoproterenol/pharmacology , Mechanotransduction, Cellular , Motion , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/metabolism , Phenotype , Stress, Mechanical , Time Factors , Troponin T/metabolism
12.
Biotechnol J ; 9(5): 675-83, 2014 May.
Article in English | MEDLINE | ID: mdl-24619905

ABSTRACT

Cardiomyocytes (CM) derived from human embryonic stem cells (hESC) are used for cardio-toxicity evaluation and tested in many preclinical trials for their potential use in regenerative therapeutics. As more efficient CM differentiation protocols are developed, reliable automated platforms for characterization and detection are needed. An automated time-resolved video analysis and management system (TVAMS) has been developed for the evaluation of hESC differentiation to CM. The system was used for monitoring the kinetics of embryoid bodies (EB) generation (numbers and size) and differentiation into beating EBs (percentage beating area and beating EB count) in two differentiation protocols. We show that the percentage beating areas of EBs (from total area of the EBs) is a more sensitive and better predictor of CM differentiation efficiency than percentage of beating EBs (from total EBs) as the percentage beating areas of EBs correlates with cardiac troponin-T and myosin heavy chain expression levels. TVAMS can also be used to evaluate the effect of drugs and inhibitors (e.g. isoproterenol and ZD7288) on CM beating frequency. TVAMS can reliably replace the commonly practiced, time consuming, manual counting of total and beating EBs during CM differentiation. TVAMS is a high-throughput non-invasive video imaging platform that can be applied for the development of new CM differentiation protocols, as well as a tool to conduct CM toxicology assays.


Subject(s)
Bioengineering/methods , Cell Differentiation/physiology , Time-Lapse Imaging/methods , Video Recording/methods , Embryoid Bodies/cytology , Embryonic Stem Cells/cytology , Humans , Myocytes, Cardiac/cytology
13.
Stem Cell Res Ther ; 5(1): 12, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24444355

ABSTRACT

Human pluripotent stem cell (hPSC)-derived cardiomyocytes have attracted attention as an unlimited source of cells for cardiac therapies. One of the factors to surmount to achieve this is the production of hPSC-derived cardiomyocytes at a commercial or clinical scale with economically and technically feasible platforms. Given the limited proliferation capacity of differentiated cardiomyocytes and the difficulties in isolating and culturing committed cardiac progenitors, the strategy for cardiomyocyte production would be biphasic, involving hPSC expansion to generate adequate cell numbers followed by differentiation to cardiomyocytes for specific applications. This review summarizes and discusses up-to-date two-dimensional cell culture, cell-aggregate and microcarrier-based platforms for hPSC expansion. Microcarrier-based platforms are shown to be the most suitable for up-scaled production of hPSCs. Subsequently, different platforms for directing hPSC differentiation to cardiomyocytes are discussed. Monolayer differentiation can be straightforward and highly efficient and embryoid body-based approaches are also yielding reasonable cardiomyocyte efficiencies, whereas microcarrier-based approaches are in their infancy but can also generate high cardiomyocyte yields. The optimal target is to establish an integrated scalable process that combines hPSC expansion and cardiomyocyte differentiation into a one unit operation. This review discuss key issues such as platform selection, bioprocess parameters, medium development, downstream processing and parameters that meet current good manufacturing practice standards.


Subject(s)
Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology , Stem Cell Transplantation/methods , Tissue Engineering/methods , Humans , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/metabolism , Stem Cell Research , Stem Cell Transplantation/standards , Tissue Engineering/standards
14.
J Cardiovasc Transl Res ; 6(6): 989-99, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24081385

ABSTRACT

While human embryonic stem cells (hESCs) can differentiate into functional cardiomyocytes, their immature phenotypes limit their therapeutic application for myocardial regeneration. We sought to determine whether electrical stimulation could enhance the differentiation and maturation of hESC-derived cardiomyocytes. Cardiac differentiation was induced in a HES3 hESC line via embryoid bodies formation treated with a p38 MAP kinase inhibitor. Detailed molecular and functional analysis were performed in those hESC-derived cardiomyocytes cultured for 4 days in the absence or presence of electrical field stimulation (6.6 V/cm, 1 Hz, and 2 ms pulses) using an eight-channel C-Pace stimulator (Ion-Optics Co., MA). Upon electrical stimulation, quantitative polymerase chain reaction demonstrated significant upregulation of cardiac-specific gene expression including HCN1, MLC2V, SCN5A, SERCA, Kv4.3, and GATA4; immunostaining and flow cytometry analysis revealed cellular elongation and an increased proportion of troponin-T positive cells (6.3 ± 1.2% vs. 15.8 ± 2.1%; n = 3, P < 0.01). Electrophysiological studies showed an increase in the proportion of ventricular-like hESC-derived cardiomyocytes (48 vs. 29%, P < 0.05) with lengthening of their action potential duration at 90% repolarization (387.7 ± 35.35; n = 11 vs. 291.8 ± 20.82; n = 10, P < 0.05) and 50% repolarization (313.9 ± 27.94; n = 11 vs. 234.0 ± 16.10; n = 10, P < 0.05) after electrical stimulation. Nonetheless, the membrane diastolic potentials and action potential upstrokes of different hESC-derived cardiomyocyte phenotypes, and the overall beating rate remained unchanged (all P > 0.05). Fluorescence confocal imaging revealed that electrical stimulation significantly increased both spontaneous and caffeine-induced calcium flux in the hESC-derived cardiomyocytes (approximately 1.6-fold for both cases; P < 0.01). In conclusion, electrical field stimulation increased the expression of cardiac-specific genes and the yield of differentiation, promoted ventricular-like phenotypes, and improved the calcium handling of hESC-derived cardiomyocytes.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/physiology , Myocytes, Cardiac/physiology , Calcium Signaling , Cell Line , Cell Lineage , Electric Stimulation , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/enzymology , Gene Expression Profiling , Gene Expression Regulation , Humans , Membrane Potentials , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Phenotype , Protein Kinase Inhibitors/pharmacology , Time Factors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
15.
World J Stem Cells ; 5(3): 86-97, 2013 Jul 26.
Article in English | MEDLINE | ID: mdl-23904910

ABSTRACT

AIM: To development of an improved p38 MAPK inhibitor-based serum-free medium for embryoid body cardiomyocyte differentiation of human pluripotent stem cells. METHODS: Human embryonic stem cells (hESC) differentiated to cardiomyocytes (CM) using a p38 MAPK inhibitor (SB203580) based serum-free medium (SB media). Nutrient supplements known to increase cell viability were added to SB medium. The ability of these supplements to improve cardiomyogenesis was evaluated by measurements of cell viability, total cell count, and the expression of cardiac markers via flow cytometry. An improved medium containing Soy hydrolysate (HySoy) and bovine serum albumin (BSA) (SupSB media) was developed and tested on 2 additional cell lines (H1 and Siu-hiPSC). Characterization of the cardiomyocytes was done by immunohistochemistry, electrophysiology and quantitative real-time reverse transcription-polymerase chain reaction. RESULTS: hESC cell line, HES-3, differentiating in SB medium for 16 d resulted in a cardiomyocyte yield of 0.07 ± 0.03 CM/hESC. A new medium (SupSB media) was developed with the addition of HySoy and BSA to SB medium. This medium resulted in 2.6 fold increase in cardiomyocyte yield (0.21 ± 0.08 CM/hESC). The robustness of SupSB medium was further demonstrated using two additional pluripotent cell lines (H1, hESC and Siu1, hiPSC), showing a 15 and 9 fold increase in cardiomyocyte yield respectively. The age (passage number) of the pluripotent cells did not affect the cardiomyocyte yields. Embryoid body (EB) cardiomyocytes formed in SupSB medium expressed canonical cardiac markers (sarcomeric α-actinin, myosin heavy chain and troponin-T) and demonstrated all three major phenotypes: nodal-, atrial- and ventricular-like. Electrophysiological characteristics (maximum diastolic potentials and action potential durations) of cardiomyocytes derived from SB and SupSB media were similar. CONCLUSION: The nutrient supplementation (HySoy and BSA) leads to increase in cell viability, cell yield and cardiac marker expression during cardiomyocyte differentiation, translating to an overall increase in cardiomyocyte yield.

16.
Bioorg Med Chem Lett ; 23(11): 3300-3, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23602399

ABSTRACT

The p38α mitogen-activated protein kinase (MAPK) inhibitor SB203580 had been reported to enhance the cardiomyogenesis of human embryonic stem cells (hESCs). To investigate if tri-substituted imidazole analogues of SB203580 are equally effective inducers for cardiomyogenesis of hESCs, and if there is a correlation between p38α MAPK inhibition and cardiomyogenesis, we designed and synthesized a series of novel tri-substituted imidazoles with a range of p38α MAPK inhibitory activities. Our studies demonstrated that suitably designed analogues of SB203580 can also be inducers of cardiomyogenesis in hESCs and that cell growth is affected by changes in the imidazole structures.


Subject(s)
Embryonic Stem Cells/cytology , Imidazoles/chemistry , Pyridines/chemistry , Cell Differentiation/drug effects , Humans , Imidazoles/metabolism , Imidazoles/pharmacology , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/metabolism , Myocytes, Cardiac/cytology , Protein Binding , Pyridines/metabolism
17.
Curr Protoc Stem Cell Biol ; Chapter 1: Unit1D.7, 2012 May.
Article in English | MEDLINE | ID: mdl-22605644

ABSTRACT

We have developed an improved cardiomyocyte differentiation protocol where we stabilized embryoid bodies (EB) in serum- and insulin-free medium (bSFS) supplemented with p38 MAP kinase inhibitor (SB203580) by addition of 10 µm laminin-coated positively charged (protamine sulfate derivatized TSKgel Tresyl-5PW) microcarriers. This protocol achieved a maximum 3-fold cell expansion, differentiation efficiency of 20%, and an overall cardiomyocyte yield of 3 × 105 CM/ml in static conditions. In comparison, EB cultures achieved 1.5-fold cell expansion, differentiation efficiency of 15%, and an overall cardiomyocyte yield of 1.1 × 105 CM/ml. The scalability of this platform was demonstrated in suspended spinner cultures, producing a maximum of 2.14 × 105 CM/ml in 50-ml cultures. This yield is two-fold higher than the control static EB-based platform (1.1 × 105 CM/ml), and seven-fold higher than yields reported in literature, 3.1-9 × 104 CM/ml. The robustness of this protocol was tested with HES-3 and H1 cell lines.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Embryonic Stem Cells/cytology , Microspheres , Myocytes, Cardiac/cytology , Animals , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Embryonic Stem Cells/drug effects , Flow Cytometry , Humans , Laminin/pharmacology , Mice , Myocytes, Cardiac/drug effects
18.
Stem Cell Res ; 7(3): 198-209, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21907163

ABSTRACT

Improving cardiac differentiation of human pluripotent stem cells is mandatory to provide functional heart muscle cells for novel therapies. Here, we have investigated the enhancing effect of the small molecule SB203580, a p38 MAPK inhibitor, on cardiomyogenesis in hESC by monitoring the phosphorylation patterns of the major MAPK pathway components p38, JNK and ERK by western immunoblotting. A remarkable drop in phosphorylation levels of all three MAPK pathways was induced after overnight embryoid body (EB) formation. Upon further differentiation, phosphorylation dynamics in EBs were specifically altered by distinct inhibitor concentrations. At 5µM of SB203580, cardiomyogenesis was most efficient and associated with the expected p38 pathway inhibition. In parallel, JNK activation was observed suggesting a regulatory interlink between these pathways in hESC ultimately supporting cardiac differentiation. In contrast, moderately elevated SB203580 concentrations (15-30µM) resulted in complete disruption of cardiomyogenesis which was associated with prominent inhibition of ERK and further elevated JNK activity. We propose that a tightly-balanced pattern in MAPK phosphorylation is important for early mesoderm and subsequent cardiomyocyte formation. Our data provide novel insights into molecular consequences of small molecule supplementation in hESC differentiation, emphasizing the role of MAPK-signaling.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/enzymology , Mitogen-Activated Protein Kinases/metabolism , Myocardium/cytology , Myocardium/enzymology , Biomarkers/metabolism , Cell Differentiation/drug effects , Culture Media/pharmacology , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Embryonic Stem Cells/drug effects , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Humans , Imidazoles/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mesoderm/drug effects , Mesoderm/metabolism , Models, Biological , Organogenesis/drug effects , Organogenesis/genetics , Phosphorylation/drug effects , Pyridines/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
19.
Tissue Eng Part C Methods ; 16(6): 1609-19, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20590381

ABSTRACT

A scalable platform for human embryonic stem cell (hESC)-derived cardiomyocyte (CM) production can provide a readily available source of CMs for cell therapy, drug screening, and cardiotoxicity tests. We have designed and optimized a scalable platform using microcarrier cultures in serum-free media supplemented with SB203580 mitogen-activated protein kinase-inhibitor. Different microcarriers (DE-53, Cytodex-1 and 3, FACT, and TOSOH-10) were used to investigate the effects of type, size, shape, and microcarrier concentrations on the differentiation efficiency. hESCs propagated on TOSOH-10 (protamine derivatized 10-µm beads) at the concentration of 0.125 mg/mL produced 80% beating aggregates, threefold cell expansion, and 20% of CMs (determined by fluorescence-activated cell sorting for myosin heavy chain and α-actinin expression). The ratio of CM/hESC seeded in this system was 0.62 compared to 0.22 in the embryoid body control cultures. The platform robustness has been tested with HES-3 and H1 cell lines, and its scalability was demonstrated in suspended spinner cultures. However, spinner culture yields dropped to 0.33 CM/hESC probably due to shear stress causing some cell death. Cells dissociated from differentiated aggregates showed positive staining for cardio-specific markers such as α-actinin, myosin heavy and light chain, troponin I, desmin, and emilin-2. Finally, CM functionality was also shown by QT-prolongation (QTempo) assay with/without Astemizole. This study represents a new scalable bioprocessing system for CM production using reagents that can comply with Good Manufacturing Practice.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Microtechnology , Myocytes, Cardiac/physiology , Tissue Scaffolds , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cells, Cultured , Efficiency , Humans , Microfluidic Analytical Techniques , Microspheres , Myocytes, Cardiac/cytology , Osmolar Concentration , Suspensions , Tissue Scaffolds/chemistry
20.
Am J Obstet Gynecol ; 188(3): 617-22, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12634630

ABSTRACT

OBJECTIVE: The purpose of this study was to examine the relationship between gestational age and outcomes of outborn versus inborn preterm infants. STUDY DESIGN: Multivariable logistic regression analysis was used to examine gestational age-specific, risk-adjusted outcomes of 2962 singleton infants who were born at <32 weeks of gestation who were admitted to 17 Canadian neonatal intensive care units from 1996 through 1997. RESULTS: The risk-adjusted incidence was significantly (P <.05) higher among outborn versus inborn infants for mortality rates (odds ratio, 2.2) and > or =grade 3 intraventricular hemorrhage (odds ratio, 2.1) at < or =26 weeks of gestation and for chronic lung disease (odds ratio, 1.7) at 27 to 29 weeks of gestation. Outcomes of outborn and inborn infants at 30 to 31 weeks of gestation were not significantly different. CONCLUSION: The short-term benefit of preterm birth at tertiary centers is related inversely to gestational age and may not extend beyond 29 weeks of gestation.


Subject(s)
Intensive Care Units, Neonatal , Parturition , Cerebral Hemorrhage/epidemiology , Chronic Disease , Female , Gestational Age , Humans , Incidence , Infant Mortality , Infant, Newborn , Infant, Newborn, Diseases/epidemiology , Infant, Premature , Inpatients , Lung Diseases/epidemiology , Outpatients , Pregnancy , Pregnancy Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...