Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Stem Cells Dev ; 23(5): 467-76, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24192059

ABSTRACT

Cardiac cell replacement therapy by using human embryonic stem cell (hESC) derivatives remains a potential approach to regenerate myocardium. The major hurdles to clinical application of this technology are immunogenicity and post-transplantation cell death. Here we examined the effects of calcineurin-targeting immunosuppressants cyclosporine A (CsA) and FK506, as well as rapamycin and a selective inhibitor of calcineurin-binding downstream nuclear factor of activated T-cell (NFAT) transcription factor VIVIT on the proliferative activity, function, and survival of hESC-derived cardiomyocytes (hESC-CM) and endothelial cells (hESC-EC) in culture. As shown by automated microscopy, treatments with CsA, FK506, and rapamycin all decreased proliferation, reducing the percentage of hESC-CM and hESC-EC with the mitotic marker Ki67(+) by as much as 60% and 74%, respectively. Administration of the cell permeable analogue 11R-VIVIT protein did not modulate their proliferative activity. All immunosuppressants reversed the proapoptotic effect of chelerythrine in hESC-CM demonstrating an inhibitory role of calcineurin/NFAT and mammalian target of rapamycin (mTOR) pathways in hESC-CM survival (using apoptotic marker caspase-3), whereas the protection was less obvious in hESC-EC exposed to H2O2. Immunosuppressants did not affect cell viability in hESC-EC. Our results show that immunosuppressants reduce proliferation, while offsetting cell loss to a smaller extent by reduction in apoptosis of hESC-CM. Immunosuppressant therapy would be compatible with stem cell transplantation, but the resulting reduction in graft expansion capabilities would potentially necessitate implantation of increased cell numbers when immunosuppressants are given. The effects of NFAT-binding immunosuppressant molecules, which do not affect hESC-CM proliferation, may point the way forward for new classes of compounds better suited to cell implantation.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Embryonic Stem Cells/drug effects , Apoptosis/drug effects , Calcineurin/metabolism , Cells, Cultured , Cyclosporine/administration & dosage , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Humans , Immunosuppressive Agents/administration & dosage , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Oligopeptides/metabolism , Sirolimus/administration & dosage , Stem Cell Transplantation , Tacrolimus/administration & dosage
2.
J Cardiovasc Transl Res ; 5(5): 593-604, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22896035

ABSTRACT

Human pluripotent stem cell-derived cardiomyocytes (hPSC-CM) are being investigated as a new source of cardiac cells for drug safety assessment. We developed a novel scalable high content microscopy-based method for the detection of cell death in hPSC-CM that can serve for future predictive in vitro cardio-toxicological screens. Using rat neonatal ventricular cardiomyocytes (RVNC) or hPSC-CM, assays for nuclear remodelling, mitochondrial status, apoptosis and necrosis were designed using a combination of fluorescent dyes and antibodies on an automated microscopy platform. This allowed the observation of a chelerythrine-induced concentration-dependent apoptosis to necrosis switch and time-dependent progression of early apoptotic cells towards a necrotic-like phenotype. Susceptibility of hPSC-CM to chelerythrine-stimulated apoptosis varied with time after differentiation, but at most time points, hPSC-CM were more resistant than RVNC. This simple and scalable humanized high-content assay generates accurate cardiotoxicity profiles that can serve as a base for further assessment of cardioprotective strategies and drug safety.


Subject(s)
Apoptosis/drug effects , Benzophenanthridines/toxicity , Cell Tracking/methods , High-Throughput Screening Assays/methods , Induced Pluripotent Stem Cells/drug effects , Microscopy, Fluorescence , Myocytes, Cardiac/drug effects , Animals , Animals, Newborn , Automation, Laboratory , Caspases, Effector/metabolism , Cell Line , Cell Membrane Permeability/drug effects , Cell Nucleus Shape/drug effects , Dose-Response Relationship, Drug , Enzyme Activation , Humans , Image Processing, Computer-Assisted , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Heart/drug effects , Mitochondria, Heart/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Necrosis , Phenotype , Rats , Time Factors
3.
Nanotoxicology ; 5(3): 372-80, 2011 Sep.
Article in English | MEDLINE | ID: mdl-20858044

ABSTRACT

Because of the increased use of titanium dioxide (TiO2) nanoparticles (NPs) in tissue engineering (TE), and in new constructs for cardiac TE, their effect was studied on three relevant cell types: Adult rat ventricular cardiomyocytes, human embryonic stem cell-derived cardiomyocytes (hESC-CM) and fibroblasts. For adult rat myocytes, 10 µg/mL TiO2 NPs showed no significant effect on myocyte survival over 24 h or acute myocyte contractility. Increasing the concentration to 100 µg/mL was seen to reduce contraction amplitude (p < 0.05). For hESC-CM, 10 µg/mL TiO2 reduced the beating rate significantly by 24 h. No arrhythmias or cessation of beating were observed in either cell type. Culturing fibroblasts in 5-150 µg/mL TiO2 significantly reduced cell proliferation at day 4 and increased cell death. We conclude that there may be modest but potentially adverse effects of TiO2 NPs if used in fast degrading polymers for myocardial tissue engineering (MTE) applications.


Subject(s)
Metal Nanoparticles/toxicity , Myocardium/cytology , Myocytes, Cardiac/drug effects , Tissue Engineering/methods , Titanium/toxicity , Animals , Cell Line , Cell Proliferation , Cells, Cultured , Electrophysiological Phenomena , L-Lactate Dehydrogenase/metabolism , Metal Nanoparticles/chemistry , Myocytes, Cardiac/physiology , Particle Size , Rats , Rats, Sprague-Dawley , Titanium/chemistry
4.
J Mol Cell Cardiol ; 50(2): 367-76, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21047517

ABSTRACT

Human embryonic stem cell-derived cardiomyocytes (hESC-CM) are being developed for tissue repair and as a model system for cardiac physiology and pathophysiology. However, the signaling requirements of their growth have not yet been fully characterized. We showed that hESC-CM retain their capacity for increase in size in long-term culture. Exposing hESC-CM to hypertrophic stimuli such as equiaxial cyclic stretch, angiotensin II, and phenylephrine (PE) increased cell size and volume, percentage of hESC-CM with organized sarcomeres, levels of ANF, and cytoskeletal assembly. PE effects on cell size were separable from those on cell cycle. Changes in cell size by PE were completely inhibited by p38-MAPK, calcineurin/FKBP, and mTOR blockers. p38-MAPK and calcineurin were also implicated in basal cell growth. Inhibitors of ERK, JNK, and CaMK II partially reduced PE effects; PKG or GSK3ß inhibitors had no effect. The role of p38-MAPK was confirmed by an additional pharmacological inhibitor and adenoviral infection of hESC-CM with a dominant-inhibitory form of p38-MAPK. Infection of hESC-CM with constitutively active upstream MAP2K3b resulted in an increased cell size, sarcomere and cytoskeletal assembly, elongation of the cells, and induction of ANF mRNA levels. siRNA knockdown of p38-MAPK inhibited PE-induced effects on cell size. These results reveal an important role for active protein kinase signaling in hESC-CM growth and hypertrophy, with potential implications for hESC-CM as a novel in vitro test system. This article is part of a special issue entitled, "Cardiovascular Stem Cells Revisited".


Subject(s)
Cardiomegaly/metabolism , Cardiomegaly/pathology , Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Angiotensin II/pharmacology , Cardiomegaly/chemically induced , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Size/drug effects , Cells, Cultured , Gene Expression Regulation/drug effects , Humans , MAP Kinase Kinase 3/metabolism , Myocytes, Cardiac/drug effects , Phenylephrine/pharmacology , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Vasoconstrictor Agents/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
5.
Tissue Eng Part A ; 16(11): 3395-402, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20528670

ABSTRACT

Grafting of elastomeric biomaterial scaffolds may offer a radical strategy for the prevention of heart failure after myocardial infarction by increasing efficacy of stem cell delivery as well as acting as mechanical restraint devices to constrain scar expansion. Biomaterials can be partially optimized in vitro, but their in vivo performance is most critical and should ideally be monitored serially and noninvasively. We used magnetic resonance imaging (MRI) to assess three scaffold materials with a range of structural moduli equal to or greater than myocardial tissue: poly(glycerol sebacate) (PGS), poly(ethyleneterephathalate)/dimer fatty acid (PED), and TiO(2)-reinforced PED (PED-TiO(2)). Patches, 1 cm in diameter, were grafted onto the hearts of infarcted rats, with biomaterial-free infarcted rat hearts used as controls. MRI was able to determine scaffold size and location on the heart and identified unexpectedly rapid in vivo degradation of the PGS compared with previous in vitro testing. PED patches did not withstand in vivo attachment, but the more rigid PED-TiO(2) material was detrimental to heart function, increasing chamber and scar sizes and reducing ejection fractions compared with controls. In contrast, the mechanically compatible PGS scaffold successfully reduced hypertrophy, giving it potential for limiting excessive postinfarct remodeling. PGS was unable to support systolic function, but it would be suitable for strategies to deliver cardiac stem/progenitor cells, to limit remodeling during the period of functional cellular integration, and to degrade after cell assimilation by the heart. This work has also shown for the first time the value of using MRI as a noninvasive tool for evaluating and optimizing therapeutic biomaterials in vivo.


Subject(s)
Biocompatible Materials/pharmacology , Elastomers/pharmacology , Magnetic Resonance Imaging , Myocardial Infarction/diagnosis , Myocardial Infarction/physiopathology , Tissue Scaffolds/chemistry , Ventricular Remodeling/drug effects , Animals , Disease Models, Animal , Myocardium/pathology , Rats , Tissue Engineering
6.
PLoS One ; 5(5): e10501, 2010 May 05.
Article in English | MEDLINE | ID: mdl-20463927

ABSTRACT

Treatment of human disease with human embryonic stem cell (hESC)-derived cells is now close to reality, but little is known of their responses to physiological and pathological insult. The ability of cells to respond via activation of Toll like receptors (TLR) is critical in innate immune sensing in most tissues, but also extends to more general danger sensing, e.g. of oxidative stress, in cardiomyocytes. We used biomarker release and gene-array analysis to compare responses in hESC before and after differentiation, and to those in primary human endothelial cells. The presence of cardiomyocytes and endothelial cells was confirmed in differentiated cultures by immunostaining, FACS-sorting and, for cardiomyocytes, beating activity. Undifferentiated hESC did not respond with CXCL8 release to Gram positive or Gram negative bacteria, or a range of PAMPs (pathogen associated molecular patterns) for TLRs 1-9 (apart from flagellin, an activator of TLR5). Surprisingly, lack of TLR-dependent responses was maintained over 4 months of differentiation of hESC, in cultures which included cardiomyocytes and endothelial cells. In contrast, primary cultures of human aortic endothelial cells (HAEC) demonstrated responses to a broad range of PAMPs. Expression of downstream TLR signalling pathways was demonstrated in hESC, and IL-1beta, TNFalpha and INFgamma, which bypass the TLRs, stimulated CXCL8 release. NFkappaB pathway expression was also present in hESC and NFkappaB was able to translocate to the nucleus. Low expression levels of TLRs were detected in hESC, especially TLRs 1 and 4, explaining the lack of response of hESC to the main TLR signals. TLR5 levels were similar between differentiated hESC and HAEC, and siRNA knockdown of TLR5 abolished the response to flagellin. These findings have potential implications for survival and function of grafted hESC-derived cells.


Subject(s)
Embryonic Stem Cells/immunology , Endothelial Cells/immunology , Immunity, Innate/immunology , Adult , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Shape/drug effects , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Escherichia coli/drug effects , Escherichia coli/immunology , Flagellin/immunology , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Humans , Immunity, Innate/drug effects , Immunity, Innate/genetics , Interleukin-8/metabolism , Lipopolysaccharides/pharmacology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Phenotype , Receptors, Pattern Recognition/metabolism , Signal Transduction/drug effects , Staphylococcus aureus/drug effects , Staphylococcus aureus/immunology , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
7.
Biomaterials ; 31(14): 3885-93, 2010 May.
Article in English | MEDLINE | ID: mdl-20153041

ABSTRACT

We hypothesize that a combinatorial approach of ventricle constraint and stem cell therapy would offer a greater benefit for the treatment of heart failure than either strategy alone. A heart patch would serve two therapeutic purposes: biomechanical support and cell delivery. In this study, we describe a hybrid heart patch engineered from a synthetic elastomer, poly(glycerol sebacate) (PGS), supplemented with cardiomyocytes differentiated from human embryonic stem cells (hESCs). In line with two therapeutically relevant considerations, i.e. biocompatibility and cell delivery efficiency, the PGS was (a) pre-conditioned in culture medium for 6 days, and (b) prepared without gelatin coatings to facilitate detachment and delivery of cardiomyocytes following patch implantation. Following pre-conditioning under physiological conditions, the PGS patch material without gelatin coating was found to satisfactorily support cardiomyocyte viability and attachment, with active cell beating for periods of longer than 3 months until interrupted. Dynamic culture studies revealed that cells detached more efficiently from the uncoated surface of PGS than from gelatin-coated PGS. No significant differences were detected between the beating rates of human embryonic stem cell-derived cardiomyocytes on tissue culture plate and the pre-conditioned and gelatin-uncoated PGS. PGS patches sutured over the left ventricle of rats in vivo remained intact over a 2 week period without any deleterious effects on ventricular function. We conclude that PGS is a suitable biomaterial for stem cell-based regeneration strategies to restore cardiomyocyte function, and the hybrid heart patch engineered under optimal conditions would be a promising support device for the cardiac repair.


Subject(s)
Decanoates/pharmacology , Elastomers/pharmacology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Glycerol/analogs & derivatives , Myocardium/cytology , Polymers/pharmacology , Tissue Engineering/methods , Acids , Aging , Animals , Cell Adhesion/drug effects , Cell Death/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Decanoates/toxicity , Embryonic Stem Cells/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Glycerol/pharmacology , Glycerol/toxicity , Humans , Kinetics , Materials Testing , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Polymers/toxicity , Rats
8.
J Cell Mol Med ; 13(9B): 3730-41, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19438812

ABSTRACT

Embryonic stem cell-derived cardiomyocytes (ESC-CM) have many of the phenotypic properties of authentic cardiomyocytes, and great interest has been shown in their possibilities for modelling human disease. Obstetric cholestasis affects 1 in 200 pregnant women in the United Kingdom. It is characterized by raised serum bile acids and complicated by premature delivery and unexplained fetal death at late gestation. It has been suggested that the fetal death is caused by the enhanced arrhythmogenic effect of bile acids in the fetal heart, and shown that neonatal susceptibility to bile acid-induced arrhythmia is lost in the adult rat cardiomyocyte. However, the mechanisms of the observed bile acid effects are not fully understood and their in vivo study in human beings is difficult. Here we use ESC-CM from both human and mouse ESCs to test our proposal that immature cardiomyocytes are more susceptible to the effect of raised bile acids than mature ones. We show that early ESC-CM exhibit bile acid-induced disruption of rhythm, depression of contraction and desynchronization of cell coupling. In both species the ESC-CM become resistant to these arrhythmias as the cells mature, and this develops in line with the respective gestational periods of mouse and human. This represents the first demonstration of the use of ESC-CM as a model system for human cardiac pathology, and opens the way for both investigation of mechanisms and a high throughput screen for drug discovery.


Subject(s)
Arrhythmias, Cardiac/pathology , Bile Acids and Salts/metabolism , Fetal Diseases/pathology , Myocytes, Cardiac/cytology , Animals , Calcium/metabolism , Cell Differentiation , Cholestasis/metabolism , Disease Models, Animal , Female , Humans , Mice , Myocytes, Cardiac/metabolism , Pregnancy , Pregnancy Complications, Cardiovascular , Time Factors
9.
Tissue Eng Part C Methods ; 14(4): 311-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19055357

ABSTRACT

The most valuable property of stem cells (SCs) is their potential to differentiate into many or all cell types of the body. So far, monitoring SC differentiation has only been possible after cells were fixed or destroyed during sample preparation. It is, however, important to develop nondestructive methods of monitoring SCs. Scanning ion conductance microscopy (SICM) is a unique imaging technique that uses similar principles to the atomic force microscope, but with a pipette for the probe. This allows scanning of the surface of living cells noninvasively and enables measurement of cellular activities under more physiological conditions than is possible with other high-resolution microscopy techniques. We report here the novel use of the SICM for studying SCs to assess and monitor the status of SCs and various cell types differentiated from SCs.


Subject(s)
Microscopy, Electron, Scanning/methods , Stem Cells/cytology , Animals , Cell Differentiation , Cell Line , Cell Proliferation , Equipment Design , Humans , Mice , Microscopy, Confocal/methods , Neural Crest/pathology , Neurons/cytology , Patch-Clamp Techniques , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/ultrastructure , Surface Properties
10.
Br Med Bull ; 87: 31-47, 2008.
Article in English | MEDLINE | ID: mdl-18790825

ABSTRACT

INTRODUCTION: Regeneration of the infarcted myocardium after a heart attack is one of the most challenging aspects in tissue engineering. Suitable cell sources and optimized biocompatible materials must be identified. SOURCES OF DATA: In this review, we briefly discuss the current therapeutic options available to patients with heart failure post-myocardial infarction. We describe the various strategies currently proposed to encourage myocardial regeneration, with focus on the achievements in myocardial tissue engineering (MTE). We report on the current cell types, materials and methods being investigated for developing a tissue-engineered myocardial construct. AREAS OF AGREEMENT: Generally, there is agreement that a 'vehicle' is required to transport cells to the infarcted heart to help myocardial repair and regeneration. AREAS OF CONTROVERSY: Suitable cell source, biomaterials, cell environment and implantation time post-infarction remain obstacles in the field of MTE. GROWING POINTS: Research is being focused on optimizing natural and synthetic biomaterials for tissue engineering. The type of cell and its origin (autologous or derived from embryonic stem cells), cell density and method of cell delivery are also being explored. AREAS TIMELY FOR DEVELOPING RESEARCH: The possibility is being explored that materials may not only act as a support for the delivered cell implants, but may also add value by changing cell survival, maturation or integration, or by prevention of mechanical and electrical remodelling of the failing heart.


Subject(s)
Biocompatible Materials/therapeutic use , Cell Transplantation/methods , Myocardial Infarction/therapy , Myocardium , Myocytes, Cardiac/transplantation , Tissue Engineering/methods , Humans , Myocardial Infarction/physiopathology , Regeneration
11.
Expert Opin Biol Ther ; 8(10): 1473-83, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18774916

ABSTRACT

BACKGROUND: Despite improvements in survival with current therapies, it is now clear that major cardiovascular event rates in patients remain high. Cardiac cell replacement therapy by using human embryonic stem cell-derived cardiomyocytes has emerged as a promising future approach to regenerate functional myocardium. However, there are still many hurdles to be overcome for the clinical application of these cells. OBJECTIVE: This review describes the embryonic stem cell system, their differentiation into cardiomyocytes, and functional characterization of the cardiac lineage derivatives in culture and upon in vivo engraftment in animal models. RESULTS/CONCLUSION: A better understanding of the characteristics of the cardiomyocytes from human embryonic stem cells not only predicts their behaviour after implantation but will also help in the design of future strategies for cardiac regeneration in vivo.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Myocardium/chemistry , Cell Transplantation , Cloning, Organism , Heart/physiology , Humans , Myocardial Infarction/therapy , Regeneration
12.
Biomaterials ; 29(1): 47-57, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17915309

ABSTRACT

The myocardial tissue lacks significant intrinsic regenerative capability to replace the lost cells. Therefore, the heart is a major target of research within the field of tissue engineering, which aims to replace infarcted myocardium and enhance cardiac function. The primary objective of this work was to develop a biocompatible, degradable and superelastic heart patch from poly(glycerol sebacate) (PGS). PGS was synthesised at 110, 120 and 130 degrees C by polycondensation of glycerol and sebacic acid with a mole ratio of 1:1. The investigation was focused on the mechanical and biodegrading behaviours of the developed PGS. PGS materials synthesised at 110, 120 and 130 degrees C have Young's moduli of 0.056, 0.22 and 1.2 MPa, respectively, which satisfy the mechanical requirements on the materials applied for the heart patch and 3D myocardial tissue engineering construction. Degradation assessment in phosphate buffered saline and Knockout DMEM culture medium has demonstrated that the PGS has a wide range of degradability, from being degradable in a couple of weeks to being nearly inert. The matching of physical characteristics to those of the heart, the ability to fine tune degradation rates in biologically relevant media and initial data showing biocompatibility indicate that this material has promise for cardiac tissue engineering applications.


Subject(s)
Decanoates/chemistry , Elastomers/chemistry , Glycerol/analogs & derivatives , Myocardium , Polymers/chemistry , Cross-Linking Reagents/chemistry , Decanoates/chemical synthesis , Furans/chemistry , Glycerol/chemical synthesis , Glycerol/chemistry , Microscopy, Electron, Scanning , Polymers/chemical synthesis , Stress, Mechanical , Tensile Strength , Tissue Engineering , X-Ray Diffraction
13.
Pharmacol Ther ; 113(2): 341-53, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17081613

ABSTRACT

Embryonic stem (ES) cells are specialised cells derived from the early embryo, which are capable of both sustained propagation in the undifferentiated state as well as subsequent differentiation into the majority of cell lineages. Human ES cells are being developed for clinical tissue repair, but a number of problems must be addressed before this becomes a reality. However, they also have potential for translational benefit through its use as a test system for screening pharmaceutical compounds. In the cardiac field, present model systems are not ideal for either screening or basic pharmacological/physiological studies. Cardiomyocytes produced from human ES differentiation have advantages for these purposes over the primary isolated cells or the small number of cell lines available. This review describes the methodology for obtaining cardiomyocytes from human embryonic stem cell-derived cardiomyocyte (hESCM), for increasing the proportion of cardiomyocytes in the preparation and for isolating single embryonic stem cell-derived cardiomyocyte (ESCM) from clusters. Their morphological, contractile and electrophysiological characteristics are compared to mature and immature primary cardiomyocytes. The advantages and disadvantages of the hESCM preparation for long term culture and genetic manipulation are described. Basic pharmacological studies on adrenoceptors and muscarinic receptors in hESCM have been performed, and have given stable and reproducible responses. Prolongation of repolarisation can be detected using hESCM cultured on multielectrode arrays (MEA). Human ESCM have a clear potential to improve model systems available for both basic scientific studies and pharmaceutical screening of cardiac target compounds.


Subject(s)
Embryonic Stem Cells/cytology , Myocytes, Cardiac , Adrenergic Agonists/pharmacology , Animals , Drug Design , Humans , Muscarinic Agonists/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology
14.
Tissue Eng ; 12(4): 657-64, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16674281

ABSTRACT

We report here the novel use of scanning ion conductance microscopy (SICM) to produce surface images of embryonic stem cell-derived cardiomyocytes (ESCM) to identify individual contracting cardiomyocytes among different cell types. By measuring amplitude and rhythm we can quantitate contraction of ESCM. This method gives, within the same experiment, an assessment of the number and position of ESCM within the layer of mixed cell types, as well as an accurate measure of the response of individual ESCM. Using different modulators of contraction as examples we showed how SICM could be used for recording their responses. We subsequently demonstrated that this model can be used to investigate the protective effect of antiarrhythmogenic drugs.


Subject(s)
Embryo, Mammalian/cytology , Mesenchymal Stem Cells/cytology , Microscopy, Scanning Tunneling , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Animals , Anti-Arrhythmia Agents/pharmacology , Cell Culture Techniques , Cell Differentiation , Cell Line , Cell Lineage , Mice , Microscopy, Scanning Tunneling/instrumentation , Microscopy, Scanning Tunneling/methods , Myocardial Contraction/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects
15.
Tissue Eng ; 8(4): 541-50, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12201994

ABSTRACT

Embryonic stem (ES) cell pluripotency is being investigated increasingly to obtain specific cell lineages for tissue engineering. However, the possibility that ES cells can give rise to lung tissue has not been tested. We hypothesized that lung epithelial cells (type II pneumocytes) can be derived in vitro from murine ES cells. After withdrawal of leukemia inhibitory factor (LIF) and formation of embryoid bodies in maintenance medium for 10, 20, and 30 days, differentiating ES cells were kept in the same medium or transferred to serum-free small airway growth medium (SAGM) for a further 3 or 14 days of culture. The presence of type II pneumocytes in the resulting mixed cultures was demonstrated by reverse transcriptase-polymerase chain reaction (RT-PCR) of surfactant protein C (SPC) mRNA, immunostaining of SPC, and electron microscopy of osmiophilic lamellar bodies only at 30 days sampling time. SAGM appeared to be more favorable for type II cell formation than ES medium. No SPC transcripts were found in differentiating cells grown under the same conditions without formation of embryoid bodies. These findings could form the basis for the enrichment of ES cell-derived cultures with type II pneumocytes, and provide an in vitro system for investigating mechanisms of lung repair and regeneration.


Subject(s)
Epithelial Cells/cytology , Pulmonary Alveoli/cytology , Stem Cells/cytology , Animals , Biomarkers , Cell Differentiation , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Immunohistochemistry , Mice , Microscopy, Phase-Contrast , Pluripotent Stem Cells/cytology , Pulmonary Surfactants/metabolism , RNA, Messenger , Stem Cells/metabolism , Stem Cells/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...