Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
EMBO J ; 32(24): 3161-75, 2013 Dec 11.
Article in English | MEDLINE | ID: mdl-24213244

ABSTRACT

Patient-specific induced pluripotent stem cells (iPSCs) will assist research on genetic cardiac maladies if the disease phenotype is recapitulated in vitro. However, genetic background variations may confound disease traits, especially for disorders with incomplete penetrance, such as long-QT syndromes (LQTS). To study the LQT2-associated c.A2987T (N996I) KCNH2 mutation under genetically defined conditions, we derived iPSCs from a patient carrying this mutation and corrected it. Furthermore, we introduced the same point mutation in human embryonic stem cells (hESCs), generating two genetically distinct isogenic pairs of LQTS and control lines. Correction of the mutation normalized the current (IKr) conducted by the HERG channel and the action potential (AP) duration in iPSC-derived cardiomyocytes (CMs). Introduction of the same mutation reduced IKr and prolonged the AP duration in hESC-derived CMs. Further characterization of N996I-HERG pathogenesis revealed a trafficking defect. Our results demonstrated that the c.A2987T KCNH2 mutation is the primary cause of the LQTS phenotype. Precise genetic modification of pluripotent stem cells provided a physiologically and functionally relevant human cellular context to reveal the pathogenic mechanism underlying this specific disease phenotype.


Subject(s)
Ether-A-Go-Go Potassium Channels/genetics , Long QT Syndrome/genetics , Mutation , Pluripotent Stem Cells , Action Potentials/genetics , Adult , Cells, Cultured , ERG1 Potassium Channel , Embryonic Stem Cells/physiology , Ether-A-Go-Go Potassium Channels/metabolism , Female , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Humans , Induced Pluripotent Stem Cells , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Patch-Clamp Techniques , Phenotype , Pluripotent Stem Cells/physiology , Protein Transport/genetics , Transcription Factors/genetics
2.
Proc Natl Acad Sci U S A ; 111(50): E5383-92, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25453094

ABSTRACT

Jervell and Lange-Nielsen syndrome (JLNS) is one of the most severe life-threatening cardiac arrhythmias. Patients display delayed cardiac repolarization, associated high risk of sudden death due to ventricular tachycardia, and congenital bilateral deafness. In contrast to the autosomal dominant forms of long QT syndrome, JLNS is a recessive trait, resulting from homozygous (or compound heterozygous) mutations in KCNQ1 or KCNE1. These genes encode the α and ß subunits, respectively, of the ion channel conducting the slow component of the delayed rectifier K(+) current, IKs. We used complementary approaches, reprogramming patient cells and genetic engineering, to generate human induced pluripotent stem cell (hiPSC) models of JLNS, covering splice site (c.478-2A>T) and missense (c.1781G>A) mutations, the two major classes of JLNS-causing defects in KCNQ1. Electrophysiological comparison of hiPSC-derived cardiomyocytes (CMs) from homozygous JLNS, heterozygous, and wild-type lines recapitulated the typical and severe features of JLNS, including pronounced action and field potential prolongation and severe reduction or absence of IKs. We show that this phenotype had distinct underlying molecular mechanisms in the two sets of cell lines: the previously unidentified c.478-2A>T mutation was amorphic and gave rise to a strictly recessive phenotype in JLNS-CMs, whereas the missense c.1781G>A lesion caused a gene dosage-dependent channel reduction at the cell membrane. Moreover, adrenergic stimulation caused action potential prolongation specifically in JLNS-CMs. Furthermore, sensitivity to proarrhythmic drugs was strongly enhanced in JLNS-CMs but could be pharmacologically corrected. Our data provide mechanistic insight into distinct classes of JLNS-causing mutations and demonstrate the potential of hiPSC-CMs in drug evaluation.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Jervell-Lange Nielsen Syndrome/drug therapy , Jervell-Lange Nielsen Syndrome/genetics , Jervell-Lange Nielsen Syndrome/physiopathology , KCNQ1 Potassium Channel/genetics , Models, Biological , Phenotype , Action Potentials/physiology , Analysis of Variance , Base Sequence , Cell Line , Genes, Recessive/genetics , Genetic Engineering , Humans , In Vitro Techniques , KCNQ1 Potassium Channel/chemistry , Models, Molecular , Molecular Sequence Data , Mutation, Missense/genetics , Myocytes, Cardiac/physiology , Sequence Analysis, DNA
3.
Biochem Biophys Res Commun ; 467(4): 998-1005, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26456652

ABSTRACT

One limitation in using human pluripotent stem cell derived cardiomyocytes (hPSC-CMs) for disease modeling and cardiac safety pharmacology is their immature functional phenotype compared with adult cardiomyocytes. Here, we report that treatment of human embryonic stem cell derived cardiomyocytes (hESC-CMs) with dexamethasone, a synthetic glucocorticoid, activated glucocorticoid signaling which in turn improved their calcium handling properties and contractility. L-type calcium current and action potential properties were not affected by dexamethasone but significantly faster calcium decay, increased forces of contraction and sarcomeric lengths, were observed in hESC-CMs after dexamethasone exposure. Activating the glucocorticoid pathway can thus contribute to mediating hPSC-CMs maturation.


Subject(s)
Calcium/metabolism , Dexamethasone/pharmacology , Embryonic Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Cell Line , Dexamethasone/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Humans , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Signal Transduction
4.
Stem Cells ; 32(3): 636-48, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24549638

ABSTRACT

Bone morphogenetic proteins (BMPs) initiate differentiation in human embryonic stem cells (hESCs) but the exact mechanisms have not been fully elucidated. We demonstrate here that SLUG and MSX2, transcription factors involved in epithelial-mesenchymal transitions, essential features of gastrulation in development and tumor progression, are important mediators of BMP4-induced differentiation in hESCs. Phosphorylated Smad1/5/8 colocalized with the SLUG protein at the edges of hESC colonies where differentiation takes place. The upregulation of the BMP target SLUG was direct as shown by the binding of phosphorylated Smad1/5/8 to its promoter, which interrupted the formation of adhesion proteins, resulting in migration. Knockdown of SLUG by short hairpin RNA blocked these changes, confirming an important role for SLUG in BMP-mediated mesodermal differentiation. Furthermore, BMP4-induced MSX2 expression leads to mesoderm formation and then preferential differentiation toward the cardiovascular lineage.


Subject(s)
Bone Morphogenetic Protein 4/pharmacology , Cell Lineage/drug effects , Embryonic Stem Cells/cytology , Epithelial-Mesenchymal Transition/drug effects , Homeodomain Proteins/metabolism , Mesoderm/cytology , Transcription Factors/metabolism , Animals , Cadherins/metabolism , Cell Differentiation/drug effects , Cell Line , Cell Movement/drug effects , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Humans , Mice , Models, Biological , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Transport/drug effects , Smad Proteins/metabolism , Snail Family Transcription Factors , Transcription Factors/genetics , Up-Regulation/drug effects
5.
Nat Methods ; 8(12): 1037-40, 2011 Oct 23.
Article in English | MEDLINE | ID: mdl-22020065

ABSTRACT

NKX2-5 is expressed in the heart throughout life. We targeted eGFP sequences to the NKX2-5 locus of human embryonic stem cells (hESCs); NKX2-5(eGFP/w) hESCs facilitate quantification of cardiac differentiation, purification of hESC-derived committed cardiac progenitor cells (hESC-CPCs) and cardiomyocytes (hESC-CMs) and the standardization of differentiation protocols. We used NKX2-5 eGFP(+) cells to identify VCAM1 and SIRPA as cell-surface markers expressed in cardiac lineages.


Subject(s)
Cell Separation/methods , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Myoblasts, Cardiac/cytology , Myocytes, Cardiac/cytology , Transcription Factors/metabolism , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Biomarkers/analysis , Cell Differentiation , Gene Expression Profiling , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Humans , Myoblasts, Cardiac/metabolism , Myocytes, Cardiac/metabolism , Polymerase Chain Reaction , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Transcription Factors/genetics , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
7.
Mater Today Bio ; 23: 100818, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37810749

ABSTRACT

Heart and kidney communicate with one another in an interdependent relationship and they influence each other's behavior reciprocally, as pathological changes in one organ can damage the other. Although independent human in vitro models for heart and kidney exist, they do not capture their dynamic crosstalk. We have developed a microfluidic system which can be used to study heart and kidney interaction in vitro. Cardiac microtissues (cMTs) and kidney organoids (kOs) derived from human induced pluripotent stem cells (hiPSCs) were generated and loaded into two separated communicating chambers of a perfusion chip. Static culture conditions were compared with dynamic culture under unidirectional flow. Tissue viability was maintained for minimally 72 h under both conditions, as indicated by the presence of sarcomeric structures coupled with beating activity in cMTs and the presence of nephron structures and albumin uptake in kOs. We concluded that this system enables the study of human cardiac and kidney organoid interaction in vitro while controlling parameters like fluidic flow speed and direction. Together, this "cardiorenal-unit" provides a new in vitro model to study the cardiorenal axis and it may be further developed to investigate diseases involving both two organs and their potential treatments.

8.
Cardiovasc Res ; 119(1): 167-182, 2023 03 17.
Article in English | MEDLINE | ID: mdl-35394010

ABSTRACT

AIMS: Human-induced pluripotent stem cell-cardiomyocytes (hiPSC-CMs) are widely used to study arrhythmia-associated mutations in ion channels. Among these, the cardiac sodium channel SCN5A undergoes foetal-to-adult isoform switching around birth. Conventional hiPSC-CM cultures, which are phenotypically foetal, have thus far been unable to capture mutations in adult gene isoforms. Here, we investigated whether tri-cellular cross-talk in a three-dimensional (3D) cardiac microtissue (MT) promoted post-natal SCN5A maturation in hiPSC-CMs. METHODS AND RESULTS: We derived patient hiPSC-CMs carrying compound mutations in the adult SCN5A exon 6B and exon 4. Electrophysiological properties of patient hiPSC-CMs in monolayer were not altered by the exon 6B mutation compared with isogenic controls since it is not expressed; further, CRISPR/Cas9-mediated excision of the foetal exon 6A did not promote adult SCN5A expression. However, when hiPSC-CMs were matured in 3D cardiac MTs, SCN5A underwent isoform switch and the functional consequences of the mutation located in exon 6B were revealed. Up-regulation of the splicing factor muscleblind-like protein 1 (MBNL1) drove SCN5A post-natal maturation in microtissues since its overexpression in hiPSC-CMs was sufficient to promote exon 6B inclusion, whilst knocking-out MBNL1 failed to foster isoform switch. CONCLUSIONS: Our study shows that (i) the tri-cellular cardiac microtissues promote post-natal SCN5A isoform switch in hiPSC-CMs, (ii) adult splicing of SCN5A is driven by MBNL1 in these tissues, and (iii) this model can be used for examining post-natal cardiac arrhythmias due to mutations in the exon 6B. TRANSLATIONAL PERSPECTIVE: The cardiac sodium channel is essential for conducting the electrical impulse in the heart. Postnatal alternative splicing regulation causes mutual exclusive inclusion of fetal or adult exons of the corresponding gene, SCN5A. Typically, immature hiPSCCMs fall short in studying the effect of mutations located in the adult exon. We describe here that an innovative tri-cellular three-dimensional cardiac microtissue culture promotes hiPSC-CMs maturation through upregulation of MBNL1, thus revealing the effect of a pathogenic genetic variant located in the SCN5A adult exon. These results help advancing the use of hiPSC-CMs in studying adult heart disease and for developing personalized medicine applications.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Adult , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Alternative Splicing , Sodium/metabolism , Arrhythmias, Cardiac/metabolism , Cardiac Conduction System Disease/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/pharmacology , Action Potentials
9.
Eur J Med Chem ; 212: 113033, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33261899

ABSTRACT

We synthesized and evaluated three novel series of substituted benzophenones for their allosteric modulation of the human Kv11.1 (hERG) channel. We compared their effects with reference compound LUF7346 previously shown to shorten the action potential of cardiomyocytes derived from human stem cells. Most compounds behaved as negative allosteric modulators (NAMs) of [3H]dofetilide binding to the channel. Compound 9i was the most potent amongst all ligands, remarkably reducing the affinity of dofetilide in competitive displacement assays. One of the other derivatives (6k) tested in a second radioligand binding set-up, displayed unusual displacement characteristics with a pseudo-Hill coefficient significantly distinct from unity, further indicative of its allosteric effects on the channel. Some compounds were evaluated in a more physiologically relevant context in beating cardiomyocytes derived from human induced pluripotent stem cells. Surprisingly, the compounds tested showed effects quite different from the reference NAM LUF7346. For instance, compound 5e prolonged, rather than shortened, the field potential duration, while it did not influence this parameter when the field potential was already prolonged by dofetilide. In subsequent patch clamp studies on HEK293 cells expressing the hERG channel the compounds behaved as channel blockers. In conclusion, we successfully synthesized and identified new allosteric modulators of the hERG channel. Unexpectedly, their effects differed from the reference compound in functional assays on hERG-HEK293 cells and human cardiomyocytes, to the extent that the compounds behaved as stand-alone channel blockers.


Subject(s)
ERG1 Potassium Channel/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Allosteric Regulation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , ERG1 Potassium Channel/metabolism , HEK293 Cells , Humans , Molecular Structure , Potassium Channel Blockers/chemical synthesis , Potassium Channel Blockers/chemistry , Structure-Activity Relationship
10.
J Proteome Res ; 9(3): 1610-8, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20088484

ABSTRACT

The absence of identified cell surface proteins and corresponding antibodies to most differentiated derivatives of human embryonic stem cells (hESCs) has largely limited selection of specific cell types from mixed cell populations to genetic approaches. Here, we describe the use of mass spectrometry (MS)-based proteomics on cell membrane proteins isolated from hESCs that were differentiated into cardiomyocytes to identify candidate proteins for this particular lineage. Quantitative MS distinguished cardiomyocyte-specific plasma membrane proteins that were highly enriched or detected only in cardiomyocytes derived from hESCs and human fetal hearts compared with a heterogeneous pool of hESC-derived differentiated cells. For several candidates, cardiomyocyte-specific expression and cell surface localization were verified by conventional antibody-based methodologies. Using an antibody against elastin microfibril interfacer 2 (EMILIN2), we demonstrate that cardiomyocytes can be sorted from live cell populations. Besides showing that MS-based membrane proteomics is a powerful tool to identify candidate proteins that allow purification of specific cell lineages from heterogeneous populations, this approach generated a plasma membrane proteome profile suggesting signaling pathways that control cell behavior.


Subject(s)
Biomarkers/analysis , Embryonic Stem Cells/metabolism , Membrane Proteins/analysis , Myocytes, Cardiac/metabolism , Proteomics/methods , Antibodies/metabolism , Biomarkers/metabolism , Blotting, Western , Cell Differentiation , Cell Separation , Embryonic Stem Cells/cytology , Fibrillins , Glycoproteins/metabolism , Humans , Isotope Labeling , Mass Spectrometry , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Microscopy, Fluorescence , Myocytes, Cardiac/cytology
11.
Stem Cells ; 26(3): 724-33, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18096723

ABSTRACT

Human embryonic stem cells (hESC) can proliferate indefinitely while retaining the capacity to form derivatives of all three germ layers. We have reported previously that hESC differentiate into cardiomyocytes when cocultured with a visceral endoderm-like cell line (END-2). Insulin/insulin-like growth factors and their intracellular downstream target protein kinase Akt are known to protect many cell types from apoptosis and to promote proliferation, including hESC-derived cardiomyocytes. Here, we show that in the absence of insulin, a threefold increase in the number of beating areas was observed in hESC/END-2 coculture. In agreement, the addition of insulin strongly inhibited cardiac differentiation, as evidenced by a significant reduction in beating areas, as well as in alpha-actinin and beta-myosin heavy chain (beta-MHC)-expressing cells. Real-time reverse transcription-polymerase chain reaction and Western blot analysis showed that insulin inhibited cardiomyogenesis in the early phase of coculture by suppressing the expression of endoderm (Foxa2, GATA-6), mesoderm (brachyury T), and cardiac mesoderm (Nkx2.5, GATA-4). In contrast to previous reports, insulin was not sufficient to maintain hESC in an undifferentiated state, since expression of the pluripotency markers Oct3/4 and nanog declined independently of the presence of insulin during coculture. Instead, insulin promoted the expression of neuroectodermal markers. Since insulin triggered sustained phosphorylation of Akt in hESC, we analyzed the effect of an Akt inhibitor during coculture. Indeed, the inhibition of Akt or insulin-like growth factor-1 receptor reversed the insulin-dependent effects. We conclude that in hESC/END-2 cocultures, insulin does not prevent differentiation but favors the neuroectodermal lineage at the expense of mesendodermal lineages.


Subject(s)
Cell Differentiation/drug effects , Embryonic Stem Cells/cytology , Endoderm/cytology , Insulin/pharmacology , Mesoderm/cytology , Myocytes, Cardiac/cytology , Neural Plate/cytology , Animals , Biomarkers/metabolism , Cell Lineage/drug effects , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/enzymology , Endoderm/drug effects , Humans , Mesoderm/drug effects , Mice , Models, Biological , Myocytes, Cardiac/drug effects , Neural Plate/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Receptor, IGF Type 1/metabolism
12.
Stem Cells ; 26(11): 2777-81, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18703662

ABSTRACT

Human embryonic stem cells (hESCs) are often cocultured on mitotically inactive fibroblast feeder cells to maintain their undifferentiated state. Under these growth conditions, hESCs form multilayered colonies of morphologically heterogeneous cells surrounded by flattened mesenchymal cells. In contrast, hESCs grown in feeder cell-conditioned medium on Matrigel instead tend to grow as monolayers with uniform morphology. Using mass spectrometry and immunofluorescence microscopy, we showed that hESCs under these conditions primarily express proteins belonging to epithelium-related cell-cell adhesion complexes, including adherens junctions, tight junctions, desmosomes, and gap junctions. This indicates that monolayers of hESCs cultured under feeder-free conditions retain a homogeneous epithelial phenotype similar to that of the upper central cell layer of colonies maintained on feeder cells. Notably, feeder-free hESCs also coexpressed vimentin, which is usually associated with mesenchyme, suggesting that these cells may have undergone epithelium-to-mesenchyme transitions, indicating differentiation. However, if grown on a "soft" substrate (Hydrogel), intracellular vimentin levels were substantially reduced. Moreover, when hESCs were transferred back to feeder cells, expression of vimentin was again absent from the epithelial cell population. These results imply that on tissue culture substrates, vimentin expression is most likely a stress-induced response, unrelated to differentiation. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Cell Membrane/metabolism , Embryonic Stem Cells/cytology , Epithelial Cells/cytology , Membrane Proteins/metabolism , Antigens, Differentiation/metabolism , Cell Adhesion , Cell Differentiation , Cells, Cultured , Coculture Techniques/methods , Collagen/metabolism , Drug Combinations , Embryonic Stem Cells/metabolism , Epithelial Cells/metabolism , Humans , Laminin/metabolism , Mass Spectrometry , Microscopy, Fluorescence , Proteoglycans/metabolism
13.
Stem Cells ; 26(9): 2257-65, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18599809

ABSTRACT

Defined growth conditions are essential for many applications of human embryonic stem cells (hESC). Most defined media are presently used in combination with Matrigel, a partially defined extracellular matrix (ECM) extract from mouse sarcoma. Here, we defined ECM requirements of hESC by analyzing integrin expression and ECM production and determined integrin function using blocking antibodies. hESC expressed all major ECM proteins and corresponding integrins. We then systematically replaced Matrigel with defined medium supplements and ECM proteins. Cells attached efficiently to natural human vitronectin, fibronectin, and Matrigel but poorly to laminin + entactin and collagen IV. Integrin-blocking antibodies demonstrated that alphaVbeta5 integrins mediated adhesion to vitronectin, alpha5beta1 mediated adhesion to fibronectin, and alpha6beta1 mediated adhesion to laminin + entactin. Fibronectin in feeder cell-conditioned medium partially supported growth on all natural matrices, but in defined, nonconditioned medium only Matrigel or (natural and recombinant) vitronectin was effective. Recombinant vitronectin was the only defined functional alternative to Matrigel, supporting sustained self-renewal and pluripotency in three independent hESC lines.


Subject(s)
Embryonic Stem Cells/cytology , Receptors, Vitronectin/physiology , Vitronectin/pharmacology , Animals , Cell Adhesion , Cell Differentiation , Cell Proliferation , Cells, Cultured , Collagen , Culture Media , Drug Combinations , Embryonic Stem Cells/metabolism , Extracellular Matrix Proteins/biosynthesis , Humans , Laminin , Mice , Proteoglycans , Recombinant Proteins/pharmacology
14.
J Vis Exp ; (123)2017 05 12.
Article in English | MEDLINE | ID: mdl-28570546

ABSTRACT

Cardiomyocytes can now be derived with high efficiency from both human embryonic and human induced-Pluripotent Stem Cells (hPSC). hPSC-derived cardiomyocytes (hPSC-CMs) are increasingly recognized as having great value for modeling cardiovascular diseases in humans, especially arrhythmia syndromes. They have also demonstrated relevance as in vitro systems for predicting drug responses, which makes them potentially useful for drug-screening and discovery, safety pharmacology and perhaps eventually for personalized medicine. This would be facilitated by deriving hPSC-CMs from patients or susceptible individuals as hiPSCs. For all applications, however, precise measurement and analysis of hPSC-CM electrical properties are essential for identifying changes due to cardiac ion channel mutations and/or drugs that target ion channels and can cause sudden cardiac death. Compared with manual patch-clamp, multi-electrode array (MEA) devices offer the advantage of allowing medium- to high-throughput recordings. This protocol describes how to dissociate 2D cell cultures of hPSC-CMs to small aggregates and single cells and plate them on MEAs to record their spontaneous electrical activity as field potential. Methods for analyzing the recorded data to extract specific parameters, such as the QT and the RR intervals, are also described here. Changes in these parameters would be expected in hPSC-CMs carrying mutations responsible for cardiac arrhythmias and following addition of specific drugs, allowing detection of those that carry a cardiotoxic risk.


Subject(s)
Electrophysiologic Techniques, Cardiac , Myocytes, Cardiac/physiology , Pluripotent Stem Cells/cytology , Cell Culture Techniques , Cells, Cultured , Electrodes , Humans
15.
EMBO Mol Med ; 8(9): 1065-81, 2016 09.
Article in English | MEDLINE | ID: mdl-27470144

ABSTRACT

Long-QT syndrome (LQTS) is an arrhythmogenic disorder characterised by prolongation of the QT interval in the electrocardiogram, which can lead to sudden cardiac death. Pharmacological treatments are far from optimal for congenital forms of LQTS, while the acquired form, often triggered by drugs that (sometimes inadvertently) target the cardiac hERG channel, is still a challenge in drug development because of cardiotoxicity. Current experimental models in vitro fall short in predicting proarrhythmic properties of new drugs in humans. Here, we leveraged a series of isogenically matched, diseased and genetically engineered, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) from patients to test a novel hERG allosteric modulator for treating congenital LQTS, drug-induced LQTS or a combination of the two. By slowing IK r deactivation and positively shifting IK r inactivation, the small molecule LUF7346 effectively rescued all of these conditions, demonstrating in a human system that allosteric modulation of hERG may be useful as an approach to treat inherited and drug-induced LQTS Furthermore, our study provides experimental support of the value of isogenic pairs of patient hiPSC-CMs as platforms for testing drug sensitivities and performing safety pharmacology.


Subject(s)
Cardiovascular Agents/pharmacology , ERG1 Potassium Channel/metabolism , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/physiology , Long QT Syndrome/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Cells, Cultured , Humans
16.
Circulation ; 107(21): 2733-40, 2003 Jun 03.
Article in English | MEDLINE | ID: mdl-12742992

ABSTRACT

BACKGROUND: Cardiomyocytes derived from human embryonic stem (hES) cells could be useful in restoring heart function after myocardial infarction or in heart failure. Here, we induced cardiomyocyte differentiation of hES cells by a novel method and compared their electrophysiological properties and coupling with those of primary human fetal cardiomyocytes. METHODS AND RESULTS: hES cells were cocultured with visceral-endoderm (VE)-like cells from the mouse. This initiated differentiation to beating muscle. Sarcomeric marker proteins, chronotropic responses, and ion channel expression and function were typical of cardiomyocytes. Electrophysiology demonstrated that most cells resembled human fetal ventricular cells. Real-time intracellular calcium measurements, Lucifer yellow injection, and connexin 43 expression demonstrated that fetal and hES-derived cardiomyocytes are coupled by gap junctions in culture. Inhibition of electrical responses by verapamil demonstrated the presence of functional alpha1c-calcium ion channels. CONCLUSIONS: This is the first demonstration of induction of cardiomyocyte differentiation in hES cells that do not undergo spontaneous cardiogenesis. It provides a model for the study of human cardiomyocytes in culture and could be a step forward in the development of cardiomyocyte transplantation therapies.


Subject(s)
Cell Differentiation/physiology , Endoderm/cytology , Myocytes, Cardiac/cytology , Stem Cells/cytology , Viscera/cytology , Action Potentials/physiology , Animals , Antigens, Differentiation/biosynthesis , Calcium Signaling/physiology , Cell Communication , Cell Line , Cell Lineage , Coculture Techniques , Fluorescent Dyes , Heart/embryology , Humans , Ion Channels/biosynthesis , Mice , Myocardium/cytology , Myocytes, Cardiac/physiology , Patch-Clamp Techniques
17.
Ann N Y Acad Sci ; 1188: 52-7, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20201886

ABSTRACT

In recent years the differentiation efficiency of human embryonic stem cells (hESCs) to cardiomyocytes has improved considerably. In general, hESC-derived cardiomyocytes are formed in aggregates, which require dissociation for follow-up experimental analyses and (clinical) applications. Here, we show that inhibition of the Rho-associated kinase (ROCK) by Y-27632 improved survival of dissociated hESC-derived differentiated cells. A maximum effect on cell survival was already observed within the first 24 hours. Hereafter, no further differences in the percentage of apoptotic and proliferating cells were observed with or without ROCK-inhibitor treatment. Improved survival was observed in both cardiomyocyte as well as non-cardiomyocyte cell populations. Viable cardiomyocytes were indicated by the appearance of beating, sarcomeric organization of cardiac-specific proteins, and fluorescence of a mitochondrion-selective dye. These results facilitate development of applications of hESC-derived cardiomyocytes in multiple research areas. Furthermore, these findings may be applied to other cell types differentiated from hESCs or other stem cells.


Subject(s)
Amides/pharmacology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , rho-Associated Kinases/antagonists & inhibitors , Cell Differentiation , Cell Line , Cell Separation , Cell Survival , Embryonic Stem Cells/enzymology , Humans , Myocytes, Cardiac/enzymology , rho-Associated Kinases/metabolism
18.
Stem Cell Res ; 3(2-3): 106-12, 2009.
Article in English | MEDLINE | ID: mdl-19560991

ABSTRACT

Transplantation of human embryonic stem cell-derived cardiomyocytes (hESC-CM) has been shown to improve the function of the rodent heart 1 month after myocardial infarction (MI). However, the mechanistic basis and optimal delivery strategies are unclear. We investigated the influence of the number of injected cells, resulting graft size, and possible paracrine mechanisms in this process. MI was induced in NOD-SCID mice (n=84) followed by injection of enriched hESC-CM at different dosages, hESC-non-CM derivatives, culture medium, or no injection. Cardiac function was monitored for 12 weeks with 9.4 T MRI (n=70). Grafts were identified by epifluorescence of a transgenic GFP marker and characterized by immunofluorescence. Vascularity and paracrine effects were investigated immunohistochemically. Transplantation of differentiated hESCs improved short, mid-, and long-term cardiac performance and survival, although only cardiomyocytes formed grafts. A mid-term (4 weeks) cardiomyocyte-specific enhancement was associated with elevated vascular density around the graft and attenuated compensatory remodeling. However, increasing the number of hESC-CM for injection did not enhance heart function further. Moreover, we observed that small graft size was associated with a better functional outcome. HESC-CM increased myocardial vascularization and enhanced heart function in mice after MI, but larger graft size was associated with reduced functional improvement. Future studies should focus on advanced delivery strategies and mechanisms of action rather than increasing graft size.


Subject(s)
Embryonic Stem Cells/transplantation , Myocardium/cytology , Myocytes, Cardiac/physiology , Animals , Embryonic Stem Cells/cytology , Graft Survival , Green Fluorescent Proteins/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Myocardial Infarction/therapy , Myocardium/pathology , Myocytes, Cardiac/cytology , Transplantation, Heterologous
19.
Genome Res ; 17(12): 1731-42, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17989250

ABSTRACT

Disregulation of imprinted genes can be associated with tumorigenesis and altered cell differentiation capacity and so could provide adverse outcomes for stem cell applications. Although the maintenance of mouse and primate embryonic stem cells in a pluripotent state has been reported to disrupt the monoallelic expression of several imprinted genes, available data have suggested relatively higher imprint stability in the human equivalents. Identification of 202 heterozygous loci allowed us to examine the allelic expression of 22 imprinted genes in 22 human embryonic stem cell lines. Half of the genes examined (IPW, H19, MEG3, MEST isoforms 1 and 2, PEG10, MESTIT1, NESP55, ATP10A, PHLDA2, IGF2) showed variable allelic expression between lines, indicating vulnerability to disrupted imprinting. However, seven genes showed consistent monoallelic expression (NDN, MAGEL2, SNRPN, PEG3, KCNQ1, KCNQ1OT1, CDKN1C). Furthermore, four genes known to be monoallelic or to exhibit polymorphic imprinting in later-developing human tissues (TP73, IGF2R, WT1, SLC22A18) were always biallelic in hESCs. MEST isoform 1, PEG10, and NESP55 showed an association between the variability observed in interline allelic expression status and the DNA methylation of previously identified regulatory regions. Our results demonstrate gene-specific differences in the stability of imprinted loci in human embryonic stem cells and identify disrupted DNA methylation as one potential mechanism. We conclude the prudence of including comprehensive imprinting analysis in the continued characterization of human embryonic stem cell lines.


Subject(s)
Embryonic Stem Cells/metabolism , Genetic Variation , Genomic Imprinting , Alleles , Animals , Carcinoma, Embryonal/genetics , Cell Line , Cell Line, Tumor , DNA Methylation , Female , Gene Expression Regulation , Genomic Imprinting/physiology , Humans , Male , Mice
20.
Nat Protoc ; 2(10): 2551-67, 2007.
Article in English | MEDLINE | ID: mdl-17947998

ABSTRACT

We have developed a mouse severe combined immunodeficient (SCID) model of myocardial infarction based on permanent coronary artery occlusion that allows long-term functional analysis of engrafted human embryonic stem cell-derived cardiomyocytes, genetically marked with green fluorescent protein (GFP), in the mouse heart. We describe methods for delivery of dissociated cardiomyocytes to the left ventricle that minimize scar formation and visualization and validation of the identity of the engrafted cells using the GFP emission spectrum, and histological techniques compatible with GFP epifluorescence, for monitoring phenotypic changes in the grafts in vivo. In addition, we describe how magnetic resonance imaging can be adapted for use in mice to monitor cardiac function non-invasively and repeatedly. The model can be adapted to include multiple control or other cell populations. The procedure for a cohort of six mice can be completed in a maximum of 13 weeks, depending on follow-up, with 30 h of hands-on time.


Subject(s)
Disease Models, Animal , Embryonic Stem Cells/transplantation , Magnetic Resonance Imaging/methods , Mice , Myocardial Infarction/therapy , Animals , Cell Culture Techniques , Cell Differentiation , Cell Transplantation/instrumentation , Cell Transplantation/methods , Cicatrix , Coronary Occlusion/immunology , Coronary Occlusion/therapy , Cryoultramicrotomy , Embryonic Stem Cells/cytology , Fluorescent Antibody Technique , Green Fluorescent Proteins/analysis , Humans , Immunocompromised Host , Myocardial Infarction/immunology , Myocardium/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/transplantation
SELECTION OF CITATIONS
SEARCH DETAIL