Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
J Pharmacol Toxicol Methods ; 105: 106889, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32565326

ABSTRACT

Safety pharmacology is an essential part of drug development aiming to identify, evaluate and investigate undesirable pharmacodynamic properties of a drug primarily prior to clinical trials. In particular, cardiovascular adverse drug reactions (ADR) have halted many drug development programs. Safety pharmacology has successfully implemented a screening strategy to detect cardiovascular liabilities, but there is room for further refinement. In this setting, we present the INSPIRE project, a European Training Network in safety pharmacology for Early Stage Researchers (ESRs), funded by the European Commission's H2020-MSCA-ITN programme. INSPIRE has recruited 15 ESR fellows that will conduct an individual PhD-research project for a period of 36 months. INSPIRE aims to be complementary to ongoing research initiatives. With this as a goal, an inventory of collaborative research initiatives in safety pharmacology was created and the ESR projects have been designed to be complementary to this roadmap. Overall, INSPIRE aims to improve cardiovascular safety evaluation, either by investigating technological innovations or by adding mechanistic insight in emerging safety concerns, as observed in the field of cardio-oncology. Finally, in addition to its hands-on research pillar, INSPIRE will organize a number of summer schools and workshops that will be open to the wider community as well. In summary, INSPIRE aims to foster both research and training in safety pharmacology and hopes to inspire the future generation of safety scientists.


Subject(s)
Cardiovascular System/drug effects , Drug Development/methods , Drug-Related Side Effects and Adverse Reactions/prevention & control , Pharmacology/methods , Humans , Safety
2.
Stem Cell Reports ; 12(6): 1232-1241, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31105048

ABSTRACT

Chagas disease (ChD) is one of the most neglected tropical diseases, with cardiomyopathy being the main cause of death in Trypanosoma cruzi-infected patients. As the parasite actively replicates in cardiomyocytes (CMs), the heart remains a key target organ in the pathogenesis of ChD. Here we modeled ChD using human induced pluripotent stem cell-derived CMs (iPSC-CMs) to understand the complex interplay between the parasite and host cells. We showed that iPSC-CMs can get infected with the T. cruzi Y strain and that all parasite cycle stages can be identified in our model system. Importantly, characterization of T. cruzi-infected iPSC-CMs showed significant changes in their gene expression profile, cell contractility, and distribution of key cardiac markers. Moreover, these infected iPSC-CMs exhibited a pro-inflammatory profile as indicated by significantly elevated cytokine levels and cell-trafficking regulators. We believe our iPSC-CM model is a valuable platform to explore new treatment strategies for ChD.


Subject(s)
Chagas Cardiomyopathy/metabolism , Induced Pluripotent Stem Cells , Models, Biological , Myocytes, Cardiac , Trypanosoma cruzi/metabolism , Chagas Cardiomyopathy/pathology , Chagas Cardiomyopathy/therapy , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/parasitology , Induced Pluripotent Stem Cells/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/parasitology , Myocytes, Cardiac/pathology
3.
Stem Cells ; 36(2): 265-277, 2018 02.
Article in English | MEDLINE | ID: mdl-29086457

ABSTRACT

The ability to differentiate human pluripotent stem cells (hPSCs) into cardiomyocytes (CMs) makes them an attractive source for repairing injured myocardium, disease modeling, and drug testing. Although current differentiation protocols yield hPSC-CMs to >90% efficiency, hPSC-CMs exhibit immature characteristics. With the goal of overcoming this limitation, we tested the effects of varying passive stretch on engineered heart muscle (EHM) structural and functional maturation, guided by computational modeling. Human embryonic stem cells (hESCs, H7 line) or human induced pluripotent stem cells (IMR-90 line) were differentiated to hPSC-derived cardiomyocytes (hPSC-CMs) in vitro using a small molecule based protocol. hPSC-CMs were characterized by troponin+ flow cytometry as well as electrophysiological measurements. Afterwards, 1.2 × 106 hPSC-CMs were mixed with 0.4 × 106 human fibroblasts (IMR-90 line) (3:1 ratio) and type-I collagen. The blend was cast into custom-made 12-mm long polydimethylsiloxane reservoirs to vary nominal passive stretch of EHMs to 5, 7, or 9 mm. EHM characteristics were monitored for up to 50 days, with EHMs having a passive stretch of 7 mm giving the most consistent formation. Based on our initial macroscopic observations of EHM formation, we created a computational model that predicts the stress distribution throughout EHMs, which is a function of cellular composition, cellular ratio, and geometry. Based on this predictive modeling, we show cell alignment by immunohistochemistry and coordinated calcium waves by calcium imaging. Furthermore, coordinated calcium waves and mechanical contractions were apparent throughout entire EHMs. The stiffness and active forces of hPSC-derived EHMs are comparable with rat neonatal cardiomyocyte-derived EHMs. Three-dimensional EHMs display increased expression of mature cardiomyocyte genes including sarcomeric protein troponin-T, calcium and potassium ion channels, ß-adrenergic receptors, and t-tubule protein caveolin-3. Passive stretch affects the structural and functional maturation of EHMs. Based on our predictive computational modeling, we show how to optimize cell alignment and calcium dynamics within EHMs. These findings provide a basis for the rational design of EHMs, which enables future scale-up productions for clinical use in cardiovascular tissue engineering. Stem Cells 2018;36:265-277.


Subject(s)
Computational Biology/methods , Myocardium/cytology , Cell Line , Flow Cytometry , Humans , Myocardium/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Tissue Engineering/methods
4.
Cell Rep ; 20(8): 1978-1990, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28834758

ABSTRACT

There is growing interest in using embryonic stem cell (ESC) and induced pluripotent stem cell (iPSC) derivatives for tissue regeneration. However, an increased understanding of human immune responses to stem cell-derived allografts is necessary for maintaining long-term graft persistence. To model this alloimmunity, humanized mice engrafted with human hematopoietic and immune cells could prove to be useful. In this study, an in-depth analysis of graft-infiltrating human lymphocytes and splenocytes revealed that humanized mice incompletely model human immune responses toward allogeneic stem cells and their derivatives. Furthermore, using an "allogenized" mouse model, we show the feasibility of reconstituting immunodeficient mice with a functional mouse immune system and describe a key role of innate immune cells in the rejection of mouse stem cell allografts.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Immunity, Innate/immunology , Pluripotent Stem Cells/metabolism , Transplantation Conditioning/methods , Animals , Disease Models, Animal , Graft Rejection , Humans , Mice
5.
Circ Res ; 120(10): 1561-1571, 2017 May 12.
Article in English | MEDLINE | ID: mdl-28246128

ABSTRACT

RATIONALE: Targeted genetic engineering using programmable nucleases such as transcription activator-like effector nucleases (TALENs) is a valuable tool for precise, site-specific genetic modification in the human genome. OBJECTIVE: The emergence of novel technologies such as human induced pluripotent stem cells (iPSCs) and nuclease-mediated genome editing represent a unique opportunity for studying cardiovascular diseases in vitro. METHODS AND RESULTS: By incorporating extensive literature and database searches, we designed a collection of TALEN constructs to knockout 88 human genes that are associated with cardiomyopathies and congenital heart diseases. The TALEN pairs were designed to induce double-strand DNA break near the starting codon of each gene that either disrupted the start codon or introduced a frameshift mutation in the early coding region, ensuring faithful gene knockout. We observed that all the constructs were active and disrupted the target locus at high frequencies. To illustrate the utility of the TALEN-mediated knockout technique, 6 individual genes (TNNT2, LMNA/C, TBX5, MYH7, ANKRD1, and NKX2.5) were knocked out with high efficiency and specificity in human iPSCs. By selectively targeting a pathogenic mutation (TNNT2 p.R173W) in patient-specific iPSC-derived cardiac myocytes, we demonstrated that the knockout strategy ameliorates the dilated cardiomyopathy phenotype in vitro. In addition, we modeled the Holt-Oram syndrome in iPSC-cardiac myocytes in vitro and uncovered novel pathways regulated by TBX5 in human cardiac myocyte development. CONCLUSIONS: Collectively, our study illustrates the powerful combination of iPSCs and genome editing technologies for understanding the biological function of genes, and the pathological significance of genetic variants in human cardiovascular diseases. The methods, strategies, constructs, and iPSC lines developed in this study provide a validated, readily available resource for cardiovascular research.


Subject(s)
Cardiovascular Diseases/genetics , Gene Knockout Techniques/methods , Gene Library , Genetic Engineering/methods , Induced Pluripotent Stem Cells/physiology , Base Sequence , Cardiovascular Diseases/therapy , Cells, Cultured , Gene Targeting/methods , Humans , Induced Pluripotent Stem Cells/transplantation
6.
Sci Transl Med ; 9(377)2017 02 15.
Article in English | MEDLINE | ID: mdl-28202772

ABSTRACT

Tyrosine kinase inhibitors (TKIs), despite their efficacy as anticancer therapeutics, are associated with cardiovascular side effects ranging from induced arrhythmias to heart failure. We used human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), generated from 11 healthy individuals and 2 patients receiving cancer treatment, to screen U.S. Food and Drug Administration-approved TKIs for cardiotoxicities by measuring alterations in cardiomyocyte viability, contractility, electrophysiology, calcium handling, and signaling. With these data, we generated a "cardiac safety index" to reflect the cardiotoxicities of existing TKIs. TKIs with low cardiac safety indices exhibit cardiotoxicity in patients. We also derived endothelial cells (hiPSC-ECs) and cardiac fibroblasts (hiPSC-CFs) to examine cell type-specific cardiotoxicities. Using high-throughput screening, we determined that vascular endothelial growth factor receptor 2 (VEGFR2)/platelet-derived growth factor receptor (PDGFR)-inhibiting TKIs caused cardiotoxicity in hiPSC-CMs, hiPSC-ECs, and hiPSC-CFs. With phosphoprotein analysis, we determined that VEGFR2/PDGFR-inhibiting TKIs led to a compensatory increase in cardioprotective insulin and insulin-like growth factor (IGF) signaling in hiPSC-CMs. Up-regulating cardioprotective signaling with exogenous insulin or IGF1 improved hiPSC-CM viability during cotreatment with cardiotoxic VEGFR2/PDGFR-inhibiting TKIs. Thus, hiPSC-CMs can be used to screen for cardiovascular toxicities associated with anticancer TKIs, and the results correlate with clinical phenotypes. This approach provides unexpected insights, as illustrated by our finding that toxicity can be alleviated via cardioprotective insulin/IGF signaling.


Subject(s)
Cardiotoxicity/pathology , High-Throughput Screening Assays/methods , Induced Pluripotent Stem Cells/metabolism , Protein Kinase Inhibitors/toxicity , Biomarkers/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Models, Biological , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Sarcomeres/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor Receptor-2/metabolism
7.
Microsyst Nanoeng ; 3: 16080, 2017.
Article in English | MEDLINE | ID: mdl-31057850

ABSTRACT

The measurement of the electrophysiology of human pluripotent stem cell-derived cardiomyocytes is critical for their biomedical applications, from disease modeling to drug screening. Yet, a method that enables the high-throughput intracellular electrophysiology measurement of single cardiomyocytes in adherent culture is not available. To address this area, we have fabricated vertical nanopillar electrodes that can record intracellular action potentials from up to 60 single beating cardiomyocytes. Intracellular access is achieved by highly localized electroporation, which allows for low impedance electrical access to the intracellular voltage. Herein, we demonstrate that this method provides the accurate measurement of the shape and duration of intracellular action potentials, validated by patch clamp, and can facilitate cellular drug screening and disease modeling using human pluripotent stem cells. This study validates the use of nanopillar electrodes for myriad further applications of human pluripotent stem cell-derived cardiomyocytes such as cardiomyocyte maturation monitoring and electrophysiology-contractile force correlation.

9.
Cell Stem Cell ; 19(3): 311-25, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27545504

ABSTRACT

Understanding individual susceptibility to drug-induced cardiotoxicity is key to improving patient safety and preventing drug attrition. Human induced pluripotent stem cells (hiPSCs) enable the study of pharmacological and toxicological responses in patient-specific cardiomyocytes (CMs) and may serve as preclinical platforms for precision medicine. Transcriptome profiling in hiPSC-CMs from seven individuals lacking known cardiovascular disease-associated mutations and in three isogenic human heart tissue and hiPSC-CM pairs showed greater inter-patient variation than intra-patient variation, verifying that reprogramming and differentiation preserve patient-specific gene expression, particularly in metabolic and stress-response genes. Transcriptome-based toxicology analysis predicted and risk-stratified patient-specific susceptibility to cardiotoxicity, and functional assays in hiPSC-CMs using tacrolimus and rosiglitazone, drugs targeting pathways predicted to produce cardiotoxicity, validated inter-patient differential responses. CRISPR/Cas9-mediated pathway correction prevented drug-induced cardiotoxicity. Our data suggest that hiPSC-CMs can be used in vitro to predict and validate patient-specific drug safety and efficacy, potentially enabling future clinical approaches to precision medicine.


Subject(s)
Gene Expression Profiling , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/cytology , Tacrolimus/adverse effects , Thiazolidinediones/adverse effects , CRISPR-Cas Systems/genetics , Cell Death/drug effects , Gene Editing , Genome, Human , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Humans , Inverted Repeat Sequences/genetics , Myocytes, Cardiac/metabolism , Rosiglitazone , Treatment Outcome
10.
Physiol Rev ; 96(3): 1093-126, 2016 07.
Article in English | MEDLINE | ID: mdl-27335446

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) have revolutionized the field of human disease modeling, with an enormous potential to serve as paradigm shifting platforms for preclinical trials, personalized clinical diagnosis, and drug treatment. In this review, we describe how hiPSCs could transition cardiac healthcare away from simple disease diagnosis to prediction and prevention, bridging the gap between basic and clinical research to bring the best science to every patient.


Subject(s)
Cardiovascular Diseases/therapy , Induced Pluripotent Stem Cells , Precision Medicine , Humans
11.
Nat Med ; 22(5): 547-56, 2016 05.
Article in English | MEDLINE | ID: mdl-27089514

ABSTRACT

Doxorubicin is an anthracycline chemotherapy agent effective in treating a wide range of malignancies, but it causes a dose-related cardiotoxicity that can lead to heart failure in a subset of patients. At present, it is not possible to predict which patients will be affected by doxorubicin-induced cardiotoxicity (DIC). Here we demonstrate that patient-specific human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) can recapitulate the predilection to DIC of individual patients at the cellular level. hiPSC-CMs derived from individuals with breast cancer who experienced DIC were consistently more sensitive to doxorubicin toxicity than hiPSC-CMs from patients who did not experience DIC, with decreased cell viability, impaired mitochondrial and metabolic function, impaired calcium handling, decreased antioxidant pathway activity, and increased reactive oxygen species production. Taken together, our data indicate that hiPSC-CMs are a suitable platform to identify and characterize the genetic basis and molecular mechanisms of DIC.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Doxorubicin/pharmacology , Heart Failure/chemically induced , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Adult , Aged , Antibiotics, Antineoplastic/adverse effects , Calcium/metabolism , Cardiotoxicity/genetics , Cell Survival/drug effects , DNA Damage/drug effects , Disease Susceptibility , Doxorubicin/adverse effects , Female , Flow Cytometry , Fluorescent Antibody Technique , Heart Failure/genetics , Humans , Induced Pluripotent Stem Cells , Membrane Potential, Mitochondrial/drug effects , Middle Aged , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Polymorphism, Single Nucleotide , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Transcriptome
12.
Nat Rev Cardiol ; 13(6): 333-49, 2016 06.
Article in English | MEDLINE | ID: mdl-27009425

ABSTRACT

The advent of human induced pluripotent stem cell (hiPSC) technology has revitalized the efforts in the past decade to realize more fully the potential of human embryonic stem cells for scientific research. Adding to the possibility of generating an unlimited amount of any cell type of interest, hiPSC technology now enables the derivation of cells with patient-specific phenotypes. Given the introduction and implementation of the large-scale Precision Medicine Initiative, hiPSC technology will undoubtedly have a vital role in the advancement of cardiovascular research and medicine. In this Review, we summarize the progress that has been made in the field of hiPSC technology, with particular emphasis on cardiovascular disease modelling and drug development. The growing roles of hiPSC technology in the practice of precision medicine will also be discussed.


Subject(s)
Cardiovascular Diseases/drug therapy , Induced Pluripotent Stem Cells , Models, Cardiovascular , Precision Medicine , Cardiovascular Diseases/genetics , Cardiovascular Diseases/prevention & control , Cell Differentiation , Cellular Reprogramming Techniques , Drug Discovery , Endothelial Cells/cytology , Humans , Myocytes, Cardiac/cytology , Myocytes, Smooth Muscle/cytology
13.
Methods Mol Biol ; 1353: 119-30, 2016.
Article in English | MEDLINE | ID: mdl-25690476

ABSTRACT

The generation of cardiomyocytes from human induced pluripotent stem cells (hiPSCs) provides a source of cells that accurately recapitulate the human cardiac pathophysiology. The application of these cells allows for modeling of cardiovascular diseases, providing a novel understanding of human disease mechanisms and assessment of therapies. Here, we describe a stepwise protocol developed in our laboratory for the generation of hiPSCs from patients with a specific disease phenotype, long-term hiPSC culture and cryopreservation, differentiation of hiPSCs to cardiomyocytes, and assessment of disease phenotypes. Our protocol combines a number of innovative tools that include a codon-optimized mini intronic plasmid (CoMiP), chemically defined culture conditions to achieve high efficiencies of reprogramming and differentiation, and calcium imaging for assessment of cardiomyocyte phenotypes. Thus, this protocol provides a complete guide to use a patient cohort on a testable cardiomyocyte platform for pharmacological drug assessment.


Subject(s)
Cardiomyopathy, Dilated/pathology , Cellular Reprogramming , Fibroblasts/cytology , Induced Pluripotent Stem Cells/cytology , Models, Biological , Myocytes, Cardiac/cytology , Antigens, Surface/genetics , Antigens, Surface/metabolism , Biomarkers/metabolism , Calcium/metabolism , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Cell Differentiation , Cryopreservation , Dermis/cytology , Dermis/metabolism , Fibroblasts/metabolism , Gene Expression , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Molecular Imaging , Mutation , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Plasmids/chemistry , Plasmids/metabolism , Primary Cell Culture , Proteoglycans/genetics , Proteoglycans/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Serial Passage , Troponin T/genetics , Troponin T/metabolism
14.
Cell Stem Cell ; 17(1): 89-100, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26095046

ABSTRACT

ß-adrenergic signaling pathways mediate key aspects of cardiac function. Its dysregulation is associated with a range of cardiac diseases, including dilated cardiomyopathy (DCM). Previously, we established an iPSC model of familial DCM from patients with a mutation in TNNT2, a sarcomeric protein. Here, we found that the ß-adrenergic agonist isoproterenol induced mature ß-adrenergic signaling in iPSC-derived cardiomyocytes (iPSC-CMs) but that this pathway was blunted in DCM iPSC-CMs. Although expression levels of several ß-adrenergic signaling components were unaltered between control and DCM iPSC-CMs, we found that phosphodiesterases (PDEs) 2A and PDE3A were upregulated in DCM iPSC-CMs and that PDE2A was also upregulated in DCM patient tissue. We further discovered increased nuclear localization of mutant TNNT2 and epigenetic modifications of PDE genes in both DCM iPSC-CMs and patient tissue. Notably, pharmacologic inhibition of PDE2A and PDE3A restored cAMP levels and ameliorated the impaired ß-adrenergic signaling of DCM iPSC-CMs, suggesting therapeutic potential.


Subject(s)
Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/genetics , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Induced Pluripotent Stem Cells/physiology , Receptors, Adrenergic, beta/metabolism , Adrenergic beta-Agonists/pharmacology , Cardiomyopathy, Dilated/pathology , Cell Differentiation , Cells, Cultured , Epigenesis, Genetic , Heart Rate/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/pathology , Isoproterenol/pharmacology , Models, Cardiovascular , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Signal Transduction , Troponin T/genetics , Troponin T/metabolism , Up-Regulation
15.
Stem Cell Reports ; 3(5): 905-14, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25418732

ABSTRACT

Cardiomyocytes from human embryonic stem cells (hESC-CMs) and induced pluripotent stem cells (hiPSC-CMs) represent new models for drug discovery. Although hypertrophy is a high-priority target, we found that hiPSC-CMs were systematically unresponsive to hypertrophic signals such as the α-adrenoceptor (αAR) agonist phenylephrine (PE) compared to hESC-CMs. We investigated signaling at multiple levels to understand the underlying mechanism of this differential responsiveness. The expression of the normal α1AR gene, ADRA1A, was reversibly silenced during differentiation, accompanied by ADRA1B upregulation in either cell type. ADRA1B signaling was intact in hESC-CMs, but not in hiPSC-CMs. We observed an increased tonic activity of inhibitory kinase pathways in hiPSC-CMs, and inhibition of antihypertrophic kinases revealed hypertrophic increases. There is tonic suppression of cell growth in hiPSC-CMs, but not hESC-CMs, limiting their use in investigation of hypertrophic signaling. These data raise questions regarding the hiPSC-CM as a valid model for certain aspects of cardiac disease.


Subject(s)
Adrenergic Agents/pharmacology , Cell Size/drug effects , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Adrenergic alpha-Agonists/pharmacology , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Gene Expression/drug effects , Humans , Hypertrophy , Imidazoles/pharmacology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Isoproterenol/pharmacology , Microscopy, Confocal , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Phenylephrine/pharmacology , Receptors, Adrenergic, alpha-1/genetics , Receptors, Adrenergic, alpha-1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics
16.
Circ Res ; 115(6): 556-66, 2014 Aug 29.
Article in English | MEDLINE | ID: mdl-25015077

ABSTRACT

RATIONALE: Viral myocarditis is a life-threatening illness that may lead to heart failure or cardiac arrhythmias. A major causative agent for viral myocarditis is the B3 strain of coxsackievirus, a positive-sense RNA enterovirus. However, human cardiac tissues are difficult to procure in sufficient enough quantities for studying the mechanisms of cardiac-specific viral infection. OBJECTIVE: This study examined whether human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) could be used to model the pathogenic processes of coxsackievirus-induced viral myocarditis and to screen antiviral therapeutics for efficacy. METHODS AND RESULTS: hiPSC-CMs were infected with a luciferase-expressing coxsackievirus B3 strain (CVB3-Luc). Brightfield microscopy, immunofluorescence, and calcium imaging were used to characterize virally infected hiPSC-CMs for alterations in cellular morphology and calcium handling. Viral proliferation in hiPSC-CMs was quantified using bioluminescence imaging. Antiviral compounds including interferonß1, ribavirin, pyrrolidine dithiocarbamate, and fluoxetine were tested for their capacity to abrogate CVB3-Luc proliferation in hiPSC-CMs in vitro. The ability of these compounds to reduce CVB3-Luc proliferation in hiPSC-CMs was consistent with reported drug effects in previous studies. Mechanistic analyses via gene expression profiling of hiPSC-CMs infected with CVB3-Luc revealed an activation of viral RNA and protein clearance pathways after interferonß1 treatment. CONCLUSIONS: This study demonstrates that hiPSC-CMs express the coxsackievirus and adenovirus receptor, are susceptible to coxsackievirus infection, and can be used to predict antiviral drug efficacy. Our results suggest that the hiPSC-CM/CVB3-Luc assay is a sensitive platform that can screen novel antiviral therapeutics for their effectiveness in a high-throughput fashion.


Subject(s)
Antiviral Agents/therapeutic use , Enterovirus B, Human/isolation & purification , Enterovirus Infections/drug therapy , Models, Cardiovascular , Myocarditis/drug therapy , Myocytes, Cardiac/pathology , Pluripotent Stem Cells/pathology , Antiviral Agents/pharmacology , Calcium/metabolism , Cell Proliferation , Cells, Cultured , Drug Evaluation, Preclinical , Enterovirus Infections/metabolism , Humans , In Vitro Techniques , Myocarditis/metabolism , Myocarditis/virology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/virology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/virology , RNA, Viral/metabolism , Treatment Outcome
17.
Nat Methods ; 11(8): 855-60, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24930130

ABSTRACT

Existing methods for human induced pluripotent stem cell (hiPSC) cardiac differentiation are efficient but require complex, undefined medium constituents that hinder further elucidation of the molecular mechanisms of cardiomyogenesis. Using hiPSCs derived under chemically defined conditions on synthetic matrices, we systematically developed an optimized cardiac differentiation strategy, using a chemically defined medium consisting of just three components: the basal medium RPMI 1640, L-ascorbic acid 2-phosphate and rice-derived recombinant human albumin. Along with small molecule-based induction of differentiation, this protocol produced contractile sheets of up to 95% TNNT2(+) cardiomyocytes at a yield of up to 100 cardiomyocytes for every input pluripotent cell and was effective in 11 hiPSC lines tested. This chemically defined platform for cardiac specification of hiPSCs will allow the elucidation of cardiomyocyte macromolecular and metabolic requirements and will provide a minimal system for the study of maturation and subtype specification.


Subject(s)
Myocytes, Cardiac/cytology , Cell Differentiation , Culture Media , Humans , Induced Pluripotent Stem Cells/cytology
18.
Sci Transl Med ; 6(239): 239ps6, 2014 Jun 04.
Article in English | MEDLINE | ID: mdl-24898747

ABSTRACT

A major research focus in the field of cardiovascular medicine is the prospect of using stem cells and progenitor cells for cardiac regeneration. With the advent of induced pluripotent stem cell (iPSC) technology, major efforts are also underway to use iPSCs to model heart disease, to screen for new drugs, and to test candidate drugs for cardiotoxicity. Here, we discuss recent advances in the exciting fields of stem cells and cardiovascular disease.


Subject(s)
Drug Discovery , Heart Diseases/therapy , Models, Cardiovascular , Stem Cell Transplantation , Stem Cells/cytology , Humans , Wound Healing
19.
Circ Res ; 114(1): 21-7, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24385505

ABSTRACT

Cardiac regeneration strategies and de novo generation of cardiomyocytes have long been significant areas of research interest in cardiovascular medicine. In this review, we outline a variety of common cell sources and methods used to regenerate cardiomyocytes and highlight the important role that key Circulation Research articles have played in this flourishing field.


Subject(s)
Heart/physiology , Myocytes, Cardiac/cytology , Regeneration , Stem Cell Transplantation , Animals , Cell Differentiation , Heart Diseases/surgery , History, 20th Century , History, 21st Century , Humans , Stem Cell Research/history , Stem Cell Transplantation/history , Stem Cell Transplantation/trends , Stem Cells/classification , Stem Cells/cytology
20.
Eur Heart J ; 35(16): 1078-87, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23470493

ABSTRACT

AIMS: Long-QT syndromes (LQTS) are mostly autosomal-dominant congenital disorders associated with a 1:1000 mutation frequency, cardiac arrest, and sudden death. We sought to use cardiomyocytes derived from human-induced pluripotency stem cells (hiPSCs) as an in vitro model to develop and evaluate gene-based therapeutics for the treatment of LQTS. METHODS AND RESULTS: We produced LQTS-type 2 (LQT2) hiPSC cardiomyocytes carrying a KCNH2 c.G1681A mutation in a IKr ion-channel pore, which caused impaired glycosylation and channel transport to cell surface. Allele-specific RNA interference (RNAi) directed towards the mutated KCNH2 mRNA caused knockdown, while leaving the wild-type mRNA unaffected. Electrophysiological analysis of patient-derived LQT2 hiPSC cardiomyocytes treated with mutation-specific siRNAs showed normalized action potential durations (APDs) and K(+) currents with the concurrent rescue of spontaneous and drug-induced arrhythmias (presented as early-afterdepolarizations). CONCLUSIONS: These findings provide in vitro evidence that allele-specific RNAi can rescue diseased phenotype in LQTS cardiomyocytes. This is a potentially novel route for the treatment of many autosomal-dominant-negative disorders, including those of the heart.


Subject(s)
Ether-A-Go-Go Potassium Channels/genetics , Long QT Syndrome/genetics , Myocytes, Cardiac/physiology , Pluripotent Stem Cells/physiology , RNA Interference/physiology , ERG1 Potassium Channel , Electrophysiological Phenomena/genetics , Gene Expression/genetics , Gene Knockdown Techniques , Genetic Therapy , Humans , Long QT Syndrome/physiopathology , Long QT Syndrome/therapy , Mutation, Missense/genetics , Phenotype , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL