Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
2.
Cell Stem Cell ; 30(4): 396-414.e9, 2023 04 06.
Article in English | MEDLINE | ID: mdl-37028405

ABSTRACT

Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) offer a promising cell-based therapy for myocardial infarction. However, the presence of transitory ventricular arrhythmias, termed engraftment arrhythmias (EAs), hampers clinical applications. We hypothesized that EA results from pacemaker-like activity of hPSC-CMs associated with their developmental immaturity. We characterized ion channel expression patterns during maturation of transplanted hPSC-CMs and used pharmacology and genome editing to identify those responsible for automaticity in vitro. Multiple engineered cell lines were then transplanted in vivo into uninjured porcine hearts. Abolishing depolarization-associated genes HCN4, CACNA1H, and SLC8A1, along with overexpressing hyperpolarization-associated KCNJ2, creates hPSC-CMs that lack automaticity but contract when externally stimulated. When transplanted in vivo, these cells engrafted and coupled electromechanically with host cardiomyocytes without causing sustained EAs. This study supports the hypothesis that the immature electrophysiological prolife of hPSC-CMs mechanistically underlies EA. Thus, targeting automaticity should improve the safety profile of hPSC-CMs for cardiac remuscularization.


Subject(s)
Gene Editing , Myocytes, Cardiac , Humans , Animals , Swine , Myocytes, Cardiac/metabolism , Cell Line , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/therapy , Arrhythmias, Cardiac/metabolism , Cell- and Tissue-Based Therapy , Cell Differentiation/genetics
3.
Stem Cell Reports ; 18(1): 159-174, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36493778

ABSTRACT

Vascular endothelial cells are a mesoderm-derived lineage with many essential functions, including angiogenesis and coagulation. The gene-regulatory mechanisms underpinning endothelial specialization are largely unknown, as are the roles of chromatin organization in regulating endothelial cell transcription. To investigate the relationships between chromatin organization and gene expression, we induced endothelial cell differentiation from human pluripotent stem cells and performed Hi-C and RNA-sequencing assays at specific time points. Long-range intrachromosomal contacts increase over the course of differentiation, accompanied by widespread heteroeuchromatic compartment transitions that are tightly associated with transcription. Dynamic topologically associating domain boundaries strengthen and converge on an endothelial cell state, and function to regulate gene expression. Chromatin pairwise point interactions (DNA loops) increase in frequency during differentiation and are linked to the expression of genes essential to vascular biology. Chromatin dynamics guide transcription in endothelial cell development and promote the divergence of endothelial cells from cardiomyocytes.


Subject(s)
Chromatin , Endothelial Cells , Humans , Cell Differentiation/genetics , Gene Expression Regulation
4.
Stem Cell Reports ; 16(10): 2473-2487, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34506727

ABSTRACT

Heart failure remains a significant cause of morbidity and mortality following myocardial infarction. Cardiac remuscularization with transplantation of human pluripotent stem cell-derived cardiomyocytes is a promising preclinical therapy to restore function. Recent large animal data, however, have revealed a significant risk of engraftment arrhythmia (EA). Although transient, the risk posed by EA presents a barrier to clinical translation. We hypothesized that clinically approved antiarrhythmic drugs can prevent EA-related mortality as well as suppress tachycardia and arrhythmia burden. This study uses a porcine model to provide proof-of-concept evidence that a combination of amiodarone and ivabradine can effectively suppress EA. None of the nine treated subjects experienced the primary endpoint of cardiac death, unstable EA, or heart failure compared with five out of eight (62.5%) in the control cohort (hazard ratio = 0.00; 95% confidence interval: 0-0.297; p = 0.002). Pharmacologic treatment of EA may be a viable strategy to improve safety and allow further clinical development of cardiac remuscularization therapy.


Subject(s)
Amiodarone/therapeutic use , Arrhythmias, Cardiac/drug therapy , Ivabradine/therapeutic use , Myocardial Infarction/drug therapy , Myocytes, Cardiac/transplantation , Stem Cell Transplantation/adverse effects , Tachycardia/drug therapy , Animals , Anti-Arrhythmia Agents/therapeutic use , Cell Line , Cell- and Tissue-Based Therapy/adverse effects , Disease Models, Animal , Drug Combinations , Humans , Male , Pluripotent Stem Cells/transplantation , Swine
5.
Stem Cell Reports ; 16(3): 478-492, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33657418

ABSTRACT

COVID-19 patients often develop severe cardiovascular complications, but it remains unclear if these are caused directly by viral infection or are secondary to a systemic response. Here, we examine the cardiac tropism of SARS-CoV-2 in human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) and smooth muscle cells (hPSC-SMCs). We find that that SARS-CoV-2 selectively infects hPSC-CMs through the viral receptor ACE2, whereas in hPSC-SMCs there is minimal viral entry or replication. After entry into cardiomyocytes, SARS-CoV-2 is assembled in lysosome-like vesicles and egresses via bulk exocytosis. The viral transcripts become a large fraction of cellular mRNA while host gene expression shifts from oxidative to glycolytic metabolism and upregulates chromatin modification and RNA splicing pathways. Most importantly, viral infection of hPSC-CMs progressively impairs both their electrophysiological and contractile function, and causes widespread cell death. These data support the hypothesis that COVID-19-related cardiac symptoms can result from a direct cardiotoxic effect of SARS-CoV-2.


Subject(s)
COVID-19/virology , Induced Pluripotent Stem Cells/virology , Myocytes, Cardiac/virology , SARS-CoV-2/pathogenicity , Cells, Cultured , Humans , RNA Splicing/genetics , RNA, Messenger/genetics , SARS-CoV-2/genetics , Virus Internalization
6.
Cardiovasc Res ; 116(2): 368-382, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31049579

ABSTRACT

AIMS: Heart failure invariably affects patients with various forms of muscular dystrophy (MD), but the onset and molecular sequelae of altered structure and function resulting from full-length dystrophin (Dp427) deficiency in MD heart tissue are poorly understood. To better understand the role of dystrophin in cardiomyocyte development and the earliest phase of Duchenne muscular dystrophy (DMD) cardiomyopathy, we studied human cardiomyocytes differentiated from induced pluripotent stem cells (hiPSC-CMs) obtained from the urine of a DMD patient. METHODS AND RESULTS: The contractile properties of patient-specific hiPSC-CMs, with no detectable dystrophin (DMD-CMs with a deletion of exon 50), were compared to CMs containing a CRISPR-Cas9 mediated deletion of a single G base at position 263 of the dystrophin gene (c.263delG-CMs) isogenic to the parental line of hiPSC-CMs from a healthy individual. We hypothesized that the absence of a dystrophin-actin linkage would adversely affect myofibril and cardiomyocyte structure and function. Cardiomyocyte maturation was driven by culturing long-term (80-100 days) on a nanopatterned surface, which resulted in hiPSC-CMs with adult-like dimensions and aligned myofibrils. CONCLUSIONS: Our data demonstrate that lack of Dp427 results in reduced myofibril contractile tension, slower relaxation kinetics, and to Ca2+ handling abnormalities, similar to DMD cells, suggesting either retarded or altered maturation of cardiomyocyte structures associated with these functions. This study offers new insights into the functional consequences of Dp427 deficiency at an early stage of cardiomyocyte development in both patient-derived and CRISPR-generated models of dystrophin deficiency.


Subject(s)
Cardiomyopathies/etiology , Cell Differentiation , Dystrophin/deficiency , Induced Pluripotent Stem Cells/metabolism , Muscular Dystrophy, Duchenne/complications , Myocardial Contraction , Myocytes, Cardiac/metabolism , Myofibrils/metabolism , Calcium Signaling , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/physiopathology , Case-Control Studies , Cell Line , Dystrophin/genetics , Humans , Induced Pluripotent Stem Cells/ultrastructure , Kinetics , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Myocytes, Cardiac/ultrastructure , Myofibrils/ultrastructure
7.
Circulation ; 140(20): 1647-1660, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31587567

ABSTRACT

BACKGROUND: The giant sarcomere protein titin is important in both heart health and disease. Mutations in the gene encoding for titin (TTN) are the leading known cause of familial dilated cardiomyopathy. The uneven distribution of these mutations within TTN motivated us to seek a more complete understanding of this gene and the isoforms it encodes in cardiomyocyte (CM) sarcomere formation and function. METHODS: To investigate the function of titin in human CMs, we used CRISPR/Cas9 to generate homozygous truncations in the Z disk (TTN-Z-/-) and A-band (TTN-A-/-) regions of the TTN gene in human induced pluripotent stem cells. The resulting CMs were characterized with immunostaining, engineered heart tissue mechanical measurements, and single-cell force and calcium measurements. RESULTS: After differentiation, we were surprised to find that despite the more upstream mutation, TTN-Z-/--CMs had sarcomeres and visibly contracted, whereas TTN-A-/--CMs did not. We hypothesized that sarcomere formation was caused by the expression of a recently discovered isoform of titin, Cronos, which initiates downstream of the truncation in TTN-Z-/--CMs. Using a custom Cronos antibody, we demonstrate that this isoform is expressed and integrated into myofibrils in human CMs. TTN-Z-/--CMs exclusively express Cronos titin, but these cells produce lower contractile force and have perturbed myofibril bundling compared with controls expressing both full-length and Cronos titin. Cronos titin is highly expressed in human fetal cardiac tissue, and when knocked out in human induced pluripotent stem cell derived CMs, these cells exhibit reduced contractile force and myofibrillar disarray despite the presence of full-length titin. CONCLUSIONS: We demonstrate that Cronos titin is expressed in developing human CMs and is able to support partial sarcomere formation in the absence of full-length titin. Furthermore, Cronos titin is necessary for proper sarcomere function in human induced pluripotent stem cell derived CMs. Additional investigation is necessary to understand the molecular mechanisms of this novel isoform and how it contributes to human cardiac disease.


Subject(s)
Connectin/metabolism , Induced Pluripotent Stem Cells/metabolism , Myocardial Contraction , Myocytes, Cardiac/metabolism , Sarcomeres/metabolism , CRISPR-Cas Systems , Calcium Signaling , Cells, Cultured , Connectin/genetics , Fetal Heart/metabolism , Gene Editing , Genotype , Humans , Mutation , Myocardial Contraction/genetics , Phenotype
8.
Stem Cell Reports ; 13(4): 657-668, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31564645

ABSTRACT

Although human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have emerged as a novel platform for heart regeneration, disease modeling, and drug screening, their immaturity significantly hinders their application. A hallmark of postnatal cardiomyocyte maturation is the metabolic substrate switch from glucose to fatty acids. We hypothesized that fatty acid supplementation would enhance hPSC-CM maturation. Fatty acid treatment induces cardiomyocyte hypertrophy and significantly increases cardiomyocyte force production. The improvement in force generation is accompanied by enhanced calcium transient peak height and kinetics, and by increased action potential upstroke velocity and membrane capacitance. Fatty acids also enhance mitochondrial respiratory reserve capacity. RNA sequencing showed that fatty acid treatment upregulates genes involved in fatty acid ß-oxidation and downregulates genes in lipid synthesis. Signal pathway analyses reveal that fatty acid treatment results in phosphorylation and activation of multiple intracellular kinases. Thus, fatty acids increase human cardiomyocyte hypertrophy, force generation, calcium dynamics, action potential upstroke velocity, and oxidative capacity. This enhanced maturation should facilitate hPSC-CM usage for cell therapy, disease modeling, and drug/toxicity screens.


Subject(s)
Cell Differentiation , Fatty Acids/metabolism , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Calcium/metabolism , Carnitine/metabolism , Cell Line , Dietary Supplements , Humans , Kinetics , Membrane Potentials , Mitochondria, Heart/metabolism , Muscle Contraction , Oxidation-Reduction , Oxidative Phosphorylation , Signal Transduction
9.
J Cell Biol ; 218(9): 2919-2944, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31395619

ABSTRACT

Mutations in A-type nuclear lamins cause dilated cardiomyopathy, which is postulated to result from dysregulated gene expression due to changes in chromatin organization into active and inactive compartments. To test this, we performed genome-wide chromosome conformation analyses in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) with a haploinsufficient mutation for lamin A/C. Compared with gene-corrected cells, mutant hiPSC-CMs have marked electrophysiological and contractile alterations, with modest gene expression changes. While large-scale changes in chromosomal topology are evident, differences in chromatin compartmentalization are limited to a few hotspots that escape segregation to the nuclear lamina and inactivation during cardiogenesis. These regions exhibit up-regulation of multiple noncardiac genes including CACNA1A, encoding for neuronal P/Q-type calcium channels. Pharmacological inhibition of the resulting current partially mitigates the electrical alterations. However, chromatin compartment changes do not explain most gene expression alterations in mutant hiPSC-CMs. Thus, global errors in chromosomal compartmentation are not the primary pathogenic mechanism in heart failure due to lamin A/C haploinsufficiency.


Subject(s)
Cardiomyopathy, Dilated , Chromatin Assembly and Disassembly , Chromatin , Haploinsufficiency , Laminin/genetics , Models, Cardiovascular , Myocytes, Cardiac/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Chromatin/genetics , Chromatin/metabolism , Chromatin/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Laminin/metabolism , Myocytes, Cardiac/pathology
10.
Nat Commun ; 10(1): 1538, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30948719

ABSTRACT

Functional changes in spatial genome organization during human development are poorly understood. Here we report a comprehensive profile of nuclear dynamics during human cardiogenesis from pluripotent stem cells by integrating Hi-C, RNA-seq and ATAC-seq. While chromatin accessibility and gene expression show complex on/off dynamics, large-scale genome architecture changes are mostly unidirectional. Many large cardiac genes transition from a repressive to an active compartment during differentiation, coincident with upregulation. We identify a network of such gene loci that increase their association inter-chromosomally, and are targets of the muscle-specific splicing factor RBM20. Genome editing studies show that TTN pre-mRNA, the main RBM20-regulated transcript in the heart, nucleates RBM20 foci that drive spatial proximity between the TTN locus and other inter-chromosomal RBM20 targets such as CACNA1C and CAMK2D. This mechanism promotes RBM20-dependent alternative splicing of the resulting transcripts, indicating the existence of a cardiac-specific trans-interacting chromatin domain (TID) functioning as a splicing factory.


Subject(s)
Alternative Splicing , Cell Differentiation/genetics , RNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Heart/growth & development , Humans , Myocardium/cytology , Myocardium/metabolism , Organogenesis/genetics , Pluripotent Stem Cells , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
11.
Sci Rep ; 9(1): 6433, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31015521

ABSTRACT

Concentration gradients of biochemical stimuli such as morphogens play a critical role in directing cell fate patterning across species and throughout development but are not commonly recapitulated in vitro. While in vitro biomolecule gradients have been generated using customized microfluidic platforms, broad implementation has been limited because these platforms introduce new variables to cell culture such as externally driven flow, culture in a specialized matrix, or extended time for in situ long range diffusion. Here we introduce a method that enables preforming and then transferring user-controlled gradients to cells in standard "open" cultures. Our gradient patterning devices are modular and decoupled from the culture substrate. We find that gradient generation and transfer are predictable by finite element modeling and that device and loading parameters can be used to tune the stimulus pattern. Furthermore, we demonstrate use of these devices to spatially define morphogen signal gradients and direct peri-gastrulation fate stratification of human pluripotent stem cells. This method for extrinsic application of biochemical signal gradients can thus be used to spatially influence cellular fate decisions in a user-controlled manner.


Subject(s)
Body Patterning/physiology , Cell Culture Techniques , Human Umbilical Vein Endothelial Cells/cytology , Induced Pluripotent Stem Cells/cytology , Signal Transduction/physiology , Cell Differentiation , Cell Line , Collagen/chemistry , Drug Combinations , Finite Element Analysis , Gastrulation/physiology , Human Umbilical Vein Endothelial Cells/physiology , Humans , Induced Pluripotent Stem Cells/physiology , Lab-On-A-Chip Devices , Laminin/chemistry , Proteoglycans/chemistry
14.
Nat Biotechnol ; 36(7): 597-605, 2018 08.
Article in English | MEDLINE | ID: mdl-29969440

ABSTRACT

Pluripotent stem cell-derived cardiomyocyte grafts can remuscularize substantial amounts of infarcted myocardium and beat in synchrony with the heart, but in some settings cause ventricular arrhythmias. It is unknown whether human cardiomyocytes can restore cardiac function in a physiologically relevant large animal model. Here we show that transplantation of ∼750 million cryopreserved human embryonic stem cell-derived cardiomyocytes (hESC-CMs) enhances cardiac function in macaque monkeys with large myocardial infarctions. One month after hESC-CM transplantation, global left ventricular ejection fraction improved 10.6 ± 0.9% vs. 2.5 ± 0.8% in controls, and by 3 months there was an additional 12.4% improvement in treated vs. a 3.5% decline in controls. Grafts averaged 11.6% of infarct size, formed electromechanical junctions with the host heart, and by 3 months contained ∼99% ventricular myocytes. A subset of animals experienced graft-associated ventricular arrhythmias, shown by electrical mapping to originate from a point-source acting as an ectopic pacemaker. Our data demonstrate that remuscularization of the infarcted macaque heart with human myocardium provides durable improvement in left ventricular function.


Subject(s)
Cell Differentiation/genetics , Human Embryonic Stem Cells/transplantation , Myocardial Infarction/therapy , Myocytes, Cardiac/transplantation , Animals , Cryopreservation , Disease Models, Animal , Humans , Macaca , Myocardial Infarction/pathology , Myocardium/pathology , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/transplantation , Primates
15.
iScience ; 2: 88-100, 2018 Apr 27.
Article in English | MEDLINE | ID: mdl-29888752

ABSTRACT

Cardiac development requires coordinated biphasic regulation of the WNT/ß-catenin signaling pathway. By intersecting gene expression and loss-of-function siRNA screens we identified Alpha Protein Kinase 2 (ALPK2) as a candidate negative regulator of WNT/ß-catenin signaling in cardiogenesis. In differentiating human embryonic stem cells (hESCs), ALPK2 is highly induced as hESCs transition from mesoderm to cardiac progenitors. Using antisense knockdown and CRISPR/Cas9 mutagenesis in hESCs and zebrafish, we demonstrate that ALPK2 promotes cardiac function and cardiomyocyte differentiation. Quantitative phosphoproteomics, protein expression profiling, and ß-catenin reporter assays demonstrate that loss of ALPK2 led to stabilization of ß-catenin and increased WNT signaling. Furthermore, cardiac defects attributed to ALPK2 depletion can be rescued in a dose-dependent manner by direct inhibition of WNT signaling through the small molecule XAV939. Together, these results demonstrate that ALPK2 regulates ß-catenin-dependent signaling during developmental commitment of cardiomyocytes.

16.
JACC Basic Transl Sci ; 3(6): 728-740, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30623132

ABSTRACT

A novel myosin heavy chain 7 mutation (E848G) identified in a familial cardiomyopathy was studied in patient-specific induced pluripotent stem cell-derived cardiomyocytes. The cardiomyopathic human induced pluripotent stem cell-derived cardiomyocytes exhibited reduced contractile function as single cells and engineered heart tissues, and genome-edited isogenic cells confirmed the pathogenic nature of the E848G mutation. Reduced contractility may result from impaired interaction between myosin heavy chain 7 and cardiac myosin binding protein C.

17.
Cell Rep ; 20(7): 1597-1608, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28813672

ABSTRACT

We analyzed chromatin dynamics and transcriptional activity of human embryonic stem cell (hESC)-derived cardiac progenitor cells (CPCs) and KDR+/CD34+ endothelial cells generated from different mesodermal origins. Using an unbiased algorithm to hierarchically rank genes modulated at the level of chromatin and transcription, we identified candidate regulators of mesodermal lineage determination. HOPX, a non-DNA-binding homeodomain protein, was identified as a candidate regulator of blood-forming endothelial cells. Using HOPX reporter and knockout hESCs, we show that HOPX regulates blood formation. Loss of HOPX does not impact endothelial fate specification but markedly reduces primitive hematopoiesis, acting at least in part through failure to suppress Wnt/ß-catenin signaling. Thus, chromatin state analysis permits identification of regulators of mesodermal specification, including a conserved role for HOPX in governing primitive hematopoiesis.


Subject(s)
Chromatin/metabolism , Hematopoiesis/genetics , Homeodomain Proteins/genetics , Human Embryonic Stem Cells/metabolism , Mesoderm/metabolism , T-Cell Acute Lymphocytic Leukemia Protein 1/genetics , Transcription, Genetic , Tumor Suppressor Proteins/genetics , Algorithms , CRISPR-Cas Systems , Cell Differentiation , Cell Lineage/genetics , Chromatin/chemistry , Endothelial Cells/cytology , Endothelial Cells/metabolism , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Genes, Reporter , Human Embryonic Stem Cells/cytology , Humans , Mesoderm/cytology , Mesoderm/growth & development , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Signal Transduction , T-Cell Acute Lymphocytic Leukemia Protein 1/metabolism , Tumor Suppressor Proteins/deficiency , beta Catenin/genetics , beta Catenin/metabolism
18.
Stem Cell Reports ; 8(2): 278-289, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28065644

ABSTRACT

We hypothesized that the neonatal rat heart would bring transplanted human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) to maturity as it grows to adult size. In neonatal rat heart, engrafted hiPSC derivatives developed partially matured myofibrils after 3 months, with increasing cell size and sarcomere length. There was no difference between grafts from hiPSC-CMs or hiPSC-derived cardiac progenitors (hiPSC-CPs) at 3 months, nor was maturation influenced by infarction. Interestingly, the infarcted adult heart induced greater human cardiomyocyte hypertrophy and induction of cardiac troponin I expression than the neonatal heart. Although human cardiomyocytes at all time points were significantly smaller than the host rat cardiomyocytes, transplanted neonatal rat cardiomyocytes reached adult size and structure by 3 months. Thus, the adult rat heart induces faster maturation than the neonatal heart, and human cardiomyocytes mature more slowly than rat cardiomyocytes. The slower maturation of human cardiomyocytes could be related to environmental mismatch or cell-autonomous factors.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Age Factors , Animals , Animals, Newborn , Cell Survival , Disease Models, Animal , Humans , Male , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Rats , Stem Cell Transplantation , Time Factors
19.
Nat Protoc ; 12(1): 15-31, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27906170

ABSTRACT

Human pluripotent stem cells (hPSCs) provide a valuable model for the study of human development and a means to generate a scalable source of cells for therapeutic applications. This protocol specifies cell fate efficiently into cardiac and endothelial lineages from hPSCs. The protocol takes 2 weeks to complete and requires experience in hPSC culture and differentiation techniques. Building on lessons taken from early development, this monolayer-directed differentiation protocol uses different concentrations of activin A and bone morphogenetic protein 4 (BMP4) to polarize cells into mesodermal subtypes that reflect mid-primitive-streak cardiogenic mesoderm and posterior-primitive-streak hemogenic mesoderm. This differentiation platform provides a basis for generating distinct cardiovascular progenitor populations that enable the derivation of cardiomyocytes and functionally distinct endothelial cell (EC) subtypes from cardiogenic versus hemogenic mesoderm with high efficiency without cell sorting. ECs derived from cardiogenic and hemogenic mesoderm can be matured into >90% CD31+/VE-cadherin+ definitive ECs. To test the functionality of ECs at different stages of differentiation, we provide methods for assaying the blood-forming potential and de novo lumen-forming activity of ECs. To our knowledge, this is the first protocol that provides a common platform for directed differentiation of cardiomyocytes and endothelial subtypes from hPSCs. This protocol yields endothelial differentiation efficiencies exceeding those of previously published protocols. Derivation of these cell types is a critical step toward understanding the basis of disease and generating cells with therapeutic potential.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Endothelial Cells/cytology , Mesoderm/cytology , Myocardium/cytology , Pluripotent Stem Cells/cytology , Cell Line , Cell Lineage , Humans
20.
Circulation ; 134(20): 1557-1567, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27737958

ABSTRACT

BACKGROUND: Tissue engineering enables the generation of functional human cardiac tissue with cells derived in vitro in combination with biocompatible materials. Human-induced pluripotent stem cell-derived cardiomyocytes provide a cell source for cardiac tissue engineering; however, their immaturity limits their potential applications. Here we sought to study the effect of mechanical conditioning and electric pacing on the maturation of human-induced pluripotent stem cell-derived cardiac tissues. METHODS: Cardiomyocytes derived from human-induced pluripotent stem cells were used to generate collagen-based bioengineered human cardiac tissue. Engineered tissue constructs were subjected to different mechanical stress and electric pacing conditions. RESULTS: The engineered human myocardium exhibits Frank-Starling-type force-length relationships. After 2 weeks of static stress conditioning, the engineered myocardium demonstrated increases in contractility (0.63±0.10 mN/mm2 vs 0.055±0.009 mN/mm2 for no stress), tensile stiffness, construct alignment, and cell size. Stress conditioning also increased SERCA2 (Sarco/Endoplasmic Reticulum Calcium ATPase 2) expression, which correlated with a less negative force-frequency relationship. When electric pacing was combined with static stress conditioning, the tissues showed an additional increase in force production (1.34±0.19 mN/mm2), with no change in construct alignment or cell size, suggesting maturation of excitation-contraction coupling. Supporting this notion, we found expression of RYR2 (Ryanodine Receptor 2) and SERCA2 further increased by combined static stress and electric stimulation. CONCLUSIONS: These studies demonstrate that electric pacing and mechanical stimulation promote maturation of the structural, mechanical, and force generation properties of human-induced pluripotent stem cell-derived cardiac tissues.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Myocardium/metabolism , Myocytes, Cardiac/physiology , Tissue Engineering/methods , Animals , Humans , Stress, Mechanical
SELECTION OF CITATIONS
SEARCH DETAIL