Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Cell ; 176(4): 913-927.e18, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30686581

ABSTRACT

Tissue engineering using cardiomyocytes derived from human pluripotent stem cells holds a promise to revolutionize drug discovery, but only if limitations related to cardiac chamber specification and platform versatility can be overcome. We describe here a scalable tissue-cultivation platform that is cell source agnostic and enables drug testing under electrical pacing. The plastic platform enabled on-line noninvasive recording of passive tension, active force, contractile dynamics, and Ca2+ transients, as well as endpoint assessments of action potentials and conduction velocity. By combining directed cell differentiation with electrical field conditioning, we engineered electrophysiologically distinct atrial and ventricular tissues with chamber-specific drug responses and gene expression. We report, for the first time, engineering of heteropolar cardiac tissues containing distinct atrial and ventricular ends, and we demonstrate their spatially confined responses to serotonin and ranolazine. Uniquely, electrical conditioning for up to 8 months enabled modeling of polygenic left ventricular hypertrophy starting from patient cells.


Subject(s)
Myocytes, Cardiac/cytology , Tissue Culture Techniques/instrumentation , Tissue Engineering/methods , Action Potentials , Cell Differentiation , Cells, Cultured , Electrophysiological Phenomena , Humans , Induced Pluripotent Stem Cells/cytology , Models, Biological , Myocardium/cytology , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/cytology , Tissue Culture Techniques/methods
2.
Cell ; 151(1): 206-20, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22981692

ABSTRACT

Heart development is exquisitely sensitive to the precise temporal regulation of thousands of genes that govern developmental decisions during differentiation. However, we currently lack a detailed understanding of how chromatin and gene expression patterns are coordinated during developmental transitions in the cardiac lineage. Here, we interrogated the transcriptome and several histone modifications across the genome during defined stages of cardiac differentiation. We find distinct chromatin patterns that are coordinated with stage-specific expression of functionally related genes, including many human disease-associated genes. Moreover, we discover a novel preactivation chromatin pattern at the promoters of genes associated with heart development and cardiac function. We further identify stage-specific distal enhancer elements and find enriched DNA binding motifs within these regions that predict sets of transcription factors that orchestrate cardiac differentiation. Together, these findings form a basis for understanding developmentally regulated chromatin transitions during lineage commitment and the molecular etiology of congenital heart disease.


Subject(s)
Epigenesis, Genetic , Gene Regulatory Networks , Myocardium/cytology , Animals , Cell Differentiation , Chromatin/metabolism , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic , Heart/embryology , Humans , Mice , Transcription Factors/metabolism , Transcriptome
3.
Development ; 140(12): 2597-610, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23715552

ABSTRACT

Osteoarthritis primarily affects the articular cartilage of synovial joints. Cell and/or cartilage replacement is a promising therapy, provided there is access to appropriate tissue and sufficient numbers of articular chondrocytes. Embryonic stem cells (ESCs) represent a potentially unlimited source of chondrocytes and tissues as they can generate a broad spectrum of cell types under appropriate conditions in vitro. Here, we demonstrate that mouse ESC-derived chondrogenic mesoderm arises from a Flk-1(-)/Pdgfrα(+) (F(-)P(+)) population that emerges in a defined temporal pattern following the development of an early cardiogenic F(-)P(+) population. Specification of the late-arising F(-)P(+) population with BMP4 generated a highly enriched population of chondrocytes expressing genes associated with growth plate hypertrophic chondrocytes. By contrast, specification with Gdf5, together with inhibition of hedgehog and BMP signaling pathways, generated a population of non-hypertrophic chondrocytes that displayed properties of articular chondrocytes. The two chondrocyte populations retained their hypertrophic and non-hypertrophic properties when induced to generate spatially organized proteoglycan-rich cartilage-like tissue in vitro. Transplantation of either type of chondrocyte, or tissue generated from them, into immunodeficient recipients resulted in the development of cartilage tissue and bone within an 8-week period. Significant ossification was not observed when the tissue was transplanted into osteoblast-depleted mice or into diffusion chambers that prevent vascularization. Thus, through stage-specific manipulation of appropriate signaling pathways it is possible to efficiently and reproducibly derive hypertrophic and non-hypertrophic chondrocyte populations from mouse ESCs that are able to generate distinct cartilage-like tissue in vitro and maintain a cartilage tissue phenotype within an avascular and/or osteoblast-free niche in vivo.


Subject(s)
Cartilage, Articular/cytology , Chondrocytes/cytology , Chondrogenesis , Embryonic Stem Cells/cytology , Animals , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Cartilage, Articular/metabolism , Cell Differentiation , Cell Lineage , Chondrocytes/metabolism , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/transplantation , Female , Growth Differentiation Factor 5/genetics , Growth Differentiation Factor 5/metabolism , Hypertrophy/metabolism , Immunohistochemistry , Mesoderm/cytology , Mesoderm/metabolism , Mice , Osteoblasts/metabolism , Osteogenesis , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction , Time Factors
4.
Proc Natl Acad Sci U S A ; 110(49): E4698-707, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24255110

ABSTRACT

Access to robust and information-rich human cardiac tissue models would accelerate drug-based strategies for treating heart disease. Despite significant effort, the generation of high-fidelity adult-like human cardiac tissue analogs remains challenging. We used computational modeling of tissue contraction and assembly mechanics in conjunction with microfabricated constraints to guide the design of aligned and functional 3D human pluripotent stem cell (hPSC)-derived cardiac microtissues that we term cardiac microwires (CMWs). Miniaturization of the platform circumvented the need for tissue vascularization and enabled higher-throughput image-based analysis of CMW drug responsiveness. CMW tissue properties could be tuned using electromechanical stimuli and cell composition. Specifically, controlling self-assembly of 3D tissues in aligned collagen, and pacing with point stimulation electrodes, were found to promote cardiac maturation-associated gene expression and in vivo-like electrical signal propagation. Furthermore, screening a range of hPSC-derived cardiac cell ratios identified that 75% NKX2 Homeobox 5 (NKX2-5)+ cardiomyocytes and 25% Cluster of Differentiation 90 OR (CD90)+ nonmyocytes optimized tissue remodeling dynamics and yielded enhanced structural and functional properties. Finally, we demonstrate the utility of the optimized platform in a tachycardic model of arrhythmogenesis, an aspect of cardiac electrophysiology not previously recapitulated in 3D in vitro hPSC-derived cardiac microtissue models. The design criteria identified with our CMW platform should accelerate the development of predictive in vitro assays of human heart tissue function.


Subject(s)
Cellular Microenvironment/physiology , Myocardium/cytology , Pluripotent Stem Cells/cytology , Tissue Engineering/methods , Biomechanical Phenomena , Electric Stimulation , Finite Element Analysis , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/metabolism , Humans , Thy-1 Antigens/metabolism , Transcription Factors/metabolism
5.
Nature ; 453(7194): 524-8, 2008 May 22.
Article in English | MEDLINE | ID: mdl-18432194

ABSTRACT

The functional heart is comprised of distinct mesoderm-derived lineages including cardiomyocytes, endothelial cells and vascular smooth muscle cells. Studies in the mouse embryo and the mouse embryonic stem cell differentiation model have provided evidence indicating that these three lineages develop from a common Flk-1(+) (kinase insert domain protein receptor, also known as Kdr) cardiovascular progenitor that represents one of the earliest stages in mesoderm specification to the cardiovascular lineages. To determine whether a comparable progenitor is present during human cardiogenesis, we analysed the development of the cardiovascular lineages in human embryonic stem cell differentiation cultures. Here we show that after induction with combinations of activin A, bone morphogenetic protein 4 (BMP4), basic fibroblast growth factor (bFGF, also known as FGF2), vascular endothelial growth factor (VEGF, also known as VEGFA) and dickkopf homolog 1 (DKK1) in serum-free media, human embryonic-stem-cell-derived embryoid bodies generate a KDR(low)/C-KIT(CD117)(neg) population that displays cardiac, endothelial and vascular smooth muscle potential in vitro and, after transplantation, in vivo. When plated in monolayer cultures, these KDR(low)/C-KIT(neg) cells differentiate to generate populations consisting of greater than 50% contracting cardiomyocytes. Populations derived from the KDR(low)/C-KIT(neg) fraction give rise to colonies that contain all three lineages when plated in methylcellulose cultures. Results from limiting dilution studies and cell-mixing experiments support the interpretation that these colonies are clones, indicating that they develop from a cardiovascular colony-forming cell. Together, these findings identify a human cardiovascular progenitor that defines one of the earliest stages of human cardiac development.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Myocytes, Cardiac/cytology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Activins/pharmacology , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/pharmacology , Cell Differentiation/drug effects , Cell Line , Cell Lineage/drug effects , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/transplantation , Fibroblast Growth Factor 2/pharmacology , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Proto-Oncogene Proteins c-kit/genetics , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/deficiency , Vascular Endothelial Growth Factor Receptor-2/genetics
6.
Cell Stem Cell ; 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38908380

ABSTRACT

The intricate anatomical structure and high cellular density of the myocardium complicate the bioengineering of perfusable vascular networks within cardiac tissues. In vivo neonatal studies highlight the key role of resident cardiac macrophages in post-injury regeneration and angiogenesis. Here, we integrate human pluripotent stem-cell-derived primitive yolk-sac-like macrophages within vascularized heart-on-chip platforms. Macrophage incorporation profoundly impacted the functionality and perfusability of microvascularized cardiac tissues up to 2 weeks of culture. Macrophages mitigated tissue cytotoxicity and the release of cell-free mitochondrial DNA (mtDNA), while upregulating the secretion of pro-angiogenic, matrix remodeling, and cardioprotective cytokines. Bulk RNA sequencing (RNA-seq) revealed an upregulation of cardiac maturation and angiogenesis genes. Further, single-nuclei RNA sequencing (snRNA-seq) and secretome data suggest that macrophages may prime stromal cells for vascular development by inducing insulin like growth factor binding protein 7 (IGFBP7) and hepatocyte growth factor (HGF) expression. Our results underscore the vital role of primitive macrophages in the long-term vascularization of cardiac tissues, offering insights for therapy and advancing heart-on-a-chip technologies.

7.
Development ; 137(17): 2829-39, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20659975

ABSTRACT

The efficient and reproducible generation of differentiated progenitors from pluripotent stem cells requires the recapitulation of appropriate developmental stages and pathways. Here, we have used the combination of activin A, BMP4 and VEGF under serum-free conditions to induce hematopoietic differentiation from both embryonic and induced pluripotent stem cells, with the aim of modeling the primary sites of embryonic hematopoiesis. We identified two distinct Flk1-positive hematopoietic populations that can be isolated based on temporal patterns of emergence. The earliest arising population displays characteristics of yolk sac hematopoiesis, whereas a late developing Flk1-positive population appears to reflect the para-aortic splanchnopleura hematopoietic program, as it has reduced primitive erythroid capacity and substantially enhanced myeloid and lymphoid potential compared with the earlier wave. These differences between the two populations are accompanied by differences in the expression of Sox17 and Hoxb4, as well as in the cell surface markers AA4.1 and CD41. Together, these findings support the interpretation that the two populations are representative of the early sites of mammalian hematopoiesis.


Subject(s)
Embryonic Stem Cells/cytology , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Activins/administration & dosage , Animals , Bone Morphogenetic Protein 4/administration & dosage , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Gene Expression , HMGB Proteins/genetics , HMGB Proteins/metabolism , Hematopoiesis/drug effects , Hematopoiesis/genetics , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Lymphopoiesis/drug effects , Lymphopoiesis/genetics , Lymphopoiesis/physiology , Membrane Glycoproteins/metabolism , Mice , Models, Biological , Platelet Membrane Glycoprotein IIb/metabolism , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Receptors, Complement/metabolism , Recombinant Proteins/administration & dosage , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor Receptor-2/metabolism
8.
Proc Natl Acad Sci U S A ; 107(8): 3329-34, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-19846783

ABSTRACT

Myocardial infarction resulting in irreversible loss of cardiomyocytes (CMs) remains a leading cause of heart failure. Although cell transplantation has modestly improved cardiac function, major challenges including increasing cell survival, engraftment, and functional integration with host tissue, remain. Embryonic stem cells (ESCs), which can be differentiated into cardiac progenitors (CPs) and CMs, represent a candidate cell source for cardiac cell therapy. However, it is not known what specific cell type or condition is the most appropriate for transplantation. This problem is exasperated by the lack of efficient and predictive strategies to screen the large numbers of parameters that may impact cell transplantation. We used a cardiac tissue model, engineered heart tissue (EHT), and quantitative molecular and electrophysiological analyses, to test transplantation conditions and specific cell populations for their potential to functionally integrate with the host tissue. In this study, we validated our analytical platform using contractile mouse neonatal CMs (nCMs) and noncontractile cardiac fibroblasts (cFBs), and screened for the integration potential of ESC-derived CMs and CPs (ESC-CMs and -CPs). Consistent with previous in vivo studies, cFB injection interfered with electrical signal propagation, whereas injected nCMs improved tissue function. Purified bioreactor-generated ESC-CMs exhibited a diminished capacity for electrophysiological integration; a result correlated with lower (compared with nCMs) connexin 43 expression. ESC-CPs, however, appeared able to appropriately mature and integrate into EHT, enhancing the amplitude of tissue contraction. Our results support the use of EHT as a model system to accelerate development of cardiac cell therapy strategies.


Subject(s)
Heart Failure/surgery , Myoblasts, Cardiac/physiology , Myocardial Contraction , Pluripotent Stem Cells/cytology , Tissue Engineering/methods , Animals , Bioreactors , Cell Differentiation , Connexin 43/biosynthesis , Electrophysiological Phenomena , Fibroblasts/physiology , Heart Failure/etiology , Mice , Mice, Transgenic , Myoblasts, Cardiac/metabolism , Myoblasts, Cardiac/transplantation , Myocardial Infarction/complications , Myocardial Infarction/physiopathology , Myocardial Infarction/surgery , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Rats , Rats, Sprague-Dawley
9.
Blood ; 115(14): 2769-76, 2010 Apr 08.
Article in English | MEDLINE | ID: mdl-20065292

ABSTRACT

The directed differentiation of human pluripotent stem cells offers the unique opportunity to generate a broad spectrum of human cell types and tissues for transplantation, drug discovery, and studying disease mechanisms. Here, we report the stepwise generation of bone-resorbing osteoclasts from human embryonic and induced pluripotent stem cells. Generation of a primitive streak-like population in embryoid bodies, followed by specification to hematopoiesis and myelopoiesis by vascular endothelial growth factor and hematopoietic cytokines in serum-free media, yielded a precursor population enriched for cells expressing the monocyte-macrophage lineage markers CD14, CD18, CD11b, and CD115. When plated in monolayer culture in the presence of macrophage colony-stimulating factor and receptor activator of nuclear factor-kappaB ligand (RANKL), these precursors formed large, multinucleated osteoclasts that expressed tartrate-resistant acid phosphatase and were capable of resorption. No tartrate-resistant acid phosphatase-positive multinucleated cells or resorption pits were observed in the absence of RANKL. Molecular analyses confirmed the expression of the osteoclast marker genes NFATc1, cathepsin K, and calcitonin receptor in a RANKL-dependent manner, and confocal microscopy demonstrated the coexpression of the alphavbeta3 integrin, cathepsin K and F-actin rings characteristic of active osteoclasts. Generating hematopoietic and osteoclast populations from human embryonic and induced pluripotent stem cells will be invaluable for understanding embryonic bone development and postnatal bone disease.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/metabolism , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Osteoclasts/metabolism , Antigens, Differentiation/biosynthesis , Cell Differentiation/drug effects , Cell Line , Embryonic Stem Cells/cytology , Gene Expression Regulation/drug effects , Hematopoietic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Macrophage Colony-Stimulating Factor/pharmacology , Myelopoiesis/drug effects , Myelopoiesis/physiology , Osteoclasts/cytology , RANK Ligand/pharmacology , Vascular Endothelial Growth Factor A/pharmacology
10.
Stem Cells ; 29(2): 217-28, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21732480

ABSTRACT

Complex cross-talk between endoderm and the microenvironment is an absolute requirement to orchestrate hepatic specification and expansion. In the mouse, the septum transversum and cardiac mesoderm, through secreted bone morphogenetic proteins (BMP) and fibroblast growth factors (FGF), respectively, instruct the adjacent ventral endoderm to become hepatic endoderm. Consecutively, endothelial cells promote expansion of the specified hepatic endoderm. By using a mouse reporter embryonic stem cell line, in which hCD4 and hCD25 were targeted to the Foxa2 and Foxa3 loci, we reconstituted an in vitro culture system in which committed endoderm cells coexpressing hCD4-Foxa2 and hCD25-Foxa3 were isolated and cocultured with endothelial cells in the presence of BMP4 and bFGF. In this culture setting, we provide mechanistic evidence that endothelial cells function not only to promote hepatic endoderm expansion but are also required at an earlier step for hepatic specification, at least in part through regulation of the Wnt and Notch pathways. Activation of Wnt and Notch by chemical or genetic approaches increases endoderm cell numbers but inhibits hepatic specification, and conversely, chemical inhibition of both pathways enhances hepatic specification and reduces proliferation. By using identical coculture conditions, we defined a similar dependence of endoderm harvested from embryos on endothelial cells to support their growth and hepatic specification. Our findings (1) confirm a conserved role of Wnt repression for mouse hepatic specification, (2) uncover a novel role for Notch repression in the hepatic fate decision, and (3) demonstrate that repression of Wnt and Notch signaling in hepatic endoderm is controlled by the endothelial cell niche.


Subject(s)
Embryonic Stem Cells/metabolism , Endothelial Cells/metabolism , Hepatocytes/metabolism , Wnt Signaling Pathway , Animals , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/pharmacology , CD4 Antigens/biosynthesis , CD4 Antigens/genetics , Cell Differentiation/drug effects , Cell Line , Embryonic Stem Cells/cytology , Endoderm/cytology , Endoderm/metabolism , Fibroblast Growth Factors/pharmacology , Hepatocyte Nuclear Factor 3-beta/biosynthesis , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-gamma/biosynthesis , Hepatocyte Nuclear Factor 3-gamma/genetics , Hepatocytes/cytology , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-2 Receptor alpha Subunit/genetics , Mice , Receptors, Notch/metabolism , Wnt Proteins/metabolism
11.
Proc Natl Acad Sci U S A ; 106(18): 7571-6, 2009 May 05.
Article in English | MEDLINE | ID: mdl-19372372

ABSTRACT

A variety of viruses establish latency by integrating their genome into the host genome. The integration event generally occurs in a nonspecific manner, precluding the prediction of functional consequences from resulting disruptions of affected host genes. The nonpathogenic adeno-associated virus (AAV) is unique in its ability to stably integrate in a site-specific manner into the human MBS85 gene. To gain a better understanding of the integration mechanism and the consequences of MBS85 disruption, we analyzed the molecular structure of AAV integrants in various latently infected human cell lines. Our study led to the observation that AAV integration causes an extensive but partial duplication of the target gene. Intriguingly, the molecular organization of the integrant leaves the possibility that a functional copy of the disrupted target gene could potentially be preserved despite the resulting rearrangements. A latently infected, Mbs85-targeted mouse ES cell line was generated to study the functional consequences of the observed duplication-based integration mechanism. AAV-modified ES cell lines continued to self-renew, maintained their multilineage differentiation potential and contributed successfully to mouse development when injected into blastocysts. Thus, our study reveals a viral strategy for targeted genome addition with the apparent absence of functional consequences.


Subject(s)
Dependovirus/genetics , Gene Targeting/methods , Proviruses/genetics , Virus Integration , Virus Latency , Animals , Cell Line , Embryonic Stem Cells/metabolism , Gene Expression , Humans , Mice , Protein Phosphatase 1/genetics
12.
Cell Rep ; 39(1): 110621, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35385743

ABSTRACT

Liver sinusoidal endothelial cells (LSECs) form the predominant microvasculature in the liver where they carry out many functions including the secretion of coagulation factor VIII (FVIII). To investigate the early origins of this lineage, we develop an efficient and scalable protocol to produce human pluripotent stem cell (hPSC)-derived LSEC progenitors characterized as venous endothelial cells (VECs) from different mesoderm subpopulations. Using a sensitive and quantitative vascular competitive transplantation assay, we demonstrate that VECs generated from BMP4 and activin A-induced KDR+CD235a/b+ mesoderm are 50-fold more efficient at LSEC engraftment than venous cells from BMP4 and WNT-induced KDR+CD235a/b- mesoderm. When transplanted into immunocompromised hemophilia A mice (NSG-HA), these VECs engraft the liver, proliferate, and mature to functional LSECs that secrete bioactive FVIII capable of correcting the bleeding phenotype. Together, these findings highlight the importance of appropriate mesoderm induction for generating hPSC-derived LSECs capable of functioning in a preclinical model of hemophilia A.


Subject(s)
Endothelial Progenitor Cells , Hemophilia A , Pluripotent Stem Cells , Animals , Hemophilia A/genetics , Hemophilia A/therapy , Hepatocytes , Liver , Mice
13.
J Exp Med ; 219(3)2022 03 07.
Article in English | MEDLINE | ID: mdl-34928315

ABSTRACT

In the mouse, the first hematopoietic cells are generated in the yolk sac from the primitive, erythro-myeloid progenitor (EMP) and lymphoid programs that are specified before the emergence of hematopoietic stem cells. While many of the yolk sac-derived populations are transient, specific immune cell progeny seed developing tissues, where they function into adult life. To access the human equivalent of these lineages, we modeled yolk sac hematopoietic development using pluripotent stem cell differentiation. Here, we show that the combination of Activin A, BMP4, and FGF2 induces a population of KDR+CD235a/b+ mesoderm that gives rise to the spectrum of erythroid, myeloid, and T lymphoid lineages characteristic of the mouse yolk sac hematopoietic programs, including the Vδ2+ subset of γ/δ T cells that develops early in the human embryo. Through clonal analyses, we identified a multipotent hematopoietic progenitor with erythroid, myeloid, and T lymphoid potential, suggesting that the yolk sac EMP and lymphoid lineages may develop from a common progenitor.


Subject(s)
Hematopoiesis , Models, Biological , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Yolk Sac/cytology , Animals , Biomarkers , Cell Differentiation/genetics , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Immunophenotyping , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Lymphopoiesis/genetics , Mice , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism
14.
Cell Stem Cell ; 29(9): 1382-1401.e8, 2022 09 01.
Article in English | MEDLINE | ID: mdl-36055193

ABSTRACT

The cardiomyocyte (CM) subtypes in the mammalian heart derive from distinct lineages known as the first heart field (FHF), the anterior second heart field (aSHF), and the posterior second heart field (pSHF) lineages that are specified during gastrulation. We modeled human heart field development from human pluripotent stem cells (hPSCs) by using single-cell RNA-sequencing to delineate lineage specification and progression. Analyses of hPSC-derived and mouse mesoderm transcriptomes enabled the identification of distinct human FHF, aSHF, and pSHF mesoderm subpopulations. Through staged manipulation of signaling pathways identified from transcriptomics, we generated myocyte populations that display molecular characteristics of key CM subtypes. The developmental trajectory of the human cardiac lineages recapitulated that of the mouse, demonstrating conserved cardiovascular programs. These findings establish a comprehensive landscape of human embryonic cardiogenesis that provides access to a broad spectrum of cardiomyocytes for modeling congenital heart diseases and chamber-specific cardiomyopathies as well as for developing new therapies to treat them.


Subject(s)
Pluripotent Stem Cells , Animals , Cell Differentiation , Embryo, Mammalian , Humans , Mammals , Mesoderm , Mice , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/metabolism
15.
Dev Cell ; 11(5): 723-32, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17084363

ABSTRACT

Cell-tracing studies in the mouse indicate that the cardiac lineage arises from a population that expresses the vascular endothelial growth factor receptor 2 (VEGFR2, Flk-1), suggesting that it may develop from a progenitor with vascular potential. Using the embryonic stem (ES) cell differentiation model, we have identified a cardiovascular progenitor based on the temporal expression of the primitive streak (PS) marker brachyury and Flk-1. Comparable progenitors could also be isolated from head-fold stage embryos. When cultured with cytokines known to function during cardiogenesis, individual cardiovascular progenitors generated colonies that displayed cardiomyocyte, endothelial, and vascular smooth muscle (VSM) potential. Isolation and characterization of this previously unidentified population suggests that the mammalian cardiovascular system develops from multipotential progenitors.


Subject(s)
Embryonic Stem Cells/cytology , Endothelial Cells/cytology , Multipotent Stem Cells/cytology , Muscle, Smooth, Vascular/cytology , Myocytes, Cardiac/cytology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cardiovascular System/cytology , Cardiovascular System/embryology , Cell Differentiation , Cell Lineage , Cells, Cultured , Fetal Proteins/metabolism , Mice , T-Box Domain Proteins/metabolism , Transcription Factors/biosynthesis
16.
Cell Stem Cell ; 28(1): 96-111.e7, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33142114

ABSTRACT

The embryonic endocardium is essential for early heart development as it functions to induce trabecular myocardium, the first heart tissue to form, and is the source of the cells that make up the valves and a portion of the coronary vasculature. With this potential, human endocardial cells could provide unique therapeutic opportunities that include engineering biological valves and cell-based therapy strategies to replace coronary vasculature in damaged hearts. To access human endocardial cells, we generated a human pluripotent stem cell (hPSC)-derived endothelial population that displays many characteristics of endocardium, including expression of the cohort of genes that identifies this lineage in vivo, the capacity to induce a trabecular fate in immature cardiomyocytes in vitro, and the ability to undergo an endothelial-to-mesenchymal transition. Analyses of the signaling pathways required for development of the hPSC-derived endocardial cells identified a novel role for BMP10 in the specification of this lineage from cardiovascular mesoderm.


Subject(s)
Endocardium , Pluripotent Stem Cells , Bone Morphogenetic Proteins , Cell Differentiation , Humans , Myocardium , Signal Transduction
17.
Adv Healthc Mater ; 10(8): e2001746, 2021 04.
Article in English | MEDLINE | ID: mdl-33694327

ABSTRACT

Tubular biological structures consisting of extracellular matrix (ECM) proteins and cells are basic functional units of all organs in animals and humans. ECM protein solutions at low concentrations (5-10 milligrams per milliliter) are abundantly used in 3D cell culture. However, their poor "printability" and minute-long gelation time have made the direct extrusion of tubular structures in bioprinting applications challenging. Here, this limitation is overcome and the continuous, template-free conversion of low-concentration collagen, elastin, and fibrinogen solutions into tubular structures of tailored size and radial, circumferential and axial organization is demonstrated. The approach is enabled by a microfabricated printhead for the consistent circumferential distribution of ECM protein solutions and lends itself to scalable manufacture. The attached confinement accommodates minute-long residence times for pH, temperature, light, ionic and enzymatic gelation. Chip hosted ECM tubular structures are amenable to perfusion with aqueous solutions and air, and cyclic stretching. Predictive collapse and reopening in a crossed-tube configuration promote all-ECM valves and pumps. Tissue level function is demonstrated by factors secreted from cells embedded within the tube wall, as well as endothelial or epithelial barriers lining the lumen. The described approaches are anticipated to find applications in ECM-based organ-on-chip and biohybrid structures, hydraulic actuators, and soft machines.


Subject(s)
Bioprinting , Tissue Engineering , Animals , Collagen , Elastin , Extracellular Matrix , Humans
18.
Stem Cells ; 27(9): 2103-13, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19522011

ABSTRACT

The development of functional cell populations such as hepatocytes and pancreatic beta cells from embryonic stem cell (ESC) is dependent on the efficient induction of definitive endoderm early in the differentiation process. To monitor definitive endoderm formation in mouse ESC differentiation cultures in a quantitative fashion, we generated a reporter cell line that expresses human CD25 from the Foxa3 locus and human CD4 from the Foxa2 locus. Induction of these reporter ESCs with high concentrations of activin A led to the development of a CD25-Foxa3+CD4-Foxa2+ population within 4-5 days of culture. Isolation and characterization of this population showed that it consists predominantly of definitive endoderm that is able to undergo hepatic specification under the appropriate conditions. To develop reagents that can be used for studies on endoderm development from unmanipulated ESCs, from induced pluripotent stem cells, and from the mouse embryo, we generated monoclonal antibodies against the CD25-Foxa3+CD4-Foxa2+ population. With this approach, we identified two antibodies that react specifically with endoderm from ESC cultures and from the early embryo. The specificity of these antibodies enables one to quantitatively monitor endoderm development in ESC differentiation cultures, to study endoderm formation in the embryo, and to isolate pure populations of culture- or embryo-derived endodermal cells.


Subject(s)
Antibodies, Monoclonal/immunology , CD4 Antigens/metabolism , Endoderm/immunology , Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 3-beta/metabolism , Hepatocyte Nuclear Factor 3-gamma/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Activins/pharmacology , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/immunology , Embryonic Stem Cells/metabolism , Endoderm/cytology , Flow Cytometry , Humans , Mice
19.
Stem Cell Reports ; 15(3): 587-596, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32763158

ABSTRACT

Current platforms for studying the mechanical properties of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) as single cells do not measure forces directly, require numerous assumptions, and cannot study cell mechanics at different loading conditions. We present a method for directly measuring the active and passive forces generated by single-cell hPSC-CMs at different stretch levels. Utilizing this technique, single hPSC-CMs exhibited positive length-tension relationship and appropriate inotropic, klinotropic, and lusitropic changes in response to pharmacological treatments (isoproterenol and verapamil). The unique potential of the approach for drug testing and disease modeling was exemplified by doxorubicin and omecamtiv mecarbil drug studies revealing their known actions to suppress (doxorubicin) or augment (omecamtiv mecarbil at low dose) cardiomyocyte contractility, respectively. Finally, mechanistic insights were gained regarding the cellular effects of these drugs as doxorubicin treatment led to cellular mechanical alternans and high doses of omecamtiv mecarbil suppressed contractility and worsened the cellular diastolic properties.


Subject(s)
Drug Evaluation, Preclinical , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology , Single-Cell Analysis , Biomechanical Phenomena/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Shape/drug effects , Doxorubicin/pharmacology , Humans , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Urea/analogs & derivatives , Urea/pharmacology
20.
Cell Stem Cell ; 27(2): 254-269.e9, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32640183

ABSTRACT

Liver sinusoidal endothelial cells (LSECs) form a highly specialized microvasculature that plays a critical role in liver function and disease. To better understand this role, we developed a strategy to generate LSECs from human pluripotent stem cells (hPSCs) by first optimizing the specification of arterial and venous angioblasts and derivative endothelial populations. Induction of a LSEC-like fate by hypoxia, cyclic AMP (cAMP) agonism, and transforming growth factor ß (TGF-ß) inhibition revealed that venous endothelial cells responded more rapidly and robustly than the arterial cells to upregulate LSEC markers and functions in vitro. Upon intrahepatic transplantation in neonates, venous angioblasts engrafted the liver and generated mature, fenestrated LSECs with scavenger functions and molecular profiles of primary human LSECs. When transplanted into the liver of adult mice, angioblasts efficiently gave rise to mature LSECs with robust factor VIII (FVIII) production. Humanization of the murine liver with hPSC-derived LSECs provides a tractable system for studying the biology of this key liver cell type.


Subject(s)
Endothelial Cells , Pluripotent Stem Cells , Animals , Hepatocytes , Humans , Liver , Mice , Transforming Growth Factor beta
SELECTION OF CITATIONS
SEARCH DETAIL