Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Basic Res Cardiol ; 119(2): 309-327, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38305903

ABSTRACT

Statins are effective drugs in reducing cardiovascular morbidity and mortality by inhibiting cholesterol synthesis. These effects are primarily beneficial for the patient's vascular system. A significant number of statin users suffer from muscle complaints probably due to mitochondrial dysfunction, a mechanism that has recently been elucidated. This has raised our interest in exploring the effects of statins on cardiac muscle cells in an era where the elderly and patients with poorer functioning hearts and less metabolic spare capacity start dominating our patient population. Here, we investigated the effects of statins on human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-derived CMs). hiPSC-derived CMs were exposed to simvastatin, atorvastatin, rosuvastatin, and cerivastatin at increasing concentrations. Metabolic assays and fluorescent microscopy were employed to evaluate cellular viability, metabolic capacity, respiration, intracellular acidity, and mitochondrial membrane potential and morphology. Over a concentration range of 0.3-100 µM, simvastatin lactone and atorvastatin acid showed a significant reduction in cellular viability by 42-64%. Simvastatin lactone was the most potent inhibitor of basal and maximal respiration by 56% and 73%, respectively, whereas simvastatin acid and cerivastatin acid only reduced maximal respiration by 50% and 42%, respectively. Simvastatin acid and lactone and atorvastatin acid significantly decreased mitochondrial membrane potential by 20%, 6% and 3%, respectively. The more hydrophilic atorvastatin acid did not seem to affect cardiomyocyte metabolism. This calls for further research on the translatability to the clinical setting, in which a more conscientious approach to statin prescribing might be considered, especially regarding the current shift in population toward older patients with poor cardiac function.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Induced Pluripotent Stem Cells , Simvastatin/analogs & derivatives , Humans , Aged , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Myocytes, Cardiac/metabolism , Atorvastatin/pharmacology , Simvastatin/pharmacology , Mitochondria/metabolism , Lactones/metabolism , Lactones/pharmacology , Hydrogen-Ion Concentration
2.
Development ; 150(15)2023 08 01.
Article in English | MEDLINE | ID: mdl-37560977

ABSTRACT

Developmental research has attempted to untangle the exact signals that control heart growth and size, with knockout studies in mice identifying pivotal roles for Wnt and Hippo signaling during embryonic and fetal heart growth. Despite this improved understanding, no clinically relevant therapies are yet available to compensate for the loss of functional adult myocardium and the absence of mature cardiomyocyte renewal that underlies cardiomyopathies of multiple origins. It remains of great interest to understand which mechanisms are responsible for the decline in proliferation in adult hearts and to elucidate new strategies for the stimulation of cardiac regeneration. Multiple signaling pathways have been identified that regulate the proliferation of cardiomyocytes in the embryonic heart and appear to be upregulated in postnatal injured hearts. In this Review, we highlight the interaction of signaling pathways in heart development and discuss how this knowledge has been translated into current technologies for cardiomyocyte production.


Subject(s)
Cues , Myocytes, Cardiac , Mice , Animals , Myocytes, Cardiac/metabolism , Heart , Myocardium , Signal Transduction , Hippo Signaling Pathway , Cell Proliferation
3.
J Vis Exp ; (193)2023 03 10.
Article in English | MEDLINE | ID: mdl-36971448

ABSTRACT

Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are of paramount importance for human cardiac disease modeling and therapeutics. We recently published a cost-effective strategy for the massive expansion of hiPSC-CMs in two dimensions (2D). Two major limitations are cell immaturity and a lack of three-dimensional (3D) arrangement and scalability in high-throughput screening (HTS) platforms. To overcome these limitations, the expanded cardiomyocytes form an ideal cell source for the generation of 3D cardiac cell culture and tissue engineering techniques. The latter holds great potential in the cardiovascular field, providing more advanced and physiologically relevant HTS. Here, we describe an HTS-compatible workflow with easy scalability for the generation, maintenance, and optical analysis of cardiac spheroids (CSs) in a 96-well-format. These small CSs are essential to fill the gap present in current in vitro disease models and/or generation for 3D tissue engineering platforms. The CSs present a highly structured morphology, size, and cellular composition. Furthermore, hiPSC-CMs cultured as CSs display increased maturation and several functional features of the human heart, such as spontaneous calcium handling and contractile activity. By automatization of the complete workflow, from the generation of CSs to functional analysis, we increase intra- and inter-batch reproducibility as demonstrated by high-throughput (HT) imaging and calcium handling analysis. The described protocol allows modeling of cardiac diseases and assessing drug/therapeutic effects at the single-cell level within a complex 3D cell environment in a fully automated HTS workflow. In addition, the study describes a straightforward procedure for long-term preservation and biobanking of whole-spheroids, thereby providing researchers the opportunity to create next-generation functional tissue storage. HTS combined with long-term storage will substantially contribute to translational research in a wide range of areas, including drug discovery and testing, regenerative medicine, and the development of personalized therapies.


Subject(s)
Heart Diseases , Induced Pluripotent Stem Cells , Humans , High-Throughput Screening Assays , Calcium/pharmacology , Biological Specimen Banks , Reproducibility of Results , Myocytes, Cardiac , Cell Differentiation/physiology
4.
Ned Tijdschr Geneeskd ; 1672023 03 16.
Article in Dutch | MEDLINE | ID: mdl-36928812

ABSTRACT

BACKGROUND: Toxic inhalations form a rare cause of poisoning in the Netherlands. The initial symptoms of toxic inhalations may appear similar to acute viral infections. In the maritime sector aluminum or zinc phosphide is used to overcome rodent infestations during transportation. CASE DESCRIPTION: Here we discuss two patients intoxicated with gaseous phosphide used as fumigant in the transport of grains. The exposure to phosphide gas resulted in respiratory and gastrointestinal tract symptoms. Upon admission one of the patients deteriorated resulting in respiratory insufficiency, multi-organ failure and cardiogenic shock. CONCLUSION: Phosphide gas poisoning forms a rare cause for transient acute heart and multiorgan failure largely due to mitochondria dysfunction. In the case of unexplained incapacitation of multiple patients and/or pets toxic inhalations should differentially diagnostically be considered.


Subject(s)
Gastrointestinal Diseases , Phosphines , Poisoning , Humans , Multiple Organ Failure/chemically induced , Phosphines/poisoning , Shock, Cardiogenic/chemically induced
5.
J Clin Invest ; 132(20)2022 10 17.
Article in English | MEDLINE | ID: mdl-35951416

ABSTRACT

Accidental injury to the cardiac conduction system (CCS), a network of specialized cells embedded within the heart and indistinguishable from the surrounding heart muscle tissue, is a major complication in cardiac surgeries. Here, we addressed this unmet need by engineering targeted antibody-dye conjugates directed against the CCS, allowing for the visualization of the CCS in vivo following a single intravenous injection in mice. These optical imaging tools showed high sensitivity, specificity, and resolution, with no adverse effects on CCS function. Further, with the goal of creating a viable prototype for human use, we generated a fully human monoclonal Fab that similarly targets the CCS with high specificity. We demonstrate that, when conjugated to an alternative cargo, this Fab can also be used to modulate CCS biology in vivo, providing a proof of principle for targeted cardiac therapeutics. Finally, in performing differential gene expression analyses of the entire murine CCS at single-cell resolution, we uncovered and validated a suite of additional cell surface markers that can be used to molecularly target the distinct subcomponents of the CCS, each prone to distinct life-threatening arrhythmias. These findings lay the foundation for translational approaches targeting the CCS for visualization and therapy in cardiothoracic surgery, cardiac imaging, and arrhythmia management.


Subject(s)
Heart Conduction System , Molecular Targeted Therapy , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Heart/physiology , Heart Conduction System/metabolism , Humans , Mice , Myocardium
6.
Stem Cells Transl Med ; 11(10): 1040-1051, 2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36018047

ABSTRACT

The development of new cardioprotective approaches using in vivo models of ischemic heart disease remains challenging as differences in cardiac physiology, phenotype, and disease progression between humans and animals influence model validity and prognostic value. Furthermore, economical and ethical considerations have to be taken into account, especially when using large animal models with relevance for conducting preclinical studies. The development of human-induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) has opened new opportunities for in vitro studies on cardioprotective compounds. However, the immature cellular phenotype of iPSC-CMs remains a roadblock for disease modeling. Here, we show that metabolic maturation renders the susceptibility of iPSC-CMs to hypoxia further toward a clinically representative phenotype. iPSC-CMs cultured in a conventional medium did not show significant cell death after exposure to hypoxia. In contrast, metabolically matured (MM) iPSC-CMs showed inhibited mitochondrial respiration after exposure to hypoxia and increased cell death upon increased durations of hypoxia. Furthermore, we confirmed the applicability of MM iPSC-CMs for in vitro studies of hypoxic damage by validating the known cardioprotective effect of necroptosis inhibitor necrostatin-1. Our results provide important steps to improving and developing valid and predictive human in vitro models of ischemic heart disease.


Subject(s)
Induced Pluripotent Stem Cells , Myocardial Ischemia , Animals , Humans , Myocytes, Cardiac/metabolism , Cell Differentiation , Hypoxia/metabolism
7.
J Cardiovasc Dev Dis ; 9(2)2022 Jan 27.
Article in English | MEDLINE | ID: mdl-35200697

ABSTRACT

Contractility of the adult heart relates to the architectural degree of sarcomeres in individual cardiomyocytes (CMs) and appears to be inversely correlated with the ability to regenerate. In this study we utilized multiple imaging techniques to follow the sequence of sarcomere disassembly during mitosis resulting in cellular or nuclear division in a source of proliferating human pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). We observed that both mono- and binuclear hiPSC-CMs give rise to mononuclear daughter cells or binuclear progeny. Within this source of highly proliferative hiPSC-CMs, treated with the CHIR99021 small molecule, we found that Wnt and Hippo signaling was more present when compared to metabolic matured non-proliferative hiPSC-CMs and adult human heart tissue. Furthermore, we found that CHIR99021 increased the efficiency of non-viral vector incorporation in high-proliferative hiPSC-CMs, in which fluorescent transgene expression became present after the chromosomal segregation (M phase). This study provides a tool for gene manipulation studies in hiPSC-CMs and engineered cardiac tissue. Moreover, our data illustrate that there is a complex biology behind the cellular and nuclear division of mono- and binuclear CMs, with a shared-phenomenon of sarcomere disassembly during mitosis.

8.
Sci Rep ; 11(1): 3026, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33542270

ABSTRACT

Generating cardiomyocytes (CMs) from human induced pluripotent stem cells (hiPSCs) has represented a significant advance in our ability to model cardiac disease. Current differentiation protocols, however, have limited use due to their production of heterogenous cell populations, primarily consisting of ventricular-like CMs. Here we describe the creation of two chamber-specific reporter hiPSC lines by site-directed genomic integration using CRISPR-Cas9 technology. In the MYL2-tdTomato reporter, the red fluorescent tdTomato was inserted upstream of the 3' untranslated region of the Myosin Light Chain 2 (MYL2) gene in order faithfully label hiPSC-derived ventricular-like CMs while avoiding disruption of endogenous gene expression. Similarly, in the SLN-CFP reporter, Cyan Fluorescent Protein (CFP) was integrated downstream of the coding region of the atrial-specific gene, Sarcolipin (SLN). Purification of tdTomato+ and CFP+ CMs using flow cytometry coupled with transcriptional and functional characterization validated these genetic tools for their use in the isolation of bona fide ventricular-like and atrial-like CMs, respectively. Finally, we successfully generated a double reporter system allowing for the isolation of both ventricular and atrial CM subtypes within a single hiPSC line. These tools provide a platform for chamber-specific hiPSC-derived CM purification and analysis in the context of atrial- or ventricular-specific disease and therapeutic opportunities.


Subject(s)
Cell Differentiation/genetics , Heart Atria/growth & development , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , CRISPR-Cas Systems/genetics , Cardiac Myosins/genetics , Green Fluorescent Proteins , Heart Atria/cytology , Heart Atria/metabolism , Heart Ventricles/cytology , Heart Ventricles/growth & development , Heart Ventricles/metabolism , Humans , Induced Pluripotent Stem Cells/pathology , Myocytes, Cardiac/cytology , Myosin Light Chains/genetics
9.
Eur Heart J Case Rep ; 5(2): ytaa525, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33569525

ABSTRACT

BACKGROUND: COVID-19 (severe acute respiratory syndrome coronavirus 2) infected patients have increased risk for thrombotic events, which initially may have been under recognized. The existence of cardiovascular emboli can be directly life threatening when obstructing the blood flow to vital organs such as the brain or other parts of the body. The exact mechanism for this hypercoagulable state in COVID-19 patients yet remains to be elucidated. CASE SUMMARY: A 72-year-old man critically ill with COVID-19 was diagnosed with a free-floating and mural thrombus in the thoracic aorta. Subsequent distal embolization to the limbs led to ischaemia and necrosis of the right foot. Treatment with heparin and anticoagulants reduced thrombus load in the ascending and thoracic aorta. DISCUSSION: One-third of COVID-19 patients show major thrombotic events, mostly pulmonary emboli. The endothelial expression of angiotensin-converting enzyme-2 receptors makes it feasible that in patients with viraemia direct viral-toxicity to the endothelium of also the large arteries results in local thrombus formation. Up to date, prophylactic anticoagulants are recommended in all patients that are hospitalized with COVID-19 infections to prevent venous and arterial thrombotic complications.

10.
STAR Protoc ; 2(1): 100334, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33615277

ABSTRACT

Since the discovery of human induced pluripotent stem cells (hiPSCs), numerous strategies have been established to efficiently derive cardiomyocytes from hiPSCs (hiPSC-CMs). Here, we describe a cost-effective strategy for the subsequent massive expansion (>250-fold) of high-purity hiPSC-CMs relying on two aspects: removal of cell-cell contacts and small-molecule inhibition with CHIR99021. The protocol maintains CM functionality, allows cryopreservation, and the cells can be used in downstream assays such as disease modeling, drug and toxicity screening, and cell therapy. For complete details on the use and execution of this protocol, please refer to Buikema (2020).


Subject(s)
Cell Communication/drug effects , Cryopreservation , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Pyridines/pharmacology , Pyrimidines/pharmacology , Humans
11.
Cell Stem Cell ; 27(5): 840-851.e6, 2020 11 05.
Article in English | MEDLINE | ID: mdl-32818433

ABSTRACT

Modulation of Wnt signaling has untapped potential in regenerative medicine due to its essential functions in stem cell homeostasis. However, Wnt lipidation and Wnt-Frizzled (Fzd) cross-reactivity have hindered translational Wnt applications. Here, we designed and engineered water-soluble, Fzd subtype-specific "next-generation surrogate" (NGS) Wnts that hetero-dimerize Fzd and Lrp6. NGS Wnt supports long-term expansion of multiple different types of organoids, including kidney, colon, hepatocyte, ovarian, and breast. NGS Wnts are superior to Wnt3a conditioned media in organoid expansion and single-cell organoid outgrowth. Administration of Fzd subtype-specific NGS Wnt in vivo reveals that adult intestinal crypt proliferation can be promoted by agonism of Fzd5 and/or Fzd8 receptors, while a broad spectrum of Fzd receptors can induce liver zonation. Thus, NGS Wnts offer a unified organoid expansion protocol and a laboratory "tool kit" for dissecting the functions of Fzd subtypes in stem cell biology.


Subject(s)
Frizzled Receptors , Organoids , Hepatocytes , Stem Cells , Wnt Signaling Pathway
12.
Cell Stem Cell ; 27(1): 50-63.e5, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32619518

ABSTRACT

Modulating signaling pathways including Wnt and Hippo can induce cardiomyocyte proliferation in vivo. Applying these signaling modulators to human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in vitro can expand CMs modestly (<5-fold). Here, we demonstrate massive expansion of hiPSC-CMs in vitro (i.e., 100- to 250-fold) by glycogen synthase kinase-3ß (GSK-3ß) inhibition using CHIR99021 and concurrent removal of cell-cell contact. We show that GSK-3ß inhibition suppresses CM maturation, while contact removal prevents CMs from cell cycle exit. Remarkably, contact removal enabled 10 to 25 times greater expansion beyond GSK-3ß inhibition alone. Mechanistically, persistent CM proliferation required both LEF/TCF activity and AKT phosphorylation but was independent from yes-associated protein (YAP) signaling. Engineered heart tissues from expanded hiPSC-CMs showed comparable contractility to those from unexpanded hiPSC-CMs, demonstrating uncompromised cellular functionality after expansion. In summary, we uncovered a molecular interplay that enables massive hiPSC-CM expansion for large-scale drug screening and tissue engineering applications.


Subject(s)
Induced Pluripotent Stem Cells , Cell Differentiation , Cells, Cultured , Glycogen Synthase Kinase 3 beta , Humans , Myocytes, Cardiac
14.
iScience ; 23(3): 100959, 2020 Mar 27.
Article in English | MEDLINE | ID: mdl-32179481

ABSTRACT

Defining the pathways that control cardiac development facilitates understanding the pathogenesis of congenital heart disease. Herein, we identify enrichment of a Cullin5 Ub ligase key subunit, Asb2, in myocardial progenitors and differentiated cardiomyocytes. Using two conditional murine knockouts, Nkx+/Cre.Asb2fl/fl and AHF-Cre.Asb2fl/fl, and tissue clarifying technique, we reveal Asb2 requirement for embryonic survival and complete heart looping. Deletion of Asb2 results in upregulation of its target Filamin A (Flna), and concurrent Flna deletion partially rescues embryonic lethality. Conditional AHF-Cre.Asb2 knockouts harboring one Flna allele have double outlet right ventricle (DORV), which is rescued by biallelic Flna excision. Transcriptomic and immunofluorescence analyses identify Tgfß/Smad as downstream targets of Asb2/Flna. Finally, using CRISPR/Cas9 genome editing, we demonstrate Asb2 requirement for human cardiomyocyte differentiation suggesting a conserved mechanism between mice and humans. Collectively, our study provides deeper mechanistic understanding of the role of the ubiquitin proteasome system in cardiac development and suggests a previously unidentified murine model for DORV.

15.
Circ Res ; 125(4): 379-397, 2019 08 02.
Article in English | MEDLINE | ID: mdl-31284824

ABSTRACT

RATIONALE: The cardiac conduction system (CCS) consists of distinct components including the sinoatrial node, atrioventricular node, His bundle, bundle branches, and Purkinje fibers. Despite an essential role for the CCS in heart development and function, the CCS has remained challenging to interrogate because of inherent obstacles including small cell numbers, large cell-type heterogeneity, complex anatomy, and difficulty in isolation. Single-cell RNA-sequencing allows for genome-wide analysis of gene expression at single-cell resolution. OBJECTIVE: Assess the transcriptional landscape of the entire CCS at single-cell resolution by single-cell RNA-sequencing within the developing mouse heart. METHODS AND RESULTS: Wild-type, embryonic day 16.5 mouse hearts (n=6 per zone) were harvested and 3 zones of microdissection were isolated, including: Zone I-sinoatrial node region; Zone II-atrioventricular node/His region; and Zone III-bundle branch/Purkinje fiber region. Tissue was digested into single-cell suspensions, cells isolated, mRNA reverse transcribed, and barcoded before high-throughput sequencing and bioinformatics analyses. Single-cell RNA-sequencing was performed on over 22 000 cells, and all major cell types of the murine heart were successfully captured including bona fide clusters of cells consistent with each major component of the CCS. Unsupervised weighted gene coexpression network analysis led to the discovery of a host of novel CCS genes, a subset of which were validated using fluorescent in situ hybridization as well as whole-mount immunolabeling with volume imaging (iDISCO+) in 3 dimensions on intact mouse hearts. Further, subcluster analysis unveiled isolation of distinct CCS cell subtypes, including the clinically relevant but poorly characterized transitional cells that bridge the CCS and surrounding myocardium. CONCLUSIONS: Our study represents the first comprehensive assessment of the transcriptional profiles from the entire CCS at single-cell resolution and provides a characterization in the context of development and disease.


Subject(s)
Heart Conduction System/metabolism , Transcriptome , Animals , Heart Conduction System/cytology , Heart Conduction System/embryology , Mice , RNA-Seq , Single-Cell Analysis
16.
Development ; 146(12)2019 06 14.
Article in English | MEDLINE | ID: mdl-31142541

ABSTRACT

The heart is a complex organ composed of multiple cell and tissue types. Cardiac cells from different regions of the growing embryonic heart exhibit distinct patterns of gene expression, which are thought to contribute to heart development and morphogenesis. Single cell RNA sequencing allows genome-wide analysis of gene expression at the single cell level. Here, we have analyzed cardiac cells derived from early stage developing hearts by single cell RNA-seq and identified cell cycle gene expression as a major determinant of transcriptional variation. Within cell cycle stage-matched CMs from a given heart chamber, we found that CMs in the G2/M phase downregulated sarcomeric and cytoskeletal markers. We also identified cell location-specific signaling molecules that may influence the proliferation of other nearby cell types. Our data highlight how variations in cell cycle activity selectively promote cardiac chamber growth during development, reveal profound chamber-specific cell cycle-linked transcriptional shifts, and open the way to deeper understanding of pathogenesis of congenital heart disease.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Developmental , Heart/embryology , Single-Cell Analysis/methods , Transcription, Genetic , Animals , Cell Cycle , Cluster Analysis , Computational Biology , Cytoskeleton/metabolism , Genomics , Mice , Morphogenesis , Myocardium/metabolism , Myocytes, Cardiac/cytology , RNA/metabolism , Sarcomeres/metabolism , Sequence Analysis, RNA , Signal Transduction
17.
Dev Cell ; 48(6): 741-742, 2019 03 25.
Article in English | MEDLINE | ID: mdl-30913402

ABSTRACT

The adult mammalian heart, unlike in some amphibian and fish species, is generally considered a post-mitotic organ. In this issue of Developmental Cell, Monroe et al. (2019) show that the expression of constitutively active YAP induces a remarkable degree of proliferation in preexisting adult cardiomyocytes by globally altering chromatin accessibility.


Subject(s)
Chromatin , Signal Transduction , Animals , Cell Proliferation , Myocytes, Cardiac
18.
Circ Res ; 123(4): 443-450, 2018 08 03.
Article in English | MEDLINE | ID: mdl-29986945

ABSTRACT

RATIONALE: Human-induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) have risen as a useful tool in cardiovascular research, offering a wide gamut of translational and clinical applications. However, inefficiency of the currently available iPSC-EC differentiation protocol and underlying heterogeneity of derived iPSC-ECs remain as major limitations of iPSC-EC technology. OBJECTIVE: Here, we performed droplet-based single-cell RNA sequencing (scRNA-seq) of the human iPSCs after iPSC-EC differentiation. Droplet-based scRNA-seq enables analysis of thousands of cells in parallel, allowing comprehensive analysis of transcriptional heterogeneity. METHODS AND RESULTS: Bona fide iPSC-EC cluster was identified by scRNA-seq, which expressed high levels of endothelial-specific genes. iPSC-ECs, sorted by CD144 antibody-conjugated magnetic sorting, exhibited standard endothelial morphology and function including tube formation, response to inflammatory signals, and production of NO. Nonendothelial cell populations resulting from the differentiation protocol were identified, which included immature cardiomyocytes, hepatic-like cells, and vascular smooth muscle cells. Furthermore, scRNA-seq analysis of purified iPSC-ECs revealed transcriptional heterogeneity with 4 major subpopulations, marked by robust enrichment of CLDN5, APLNR, GJA5, and ESM1 genes, respectively. CONCLUSIONS: Massively parallel, droplet-based scRNA-seq allowed meticulous analysis of thousands of human iPSCs subjected to iPSC-EC differentiation. Results showed inefficiency of the differentiation technique, which can be improved with further studies based on identification of molecular signatures that inhibit expansion of nonendothelial cell types. Subtypes of bona fide human iPSC-ECs were also identified, allowing us to sort for iPSC-ECs with specific biological function and identity.


Subject(s)
Endothelial Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Transcriptome , Apelin Receptors/genetics , Apelin Receptors/metabolism , Cell Differentiation , Cells, Cultured , Claudin-5/genetics , Claudin-5/metabolism , Connexins/genetics , Connexins/metabolism , Endothelial Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Proteoglycans/genetics , Proteoglycans/metabolism , Single-Cell Analysis , Gap Junction alpha-5 Protein
19.
Sci Rep ; 8(1): 6618, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29700394

ABSTRACT

Bioactive lipids such as sphingosine-1-phosphate (S1P) and lysophosphatidic acid (LPA) regulate diverse processes including cell proliferation, differentiation, and migration. However, their roles in cardiac differentiation and cardiomyocyte proliferation have not been explored. Using a 96-well differentiation platform for generating human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) we found that S1P and LPA can independently enhance cardiomyocyte generation when administered at an early stage of differentiation. We showed that the combined S1P and LPA treatment of undifferentiated hiPSCs resulted in increased nuclear accumulation of ß-catenin, the canonical Wnt signaling pathway mediator, and synergized with CHIR99021, a glycogen synthase kinase 3 beta inhibitor, to enhance mesodermal induction and subsequent cardiac differentiation. At later stages of cardiac differentiation, the addition of S1P and LPA resulted in cell cycle initiation in hiPSC-CMs, an effect mediated through increased ERK signaling. Although the addition of S1P and LPA alone was insufficient to induce cell division, it was able to enhance ß-catenin-mediated hiPSC-CM proliferation. In summary, we demonstrated a developmental stage-specific effect of bioactive lipids to enhance hiPSC-CM differentiation and proliferation via modulating the effect of canonical Wnt/ß-catenin and ERK signaling. These findings may improve hiPSC-CM generation for cardiac disease modeling, precision medicine, and regenerative therapies.


Subject(s)
Cell Differentiation/drug effects , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Lipids/pharmacology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Biomarkers , Cell Proliferation/drug effects , Cells, Cultured , Gene Expression Profiling , Humans , Induced Pluripotent Stem Cells/metabolism , MAP Kinase Signaling System/drug effects , Mesoderm/cytology , Mesoderm/drug effects , Models, Biological , Myocytes, Cardiac/metabolism , Wnt Signaling Pathway/drug effects
20.
Appl Phys Rev ; 5(4): 041106, 2018 Dec.
Article in English | MEDLINE | ID: mdl-32550960

ABSTRACT

Three-dimensional (3D) cardiac tissue bioprinting occupies a critical crossroads position between the fields of materials engineering, cardiovascular biology, 3D printing, and rational organ replacement design. This complex area of research therefore requires expertise from all those disciplines and it poses some unique considerations that must be accounted for. One of the chief hurdles is that there is a relatively limited systematic organization of the physical and chemical characteristics of bioinks that would make them applicable to cardiac bioprinting. This is of great significance, as heart tissue is functionally complex and the in vivo extracellular niche is under stringent controls with little room for variability before a cardiomyopathy manifests. This review explores the critical parameters that are necessary for biologically relevant bioinks to successfully be leveraged for functional cardiac tissue engineering, which can have applications in in vitro heart tissue models, cardiotoxicity studies, and implantable constructs that can be used to treat a range of cardiomyopathies, or in regenerative medicine.

SELECTION OF CITATIONS
SEARCH DETAIL
...