Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cell Rep ; 42(4): 112330, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37014753

ABSTRACT

A limitation in the application of pluripotent stem cell-derived cardiomyocytes (PSC-CMs) is the failure of these cells to achieve full functional maturity. The mechanisms by which directed differentiation differs from endogenous development, leading to consequent PSC-CM maturation arrest, remain unclear. Here, we generate a single-cell RNA sequencing (scRNA-seq) reference of mouse in vivo CM maturation with extensive sampling of previously difficult-to-isolate perinatal time periods. We subsequently generate isogenic embryonic stem cells to create an in vitro scRNA-seq reference of PSC-CM-directed differentiation. Through trajectory reconstruction, we identify an endogenous perinatal maturation program that is poorly recapitulated in vitro. By comparison with published human datasets, we identify a network of nine transcription factors (TFs) whose targets are consistently dysregulated in PSC-CMs across species. Notably, these TFs are only partially activated in common ex vivo approaches to engineer PSC-CM maturation. Our study can be leveraged toward improving the clinical viability of PSC-CMs.


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Humans , Animals , Mice , Myocytes, Cardiac , Cell Differentiation , Embryonic Stem Cells , Transcription Factors/genetics
2.
Circulation ; 147(17): 1291-1303, 2023 04 25.
Article in English | MEDLINE | ID: mdl-36970983

ABSTRACT

BACKGROUND: During cardiomyocyte maturation, the centrosome, which functions as a microtubule organizing center in cardiomyocytes, undergoes dramatic structural reorganization where its components reorganize from being localized at the centriole to the nuclear envelope. This developmentally programmed process, referred to as centrosome reduction, has been previously associated with cell cycle exit. However, understanding of how this process influences cardiomyocyte cell biology, and whether its disruption results in human cardiac disease, remains unknown. We studied this phenomenon in an infant with a rare case of infantile dilated cardiomyopathy (iDCM) who presented with left ventricular ejection fraction of 18% and disrupted sarcomere and mitochondria structure. METHODS: We performed an analysis beginning with an infant who presented with a rare case of iDCM. We derived induced pluripotent stem cells from the patient to model iDCM in vitro. We performed whole exome sequencing on the patient and his parents for causal gene analysis. CRISPR/Cas9-mediated gene knockout and correction in vitro were used to confirm whole exome sequencing results. Zebrafish and Drosophila models were used for in vivo validation of the causal gene. Matrigel mattress technology and single-cell RNA sequencing were used to characterize iDCM cardiomyocytes further. RESULTS: Whole exome sequencing and CRISPR/Cas9 gene knockout/correction identified RTTN, the gene encoding the centrosomal protein RTTN (rotatin), as the causal gene underlying the patient's condition, representing the first time a centrosome defect has been implicated in a nonsyndromic dilated cardiomyopathy. Genetic knockdowns in zebrafish and Drosophila confirmed an evolutionarily conserved requirement of RTTN for cardiac structure and function. Single-cell RNA sequencing of iDCM cardiomyocytes showed impaired maturation of iDCM cardiomyocytes, which underlie the observed cardiomyocyte structural and functional deficits. We also observed persistent localization of the centrosome at the centriole, contrasting with expected programmed perinuclear reorganization, which led to subsequent global microtubule network defects. In addition, we identified a small molecule that restored centrosome reorganization and improved the structure and contractility of iDCM cardiomyocytes. CONCLUSIONS: This study is the first to demonstrate a case of human disease caused by a defect in centrosome reduction. We also uncovered a novel role for RTTN in perinatal cardiac development and identified a potential therapeutic strategy for centrosome-related iDCM. Future study aimed at identifying variants in centrosome components may uncover additional contributors to human cardiac disease.


Subject(s)
Cardiomyopathy, Dilated , Female , Pregnancy , Animals , Humans , Cardiomyopathy, Dilated/genetics , Zebrafish , Stroke Volume , Ventricular Function, Left , Centrosome/metabolism , Myocytes, Cardiac
3.
Proc Natl Acad Sci U S A ; 120(4): e2217687120, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36649430

ABSTRACT

The heart develops in a synchronized sequence of proliferation and differentiation of cardiac progenitor cells (CPCs) from two anatomically distinct pools of cells, the first heart field (FHF) and second heart field (SHF). Congenital heart defects arise upon dysregulation of these processes, many of which are restricted to derivatives of the FHF or SHF. Of the conserved set of signaling pathways that regulate development, the Wnt signaling pathway has long been known for its importance in SHF development. The source of such Wnts has remained elusive, though it has been postulated that these Wnts are secreted from ectodermal or endodermal sources. The central question remains unanswered: Where do these Wnts come from? Here, we show that CPCs autoregulate SHF development via Wnt through genetic manipulation of a key Wnt export protein (Wls), scRNA-seq analysis of CPCs, and use of our precardiac organoid system. Through this, we identify dysregulated developmental trajectories of anterior SHF cell fate, leading to a striking single ventricle phenotype in knockout embryos. We then applied our findings to our precardiac organoid model and found that Wnt2 is sufficient to restore SHF cell fate in our model of disrupted endogenous Wnt signaling. In this study, we provide a basis for SHF cell fate decision-proliferation vs. differentiation-autoregulated by CPCs through Wnt.


Subject(s)
Heart Defects, Congenital , Heart , Humans , Heart/physiology , Cell Differentiation , Wnt Signaling Pathway , Wnt Proteins/genetics , Wnt Proteins/metabolism , Gene Expression Regulation, Developmental
4.
Cell Stem Cell ; 29(9): 1285-1287, 2022 09 01.
Article in English | MEDLINE | ID: mdl-36055186

ABSTRACT

In this issue of Cell Stem Cell, Yang et al. devise a protocol for induction and differentiation of human heart-field precursors, drawing on inspiration from in vivo development. Intricate computational analyses uncovered conserved factors governing heart-field segregation, which facilitate enhanced study of human heart development and disease in the dish.


Subject(s)
Heart , Biomarkers , Cell Differentiation , Cell Lineage , Humans
5.
Sci Adv ; 7(49): eabh4181, 2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34851661

ABSTRACT

Neurons can regulate the development, pathogenesis, and regeneration of target organs. However, the role of neurons during heart development and regeneration remains unclear. We genetically inhibited sympathetic innervation in vivo, which resulted in heart enlargement with an increase in cardiomyocyte number. Transcriptomic and protein analysis showed down-regulation of the two clock gene homologs Period1/Period2 (Per1/Per2) accompanied by up-regulation of cell cycle genes. Per1/Per2 deletion increased heart size and cardiomyocyte proliferation, recapitulating sympathetic neuron­deficient hearts. Conversely, increasing sympathetic activity by norepinephrine treatment induced Per1/Per2 and suppressed cardiomyocyte proliferation. We further found that the two clock genes negatively regulate myocyte mitosis entry through the Wee1 kinase pathway. Our findings demonstrate a previously unknown link between cardiac neurons and clock genes in regulation of cardiomyocyte proliferation and heart size and provide mechanistic insights for developing neuromodulation strategies for cardiac regen5eration.

6.
PLoS Comput Biol ; 17(9): e1009305, 2021 09.
Article in English | MEDLINE | ID: mdl-34534204

ABSTRACT

The immaturity of pluripotent stem cell (PSC)-derived tissues has emerged as a universal problem for their biomedical applications. While efforts have been made to generate adult-like cells from PSCs, direct benchmarking of PSC-derived tissues against in vivo development has not been established. Thus, maturation status is often assessed on an ad-hoc basis. Single cell RNA-sequencing (scRNA-seq) offers a promising solution, though cross-study comparison is limited by dataset-specific batch effects. Here, we developed a novel approach to quantify PSC-derived cardiomyocyte (CM) maturation through transcriptomic entropy. Transcriptomic entropy is robust across datasets regardless of differences in isolation protocols, library preparation, and other potential batch effects. With this new model, we analyzed over 45 scRNA-seq datasets and over 52,000 CMs, and established a cross-study, cross-species CM maturation reference. This reference enabled us to directly compare PSC-CMs with the in vivo developmental trajectory and thereby to quantify PSC-CM maturation status. We further found that our entropy-based approach can be used for other cell types, including pancreatic beta cells and hepatocytes. Our study presents a biologically relevant and interpretable metric for quantifying PSC-derived tissue maturation, and is extensible to numerous tissue engineering contexts.


Subject(s)
Benchmarking , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology , Single-Cell Analysis/methods , Transcriptome , Gene Expression , Hepatocytes/cytology , Humans , Insulin-Secreting Cells/cytology , Sequence Analysis, RNA/methods , Tissue Engineering
7.
Biochem Biophys Res Commun ; 577: 12-16, 2021 11 05.
Article in English | MEDLINE | ID: mdl-34487959

ABSTRACT

The Notch pathway is an ancient intercellular signaling system with crucial roles in numerous cell-fate decision processes across species. While the canonical pathway is activated by ligand-induced cleavage and nuclear localization of membrane-bound Notch, Notch can also exert its activity in a ligand/transcription-independent fashion, which is conserved in Drosophila, Xenopus, and mammals. However, the noncanonical role remains poorly understood in in vivo processes. Here we show that increased levels of the Notch intracellular domain (NICD) in the early mesoderm inhibit heart development, potentially through impaired induction of the second heart field (SHF), independently of the transcriptional effector RBP-J. Similarly, inhibiting Notch cleavage, shown to increase noncanonical Notch activity, suppressed SHF induction in embryonic stem cell (ESC)-derived mesodermal cells. In contrast, NICD overexpression in late cardiac progenitor cells lacking RBP-J resulted in an increase in heart size. Our study suggests that noncanonical Notch signaling has stage-specific roles during cardiac development.


Subject(s)
Heart/embryology , Myocardium/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , Cell Differentiation , Cells, Cultured , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Homeobox Protein Nkx-2.5/genetics , Homeobox Protein Nkx-2.5/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mesoderm/cytology , Mesoderm/embryology , Mesoderm/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Myocardium/cytology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
8.
Semin Cell Dev Biol ; 118: 119-128, 2021 10.
Article in English | MEDLINE | ID: mdl-33775518

ABSTRACT

Organoids, or miniaturized organs formed in vitro, hold potential to revolutionize how researchers approach and answer fundamental biological and pathological questions. In the context of cardiac biology, development of a bona fide cardiac organoid enables study of heart development, function, and pathogenesis in a dish, providing insight into the nature of congenital heart disease and offering the opportunity for high-throughput probing of adult heart disease and drug discovery. Recently, multiple groups have reported novel methods for generating in vitro models of the heart; however, there are substantial conceptual and methodological differences. In this review we will evaluate recent cardiac organoid studies through the lens of the core principles of organoid technology: patterned self-organization of multiple cell types resembling the in vivo organ. Based on this, we will classify systems into the following related types of tissues: developmental cardiac organoids, chamber cardiac organoids, microtissues, and engineered heart tissues. Furthermore, we highlight the interventions which allow for organoid formation, such as modulation of highly conserved cardiogenic signaling pathways mediated by developmental morphogens. We expect that consolidation and categorization of existing organoid models will help eliminate confusion in the field and facilitate progress towards creation of an ideal cardiac organoid.


Subject(s)
Heart/growth & development , Organogenesis/physiology , Organoids/growth & development , Humans
9.
Curr Cardiol Rep ; 23(5): 38, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33694131

ABSTRACT

PURPOSE OF REVIEW: Heart development is a meticulously coordinated process that involves the specification of two distinct populations of cardiac progenitor cells, namely the first and the second heart field. Disruption of heart field progenitors can result in congenital heart defects. In this review, we aim to describe the signaling pathways and transcription factors that link heart field development and congenital heart disease. RECENT FINDINGS: Single-cell transcriptomics, lineage-tracing mouse models, and stem cell-based in vitro modeling of cardiogenesis have significantly improved the spatiotemporal characterization of cardiac progenitors. Additionally, novel functional genomic studies have now linked more genetic variants with congenital heart disease. Dysregulation of cardiac progenitor cells causes malformations that can be lethal. Ongoing research will continue to shed light on cardiac morphogenesis and help us better understand and treat patients with congenital heart disease.


Subject(s)
Heart Defects, Congenital , Heart , Animals , Humans , Mice , Myocardium , Signal Transduction , Stem Cells
10.
Nat Commun ; 12(1): 1648, 2021 03 12.
Article in English | MEDLINE | ID: mdl-33712605

ABSTRACT

Cardiomyocytes undergo significant structural and functional changes after birth, and these fundamental processes are essential for the heart to pump blood to the growing body. However, due to the challenges of isolating single postnatal/adult myocytes, how individual newborn cardiomyocytes acquire multiple aspects of the mature phenotype remains poorly understood. Here we implement large-particle sorting and analyze single myocytes from neonatal to adult hearts. Early myocytes exhibit wide-ranging transcriptomic and size heterogeneity that is maintained until adulthood with a continuous transcriptomic shift. Gene regulatory network analysis followed by mosaic gene deletion reveals that peroxisome proliferator-activated receptor coactivator-1 signaling, which is active in vivo but inactive in pluripotent stem cell-derived cardiomyocytes, mediates the shift. This signaling simultaneously regulates key aspects of cardiomyocyte maturation through previously unrecognized proteins, including YAP1 and SF3B2. Our study provides a single-cell roadmap of heterogeneous transitions coupled to cellular features and identifies a multifaceted regulator controlling cardiomyocyte maturation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Myocytes, Cardiac/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , RNA Splicing Factors/metabolism , Transcription Factors/metabolism , Animals , Calcium/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation , Gene Regulatory Networks , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Peroxisome Proliferator-Activated Receptors/genetics , Pluripotent Stem Cells/metabolism , Signal Transduction , Transcription Factors/genetics , Transcriptome , YAP-Signaling Proteins
11.
Sci Transl Med ; 13(581)2021 02 17.
Article in English | MEDLINE | ID: mdl-33597260

ABSTRACT

Myocyte death occurs in many inherited and acquired cardiomyopathies, including arrhythmogenic cardiomyopathy (ACM), a genetic heart disease plagued by the prevalence of sudden cardiac death. Individuals with ACM and harboring pathogenic desmosomal variants, such as desmoglein-2 (DSG2), often show myocyte necrosis with progression to exercise-associated heart failure. Here, we showed that homozygous Dsg2 mutant mice (Dsg2 mut/mut), a model of ACM, die prematurely during swimming and display myocardial dysfunction and necrosis. We detected calcium (Ca2+) overload in Dsg2 mut/mut hearts, which induced calpain-1 (CAPN1) activation, association of CAPN1 with mitochondria, and CAPN1-induced cleavage of mitochondrial-bound apoptosis-inducing factor (AIF). Cleaved AIF translocated to the myocyte nucleus triggering large-scale DNA fragmentation and cell death, an effect potentiated by mitochondrial-driven AIF oxidation. Posttranslational oxidation of AIF cysteine residues was due, in part, to a depleted mitochondrial thioredoxin-2 redox system. Hearts from exercised Dsg2 mut/mut mice were depleted of calpastatin (CAST), an endogenous CAPN1 inhibitor, and overexpressing CAST in myocytes protected against Ca2+ overload-induced necrosis. When cardiomyocytes differentiated from Dsg2 mut/mut embryonic stem cells (ES-CMs) were challenged with ß-adrenergic stimulation, CAPN1 inhibition attenuated CAPN1-induced AIF truncation. In addition, pretreatment of Dsg2 mut/mut ES-CMs with an AIF-mimetic peptide, mirroring the cyclophilin-A (PPIA) binding site of AIF, blocked PPIA-mediated AIF-nuclear translocation, and reduced both apoptosis and necrosis. Thus, preventing CAPN1-induced AIF-truncation or barring binding of AIF to the nuclear chaperone, PPIA, may avert myocyte death and, ultimately, disease progression to heart failure in ACM and likely other forms of cardiomyopathies.


Subject(s)
Apoptosis Inducing Factor , Calpain , Cardiomyopathies , Myocytes, Cardiac/pathology , Physical Conditioning, Animal , Animals , Apoptosis Inducing Factor/metabolism , Calpain/metabolism , Cardiomyopathies/metabolism , Cell Death , Mice , Mitochondria/metabolism , Myocytes, Cardiac/metabolism
12.
Cell Stem Cell ; 28(2): 230-240.e6, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33176168

ABSTRACT

Organoids are powerful models for studying tissue development, physiology, and disease. However, current culture systems disrupt the inductive tissue-tissue interactions needed for the complex morphogenetic processes of native organogenesis. Here, we show that mouse embryonic stem cells (mESCs) can be coaxed to robustly undergo fundamental steps of early heart organogenesis with an in-vivo-like spatiotemporal fidelity. These axially patterned embryonic organoids (gastruloids) mimic embryonic development and support the generation of cardiovascular progenitors, including first and second heart fields. The cardiac progenitors self-organize into an anterior domain reminiscent of a cardiac crescent before forming a beating cardiac tissue near a putative primitive gut-like tube, from which it is separated by an endocardial-like layer. These findings unveil the surprising morphogenetic potential of mESCs to execute key aspects of organogenesis through the coordinated development of multiple tissues. This platform could be an excellent tool for studying heart development in unprecedented detail and throughput.


Subject(s)
Organogenesis , Organoids , Animals , Embryonic Development , Heart , Mice , Mouse Embryonic Stem Cells
13.
Sci Rep ; 10(1): 4249, 2020 03 06.
Article in English | MEDLINE | ID: mdl-32144297

ABSTRACT

Pluripotent stem cell-derived cardiomyocytes (PSC-CMs) hold great promise for disease modeling and drug discovery. However, PSC-CMs exhibit immature phenotypes in culture, and the lack of maturity limits their broad applications. While physical and functional analyses are generally used to determine the status of cardiomyocyte maturation, they could be time-consuming and often present challenges in comparing maturation-enhancing strategies. Therefore, there is a demand for a method to assess cardiomyocyte maturation rapidly and reproducibly. In this study, we found that Myomesin-2 (Myom2), encoding M-protein, is upregulated postnatally, and based on this, we targeted TagRFP to the Myom2 locus in mouse embryonic stem cells. Myom2-RFP+ PSC-CMs exhibited more mature phenotypes than RFP- cells in morphology, function and transcriptionally, conductive to sarcomere shortening assays. Using this system, we screened extracellular matrices (ECMs) and identified laminin-511/521 as potent enhancers of cardiomyocyte maturation. Together, we developed and characterized a novel fluorescent reporter system for the assessment of cardiomyocyte maturation and identified potent maturation-enhancing ECMs through this simple and rapid assay. This system is expected to facilitate use of PSC-CMs in a variety of scientific and medical investigations.


Subject(s)
Biomarkers , Cell Differentiation , Gene Expression , Genes, Reporter , Laminin/genetics , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Calcium , Cell Differentiation/genetics , Computational Biology/methods , Extracellular Matrix/metabolism , Gene Expression Profiling , Laminin/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Sarcomeres/metabolism , Transcription, Genetic , Transcriptome
15.
J Vis Exp ; (149)2019 07 03.
Article in English | MEDLINE | ID: mdl-31329174

ABSTRACT

Pluripotent stem cells offer great potential for understanding heart development and disease and for regenerative medicine. While recent advances in developmental cardiology have led to generating cardiac cells from pluripotent stem cells, it is unclear if the two cardiac fields - the first and second heart fields (FHF and SHF) - are induced in pluripotent stem cells systems. To address this, we generated a protocol for in vitro specification and isolation of heart field-specific cardiac progenitor cells. We used embryonic stem cells lines carrying Hcn4-GFP and Tbx1-Cre; Rosa-RFP reporters of the FHF and the SHF, respectively, and live cell immunostaining of the cell membrane protein Cxcr4, a SHF marker. With this approach, we generated progenitor cells which recapitulate the functional properties and transcriptome of their in vivo counterparts. Our protocol can be utilized to study early specification and segregation of the two heart fields and to generate chamber-specific cardiac cells for heart disease modelling. Since this is an in vitro organoid system, it may not provide precise anatomical information. However, this system overcomes the poor accessibility of gastrulation-stage embryos and can be upscaled for high-throughput screens.


Subject(s)
Heart/embryology , Myoblasts, Cardiac/cytology , Myocardium/cytology , Animals , Biomarkers/metabolism , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Myoblasts, Cardiac/metabolism , Myocardium/metabolism , Organoids/cytology , Organoids/metabolism , Signal Transduction , Transcriptome
16.
Nat Commun ; 9(1): 3140, 2018 08 07.
Article in English | MEDLINE | ID: mdl-30087351

ABSTRACT

The discovery of the first heart field (FHF) and the second heart field (SHF) led us to understand how cardiac lineages and structures arise during development. However, it remains unknown how they are specified. Here, we generate precardiac spheroids with pluripotent stem cells (PSCs) harboring GFP/RFP reporters under the control of FHF/SHF markers, respectively. GFP+ cells and RFP+ cells appear from two distinct areas and develop in a complementary fashion. Transcriptome analysis shows a high degree of similarities with embryonic FHF/SHF cells. Bmp and Wnt are among the most differentially regulated pathways, and gain- and loss-of-function studies reveal that Bmp specifies GFP+ cells and RFP+ cells via the Bmp/Smad pathway and Wnt signaling, respectively. FHF/SHF cells can be isolated without reporters by the surface protein Cxcr4. This study provides novel insights into understanding the specification of two cardiac origins, which can be leveraged for PSC-based modeling of heart field/chamber-specific disease.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Gene Expression Regulation, Developmental , Heart/physiology , Organoids/metabolism , Receptors, CXCR4/metabolism , Wnt Signaling Pathway , Animals , Cell Differentiation , Cell Lineage , Cell Separation , Crosses, Genetic , Flow Cytometry , Gene Library , Green Fluorescent Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Myocardium/metabolism , Pluripotent Stem Cells/cytology , RNA, Small Interfering/metabolism , Sequence Analysis, RNA , Time Factors , Transcriptome
17.
Biochem Biophys Res Commun ; 500(2): 256-260, 2018 06 02.
Article in English | MEDLINE | ID: mdl-29653101

ABSTRACT

Proper control of multipotent/stem cell number and fate is essential for ensuing organ formation during development. ß1-integrin, a subfamily of cell surface receptors, has a conserved role in maintenance of multipotent/stem cells, including renal progenitor cells, follicle stem cells, epidermal stem cells and neural stem cells. However, it remains unclear whether ß1-integrin has a role in cardiac progenitor cell (CPC) development. Here we show that a mesodermal deletion of ß1-integrin decreases Isl1+ cell number in the second pharyngeal arch (PA2), where CPCs undergo renewal and expansion. Mesp1 lineage-specific mosaicism revealed that ß1-integrin-deleted Isl1+ cells do not proliferate in the PA2. Consistently, ß1-integrin-deleted Isl1+ CPCs failed to expand in vitro, independent of PA2 cells. ß1-integrin co-localized and physically associated with Numb, a crucial regulator of CPC renewal and expansion. Importantly, Numb/Numbl-deleted CPCs showed dramatic reduction in ß1-integrin levels. These findings suggest that ß1-integrin is a key mediator of the Numb pathway in CPC maintenance.


Subject(s)
Integrin beta1/metabolism , Membrane Proteins/metabolism , Myocytes, Cardiac/cytology , Nerve Tissue Proteins/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Line , Cell Proliferation , Embryo, Mammalian/metabolism , Gene Deletion , Heart/embryology , Intracellular Signaling Peptides and Proteins , Mice, Knockout , Protein Binding
18.
Cell Rep ; 13(8): 1705-16, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26586429

ABSTRACT

Decades of progress in developmental cardiology has advanced our understanding of the early aspects of heart development, including cardiomyocyte (CM) differentiation. However, control of the CM maturation that is subsequently required to generate adult myocytes remains elusive. Here, we analyzed over 200 microarray datasets from early embryonic to adult hearts and identified a large number of genes whose expression shifts gradually and continuously during maturation. We generated an atlas of integrated gene expression, biological pathways, transcriptional regulators, and gene regulatory networks (GRNs), which show discrete sets of key transcriptional regulators and pathways activated or suppressed during CM maturation. We developed a GRN-based program named MatStat(CM) that indexes CM maturation status. MatStat(CM) reveals that pluripotent-stem-cell-derived CMs mature early in culture but are arrested at the late embryonic stage with aberrant regulation of key transcription factors. Our study provides a foundation for understanding CM maturation.


Subject(s)
Myocytes, Cardiac/cytology , Transcription, Genetic/genetics , Animals , Cell Differentiation/genetics , Cell Line , Embryonic Stem Cells/cytology , Gene Expression/genetics , Gene Expression Regulation, Developmental/genetics , Gene Regulatory Networks/genetics , Mice , Pluripotent Stem Cells/cytology , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...