Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Nat Commun ; 14(1): 1722, 2023 04 03.
Article in English | MEDLINE | ID: mdl-37012244

ABSTRACT

Cardiogenesis relies on the precise spatiotemporal coordination of multiple progenitor populations. Understanding the specification and differentiation of these distinct progenitor pools during human embryonic development is crucial for advancing our knowledge of congenital cardiac malformations and designing new regenerative therapies. By combining genetic labelling, single-cell transcriptomics, and ex vivo human-mouse embryonic chimeras we uncovered that modulation of retinoic acid signaling instructs human pluripotent stem cells to form heart field-specific progenitors with distinct fate potentials. In addition to the classical first and second heart fields, we observed the appearance of juxta-cardiac field progenitors giving rise to both myocardial and epicardial cells. Applying these findings to stem-cell based disease modelling we identified specific transcriptional dysregulation in first and second heart field progenitors derived from stem cells of patients with hypoplastic left heart syndrome. This highlights the suitability of our in vitro differentiation platform for studying human cardiac development and disease.


Subject(s)
Pluripotent Stem Cells , Tretinoin , Humans , Animals , Mice , Tretinoin/pharmacology , Heart , Myocardium , Cell Differentiation , Myocytes, Cardiac
2.
Circulation ; 145(22): 1663-1683, 2022 05 31.
Article in English | MEDLINE | ID: mdl-35400201

ABSTRACT

BACKGROUND: Transcriptional reconfiguration is central to heart failure, the most common cause of which is dilated cardiomyopathy (DCM). The effect of 3-dimensional chromatin topology on transcriptional dysregulation and pathogenesis in human DCM remains elusive. METHODS: We generated a compendium of 3-dimensional epigenome and transcriptome maps from 101 biobanked human DCM and nonfailing heart tissues through highly integrative chromatin immunoprecipitation (H3K27ac [acetylation of lysine 27 on histone H3]), in situ high-throughput chromosome conformation capture, chromatin immunoprecipitation sequencing, assay for transposase-accessible chromatin using sequencing, and RNA sequencing. We used human induced pluripotent stem cell-derived cardiomyocytes and mouse models to interrogate the key transcription factor implicated in 3-dimensional chromatin organization and transcriptional regulation in DCM pathogenesis. RESULTS: We discovered that the active regulatory elements (H3K27ac peaks) and their connectome (H3K27ac loops) were extensively reprogrammed in DCM hearts and contributed to transcriptional dysregulation implicated in DCM development. For example, we identified that nontranscribing NPPA-AS1 (natriuretic peptide A antisense RNA 1) promoter functions as an enhancer and physically interacts with the NPPA (natriuretic peptide A) and NPPB (natriuretic peptide B) promoters, leading to the cotranscription of NPPA and NPPB in DCM hearts. We revealed that DCM-enriched H3K27ac loops largely resided in conserved high-order chromatin architectures (compartments, topologically associating domains) and their anchors unexpectedly had equivalent chromatin accessibility. We discovered that the DCM-enriched H3K27ac loop anchors exhibited a strong enrichment for HAND1 (heart and neural crest derivatives expressed 1), a key transcription factor involved in early cardiogenesis. In line with this, its protein expression was upregulated in human DCM and mouse failing hearts. To further validate whether HAND1 is a causal driver for the reprogramming of enhancer-promoter connectome in DCM hearts, we performed comprehensive 3-dimensional epigenome mappings in human induced pluripotent stem cell-derived cardiomyocytes. We found that forced overexpression of HAND1 in human induced pluripotent stem cell-derived cardiomyocytes induced a distinct gain of enhancer-promoter connectivity and correspondingly increased the expression of their connected genes implicated in DCM pathogenesis, thus recapitulating the transcriptional signature in human DCM hearts. Electrophysiology analysis demonstrated that forced overexpression of HAND1 in human induced pluripotent stem cell-derived cardiomyocytes induced abnormal calcium handling. Furthermore, cardiomyocyte-specific overexpression of Hand1 in the mouse hearts resulted in dilated cardiac remodeling with impaired contractility/Ca2+ handling in cardiomyocytes, increased ratio of heart weight/body weight, and compromised cardiac function, which were ascribed to recapitulation of transcriptional reprogramming in DCM. CONCLUSIONS: This study provided novel chromatin topology insights into DCM pathogenesis and illustrated a model whereby a single transcription factor (HAND1) reprograms the genome-wide enhancer-promoter connectome to drive DCM pathogenesis.


Subject(s)
Cardiomyopathy, Dilated , Induced Pluripotent Stem Cells , Animals , Cardiomyopathy, Dilated/metabolism , Chromatin/genetics , Chromatin/metabolism , Histones/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Transcription Factors/genetics
3.
Stem Cell Reports ; 16(6): 1398-1408, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34048692

ABSTRACT

The International Society for Stem Cell Research has updated its Guidelines for Stem Cell Research and Clinical Translation in order to address advances in stem cell science and other relevant fields, together with the associated ethical, social, and policy issues that have arisen since the last update in 2016. While growing to encompass the evolving science, clinical applications of stem cells, and the increasingly complex implications of stem cell research for society, the basic principles underlying the Guidelines remain unchanged, and they will continue to serve as the standard for the field and as a resource for scientists, regulators, funders, physicians, and members of the public, including patients. A summary of the key updates and issues is presented here.


Subject(s)
Bioethical Issues/standards , Policy , Practice Guidelines as Topic , Societies, Scientific/standards , Stem Cell Research/ethics , Stem Cells , Humans , Societies, Scientific/ethics
4.
Stem Cell Reports ; 16(6): 1409-1415, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34048695

ABSTRACT

The newly revised 2021 ISSCR Guidelines for Stem Cell Research and Clinical Translation includes scientific and ethical guidance for the transfer of human pluripotent stem cells and their direct derivatives into animal models. In this white paper, the ISSCR subcommittee that drafted these guidelines for research involving the use of nonhuman embryos and postnatal animals explains and summarizes their recommendations.


Subject(s)
Chimera , Embryo Research/ethics , Pluripotent Stem Cells , Practice Guidelines as Topic , Societies, Scientific/standards , Stem Cell Research/ethics , Stem Cell Transplantation/standards , Animals , Humans , Societies, Scientific/ethics , Stem Cell Transplantation/ethics
5.
Cell Stem Cell ; 28(5): 787-789, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33961759

ABSTRACT

Recently in Cell, Tan et al. (2021) report the successful generation of human-monkey chimeras in vitro, providing an opportunity for new insights into the biology of human stem cells and early human development in an embryonic environment that is evolutionary closer to human than previously studied rodent and domestic species.


Subject(s)
Chimera , Pluripotent Stem Cells , Animals , Haplorhini , Humans
6.
Epigenetics Chromatin ; 12(1): 14, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30767785

ABSTRACT

BACKGROUND: Genomic imprinting, resulting in parent-of-origin specific gene expression, plays a critical role in mammalian development. Here, we apply allele-specific RNA-seq on isogenic B6D2F1 mice to assay imprinted genes in tissues from early embryonic tissues between E3.5 and E7.25 and in pluripotent cell lines to evaluate maintenance of imprinted gene expression. For the cell lines, we include embryonic stem cells (ESCs) and epiblast stem cells (EpiSCs) derived from fertilized embryos and from embryos obtained after nuclear transfer (NT) or parthenogenetic activation (PGA). RESULTS: As homozygous genomic regions of PGA-derived cells are not compatible with allele-specific RNA-seq, we developed an RNA-seq-based genotyping strategy allowing identification of informative heterozygous regions. Global analysis shows that proper imprinted gene expression as observed in embryonic tissues is largely lost in the ESC lines included in this study, which mainly consisted of female ESCs. Differentiation of ESC lines to embryoid bodies or NPCs does not restore monoallelic expression of imprinted genes, neither did reprogramming of the serum-cultured ESCs to the pluripotent ground state by the use of 2 kinase inhibitors. Fertilized EpiSC and EpiSC-NT lines largely maintain imprinted gene expression, as did EpiSC-PGA lines that show known paternally expressed genes being silent and known maternally expressed genes consistently showing doubled expression. Notably, two EpiSC-NT lines show aberrant silencing of Rian and Meg3, two critically imprinted genes in mouse iPSCs. With respect to female EpiSC, most of the lines displayed completely skewed X inactivation suggesting a (near) clonal origin. CONCLUSIONS: Altogether, our analysis provides a comprehensive overview of imprinted gene expression in pluripotency and provides a benchmark to allow identification of cell lines that faithfully maintain imprinted gene expression and therefore retain full developmental potential.


Subject(s)
Alleles , Genomic Imprinting , Mouse Embryonic Stem Cells/metabolism , RNA, Messenger/genetics , Animals , Cell Differentiation , Cell Line , Cells, Cultured , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Silencing , Germ Layers/cytology , Germ Layers/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mouse Embryonic Stem Cells/cytology
7.
Biol Open ; 7(8)2018 Aug 17.
Article in English | MEDLINE | ID: mdl-30026265

ABSTRACT

During early mammalian development, transient pools of pluripotent cells emerge that can be immortalised upon stem cell derivation. The pluripotent state, 'naïve' or 'primed', depends on the embryonic stage and derivation conditions used. Here we analyse the temporal gene expression patterns of mouse, cattle and porcine embryos at stages that harbour different types of pluripotent cells. We document conserved and divergent traits in gene expression, and identify predictor genes shared across the species that are associated with pluripotent states in vivo and in vitro Amongst these are the pluripotency-linked genes Klf4 and Lin28b The novel genes discovered include naïve- (Spic, Scpep1 and Gjb5) and primed-associated (Sema6a and Jakmip2) genes as well as naïve to primed transition genes (Dusp6 and Trip6). Both Gjb5 and Dusp6 play a role in pluripotency since their knockdown results in differentiation and downregulation of key pluripotency genes. Our interspecies comparison revealed new insights of pluripotency, pluripotent stem cell identity and a new molecular criterion for distinguishing between pluripotent states in various species, including human.

8.
Eur Respir J ; 51(4)2018 04.
Article in English | MEDLINE | ID: mdl-29449428

ABSTRACT

Genetic defects in bone morphogenetic protein type II receptor (BMPRII) signalling and inflammation contribute to the pathogenesis of pulmonary arterial hypertension (PAH). The receptor is activated by bone morphogenetic protein (BMP) ligands, which also enhance BMPR2 transcription. A small-molecule BMP upregulator with selectivity on vascular endothelium would be a desirable therapeutic intervention for PAH.We assayed compounds identified in the screening of BMP2 upregulators for their ability to increase the expression of inhibitor of DNA binding 1 (Id1), using a dual reporter driven specifically in human embryonic stem cell-derived endothelial cells. These assays identified a novel piperidine, BMP upregulator 1 (BUR1), that increased endothelial Id1 expression with a half-maximal effective concentration of 0.098 µmol·L-1 Microarray analyses and immunoblotting showed that BUR1 induced BMP2 and prostaglandin-endoperoxide synthase 2 (PTGS2) expression. BUR1 effectively rescued deficient angiogenesis in autologous BMPR2+/R899X endothelial cells generated by CRISPR/Cas9 and patient cells.BUR1 prevented and reversed PAH in monocrotaline rats, and restored BMPRII downstream signalling and modulated the arachidonic acid pathway in the pulmonary arterial endothelium in the Sugen 5416/hypoxia PAH mouse model.In conclusion, using stem cell technology we have provided a novel small-molecule compound which regulates BMP2 and PTGS2 levels that might be useful for the treatment of PAH.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Cyclooxygenase 2/metabolism , Endothelial Cells/drug effects , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/metabolism , Piperidines/pharmacology , Animals , Cell Line , Cell Proliferation , Dinoprostone/blood , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Leukotriene B4/blood , Pulmonary Artery/drug effects , Rats , Signal Transduction/drug effects , Stem Cells/cytology
10.
Stem Cell Reports ; 8(4): 803-812, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28344001

ABSTRACT

The isolation or in vitro derivation of many human cell types remains challenging and inefficient. Direct conversion of human pluripotent stem cells (hPSCs) by forced expression of transcription factors provides a potential alternative. However, deficient inducible gene expression in hPSCs has compromised efficiencies of forward programming approaches. We have systematically optimized inducible gene expression in hPSCs using a dual genomic safe harbor gene-targeting strategy. This approach provides a powerful platform for the generation of human cell types by forward programming. We report robust and deterministic reprogramming of hPSCs into neurons and functional skeletal myocytes. Finally, we present a forward programming strategy for rapid and highly efficient generation of human oligodendrocytes.


Subject(s)
Cell Differentiation , Gene Targeting/methods , Muscle Fibers, Skeletal/cytology , Neurons/cytology , Oligodendroglia/cytology , Pluripotent Stem Cells/cytology , Cell Line , Cellular Reprogramming , Gene Expression , Humans , Muscle Development , Muscle Fibers, Skeletal/metabolism , Neurogenesis , Neurons/metabolism , Oligodendroglia/metabolism , Pluripotent Stem Cells/metabolism , Transgenes , Up-Regulation
12.
Cell Rep ; 17(5): 1369-1382, 2016 10 25.
Article in English | MEDLINE | ID: mdl-27783950

ABSTRACT

The polycomb repressive complex 2 (PRC2) methylates lysine 27 of histone H3 (H3K27) through its catalytic subunit Ezh2. PRC2-mediated di- and tri-methylation (H3K27me2/H3K27me3) have been interchangeably associated with gene repression. However, it remains unclear whether these two degrees of H3K27 methylation have different functions. In this study, we have generated isogenic mouse embryonic stem cells (ESCs) with a modified H3K27me2/H3K27me3 ratio. Our findings document dynamic developmental control in the genomic distribution of H3K27me2 and H3K27me3 at regulatory regions in ESCs. They also reveal that modifying the ratio of H3K27me2 and H3K27me3 is sufficient for the acquisition and repression of defined cell lineage transcriptional programs and phenotypes and influences induction of the ESC ground state.


Subject(s)
Cell Lineage , Histones/metabolism , Lysine/metabolism , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Regulation , Genome , Methylation , Mice , Neurons/cytology , RNA Editing , Regulatory Sequences, Nucleic Acid/genetics , Transcription Activator-Like Effector Nucleases/metabolism , Transcription, Genetic
13.
Cell Stem Cell ; 19(2): 163-175, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27494674

ABSTRACT

Chimeras are widely acknowledged as the gold standard for assessing stem cell pluripotency, based on their capacity to test donor cell lineage potential in the context of an organized, normally developing tissue. Experimental chimeras provide key insights into mammalian developmental mechanisms and offer a resource for interrogating the fate potential of various pluripotent stem cell states. We highlight the applications and current limitations presented by intra- and inter-species chimeras and consider their future contribution to the stem cell field. Despite the technical and ethical demands of experimental chimeras, including human-interspecies chimeras, they are a provocative resource for achieving regenerative medicine goals.


Subject(s)
Chimera/metabolism , Pluripotent Stem Cells/cytology , Stem Cell Research , Animals , Cell Lineage , Embryo, Mammalian/cytology , Humans , Species Specificity
14.
Nat Commun ; 7: 11208, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27052461

ABSTRACT

The production of megakaryocytes (MKs)--the precursors of blood platelets--from human pluripotent stem cells (hPSCs) offers exciting clinical opportunities for transfusion medicine. Here we describe an original approach for the large-scale generation of MKs in chemically defined conditions using a forward programming strategy relying on the concurrent exogenous expression of three transcription factors: GATA1, FLI1 and TAL1. The forward programmed MKs proliferate and differentiate in culture for several months with MK purity over 90% reaching up to 2 × 10(5) mature MKs per input hPSC. Functional platelets are generated throughout the culture allowing the prospective collection of several transfusion units from as few as 1 million starting hPSCs. The high cell purity and yield achieved by MK forward programming, combined with efficient cryopreservation and good manufacturing practice (GMP)-compatible culture, make this approach eminently suitable to both in vitro production of platelets for transfusion and basic research in MK and platelet biology.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cellular Reprogramming , GATA1 Transcription Factor/genetics , Megakaryocytes/cytology , Pluripotent Stem Cells/cytology , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Proteins/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blood Platelets/cytology , Blood Platelets/metabolism , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Cryopreservation/methods , GATA1 Transcription Factor/metabolism , Gene Expression Regulation , Genes, Reporter , Genetic Vectors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Lentivirus/genetics , Megakaryocytes/metabolism , Microarray Analysis , Pluripotent Stem Cells/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , T-Cell Acute Lymphocytic Leukemia Protein 1 , Transduction, Genetic , Transgenes
16.
Methods Mol Biol ; 1357: 23-31, 2016.
Article in English | MEDLINE | ID: mdl-25687300

ABSTRACT

This protocol describes the efficient isolation of peripheral blood mononuclear cells from circulating blood via density gradient centrifugation and subsequent generation of integration-free human induced pluripotent stem cells. Peripheral blood mononuclear cells are cultured for 9 days to allow expansion of the erythroblast population. The erythroblasts are then used to derive human induced pluripotent stem cells using Sendai viral vectors, each expressing one of the four reprogramming factors Oct4, Sox2, Klf4, and c-Myc.


Subject(s)
Cell Culture Techniques/methods , Cellular Reprogramming Techniques/methods , Cellular Reprogramming , Genetic Vectors/genetics , Induced Pluripotent Stem Cells/cytology , Leukocytes, Mononuclear/cytology , Sendai virus/genetics , Animals , Cell Separation/methods , Cells, Cultured , Culture Media , Cytokines/pharmacology , Erythroblasts/cytology , Erythroblasts/drug effects , Erythroblasts/virology , Fibroblasts/cytology , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/physiology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/virology , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/physiology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/physiology , Recombinant Proteins/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/physiology
17.
Cell Stem Cell ; 18(1): 67-72, 2016 Jan 07.
Article in English | MEDLINE | ID: mdl-26712580

ABSTRACT

Pluripotent stem cells are defined by their capacity to differentiate into all three tissue layers that comprise the body. Chimera formation, generated by stem cell transplantation to the embryo, is a stringent assessment of stem cell pluripotency. However, the ability of human pluripotent stem cells (hPSCs) to form embryonic chimeras remains in question. Here we show using a stage-matching approach that human induced pluripotent stem cells (hiPSCs) and human embryonic stem cells (hESCs) have the capacity to participate in normal mouse development when transplanted into gastrula-stage embryos, providing in vivo functional validation of hPSC pluripotency. hiPSCs and hESCs form interspecies chimeras with high efficiency, colonize the embryo in a manner predicted from classical developmental fate mapping, and differentiate into each of the three primary tissue layers. This faithful recapitulation of tissue-specific fate post-transplantation underscores the functional potential of hPSCs and provides evidence that human-mouse interspecies developmental competency can occur.


Subject(s)
Cell Culture Techniques , Chimerism , Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Animals , Body Patterning , Cell Differentiation , Cell Lineage , Cell Proliferation , Cells, Cultured , Gastrula/physiology , Humans , Mice , Regenerative Medicine , Species Specificity
18.
Stem Cell Reports ; 5(4): 532-45, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26388287

ABSTRACT

Retinal progenitors are initially found in the anterior neural plate region known as the eye field, whereas neighboring areas undertake telencephalic or hypothalamic development. Eye field cells become specified by switching on a network of eye field transcription factors, but the extracellular cues activating this network remain unclear. In this study, we used chemically defined media to induce in vitro differentiation of mouse embryonic stem cells (ESCs) toward eye field fates. Inhibition of Wnt/ß-catenin signaling was sufficient to drive ESCs to telencephalic, but not retinal, fates. Instead, retinal progenitors could be generated from competent differentiating mouse ESCs by activation of Activin/Nodal signaling within a narrow temporal window corresponding to the emergence of primitive anterior neural progenitors. Activin also promoted eye field gene expression in differentiating human ESCs. Our results reveal insights into the mechanisms of eye field specification and open new avenues toward the generation of retinal progenitors for translational medicine.


Subject(s)
Activins/metabolism , Embryonic Stem Cells/cytology , Nodal Protein/metabolism , Pluripotent Stem Cells/cytology , Retina/embryology , Signal Transduction , Animals , Cell Differentiation , Cell Line , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Humans , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Retina/cytology , Retina/metabolism , Wnt Signaling Pathway
19.
Genome Biol ; 16: 112, 2015 May 30.
Article in English | MEDLINE | ID: mdl-26025256

ABSTRACT

BACKGROUND: Selective maintenance of genomic epigenetic imprints during pre-implantation development is required for parental origin-specific expression of imprinted genes. The Kruppel-like zinc finger protein ZFP57 acts as a factor necessary for maintaining the DNA methylation memory at multiple imprinting control regions in early mouse embryos and embryonic stem (ES) cells. Maternal-zygotic deletion of ZFP57 in mice presents a highly penetrant phenotype with no animals surviving to birth. Additionally, several cases of human transient neonatal diabetes are associated with somatic mutations in the ZFP57 coding sequence. RESULTS: Here, we comprehensively map sequence-specific ZFP57 binding sites in an allele-specific manner using hybrid ES cell lines from reciprocal crosses between C57BL/6J and Cast/EiJ mice, assigning allele specificity to approximately two-thirds of all binding sites. While half of these are biallelic and include endogenous retrovirus (ERV) targets, the rest show monoallelic binding based either on parental origin or on genetic background of the allele. Parental-origin allele-specific binding is methylation-dependent and maps only to imprinting control differentially methylated regions (DMRs) established in the germline. We identify a novel imprinted gene, Fkbp6, which has a critical function in mouse male germ cell development. Genetic background-specific sequence differences also influence ZFP57 binding, as genetic variation that disrupts the consensus binding motif and its methylation is often associated with monoallelic expression of neighboring genes. CONCLUSIONS: The work described here uncovers further roles for ZFP57-mediated regulation of genomic imprinting and identifies a novel mechanism for genetically determined monoallelic gene expression.


Subject(s)
Alleles , DNA-Binding Proteins/metabolism , Genomic Imprinting , Kruppel-Like Transcription Factors/metabolism , Repressor Proteins/metabolism , Animals , Binding Sites , Cell Line , DNA Methylation , Genetic Variation , Histone Code , Mice , Mice, Inbred C57BL
20.
Development ; 142(12): 2121-35, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-26015544

ABSTRACT

The transcription factor brachyury (T, BRA) is one of the first markers of gastrulation and lineage specification in vertebrates. Despite its wide use and importance in stem cell and developmental biology, its functional genomic targets in human cells are largely unknown. Here, we use differentiating human embryonic stem cells to study the role of BRA in activin A-induced endoderm and BMP4-induced mesoderm progenitors. We show that BRA has distinct genome-wide binding landscapes in these two cell populations, and that BRA interacts and collaborates with SMAD1 or SMAD2/3 signalling to regulate the expression of its target genes in a cell-specific manner. Importantly, by manipulating the levels of BRA in cells exposed to different signalling environments, we demonstrate that BRA is essential for mesoderm but not for endoderm formation. Together, our data illuminate the function of BRA in the context of human embryonic development and show that the regulatory role of BRA is context dependent. Our study reinforces the importance of analysing the functions of a transcription factor in different cellular and signalling environments.


Subject(s)
Embryonic Stem Cells/cytology , Fetal Proteins/metabolism , Gene Expression Regulation, Developmental , Neurogenesis/physiology , Smad1 Protein/metabolism , T-Box Domain Proteins/metabolism , Animals , Bone Morphogenetic Protein 4/metabolism , Cell Line , Embryonic Stem Cells/metabolism , Endoderm/cytology , Gastrulation/physiology , Humans , Mesoderm/cytology , Mice , Mice, Transgenic , Smad2 Protein/metabolism , Smad3 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...