Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Development ; 147(20)2020 10 29.
Article in English | MEDLINE | ID: mdl-33028609

ABSTRACT

The genetic regulatory network controlling early fate choices during human blood cell development are not well understood. We used human pluripotent stem cell reporter lines to track the development of endothelial and haematopoietic populations in an in vitro model of human yolk-sac development. We identified SOX17-CD34+CD43- endothelial cells at day 2 of blast colony development, as a haemangioblast-like branch point from which SOX17-CD34+CD43+ blood cells and SOX17+CD34+CD43- endothelium subsequently arose. Most human blood cell development was dependent on RUNX1. Deletion of RUNX1 only permitted a single wave of yolk sac-like primitive erythropoiesis, but no yolk sac myelopoiesis or aorta-gonad-mesonephros (AGM)-like haematopoiesis. Blocking GFI1 and/or GFI1B activity with a small molecule inhibitor abrogated all blood cell development, even in cell lines with an intact RUNX1 gene. Together, our data define the hierarchical requirements for RUNX1, GFI1 and/or GFI1B during early human haematopoiesis arising from a yolk sac-like SOX17-negative haemogenic endothelial intermediate.


Subject(s)
Blood Cells/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , DNA-Binding Proteins/metabolism , Endothelium/metabolism , Hematopoiesis , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , SOXF Transcription Factors/metabolism , Transcription Factors/metabolism , Yolk Sac/metabolism , Blood Cells/cytology , Cell Differentiation , Cell Lineage , Erythroid Cells/cytology , Erythroid Cells/metabolism , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/metabolism , Humans , Models, Biological , Transcription, Genetic
2.
Stem Cells ; 25(9): 2206-14, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17556598

ABSTRACT

We have utilized a serum- and stromal cell-free "spin embryoid body (EB)" differentiation system to investigate the roles of four growth factors, bone morphogenetic protein 4 (BMP4), vascular endothelial growth factor (VEGF), stem cell factor (SCF), and basic fibroblast growth factor (FGF2), singly and in combination, on the generation of hematopoietic cells from human embryonic stem cells (HESCs). Of the four factors, only BMP4 induced expression of genes that signaled the emergence of the primitive streak-like population required for the subsequent development of hematopoietic mesoderm. In addition, BMP4 initiated the expression of genes marking hematopoietic mesoderm and supported the generation of hematopoietic progenitor cells at a low frequency. However, the appearance of robust numbers of hematopoietic colony forming cells and their mature progeny required the inclusion of VEGF. Finally, the combination of BMP4, VEGF, SCF, and FGF2 further enhanced the total yield of hematopoietic cells. These data demonstrate the utility of the serum-free spin EB system in dissecting the roles of specific growth factors required for the directed differentiation of HESCs toward the hematopoietic lineage.


Subject(s)
Bone Morphogenetic Proteins/physiology , Cell Differentiation/drug effects , Culture Media, Serum-Free/pharmacology , Embryonic Stem Cells/cytology , Fibroblast Growth Factor 2/physiology , Hematopoiesis/physiology , Stem Cell Factor/physiology , Vascular Endothelial Growth Factor A/physiology , Animals , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/pharmacology , Cells, Cultured , Drug Combinations , Drug Synergism , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation, Developmental , Hematopoiesis/drug effects , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Primitive Streak/metabolism , Stem Cell Factor/pharmacology , Vascular Endothelial Growth Factor A/pharmacology
3.
Nat Biotechnol ; 34(11): 1168-1179, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27748754

ABSTRACT

The ability to generate hematopoietic stem cells from human pluripotent cells would enable many biomedical applications. We find that hematopoietic CD34+ cells in spin embryoid bodies derived from human embryonic stem cells (hESCs) lack HOXA expression compared with repopulation-competent human cord blood CD34+ cells, indicating incorrect mesoderm patterning. Using reporter hESC lines to track the endothelial (SOX17) to hematopoietic (RUNX1C) transition that occurs in development, we show that simultaneous modulation of WNT and ACTIVIN signaling yields CD34+ hematopoietic cells with HOXA expression that more closely resembles that of cord blood. The cultures generate a network of aorta-like SOX17+ vessels from which RUNX1C+ blood cells emerge, similar to hematopoiesis in the aorta-gonad-mesonephros (AGM). Nascent CD34+ hematopoietic cells and corresponding cells sorted from human AGM show similar expression of cell surface receptors, signaling molecules and transcription factors. Our findings provide an approach to mimic in vitro a key early stage in human hematopoiesis for the generation of AGM-derived hematopoietic lineages from hESCs.


Subject(s)
Embryonic Stem Cells/cytology , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/metabolism , Mesonephros/cytology , Mesonephros/embryology , Neovascularization, Physiologic/physiology , Aorta/cytology , Aorta/embryology , Aorta/growth & development , Cell Differentiation/physiology , Cells, Cultured , Embryonic Stem Cells/physiology , Gonads/cytology , Gonads/embryology , Gonads/growth & development , Hematopoietic Stem Cells/physiology , Humans , Mesonephros/growth & development
4.
Nat Cell Biol ; 17(5): 580-91, 2015 May.
Article in English | MEDLINE | ID: mdl-25915127

ABSTRACT

The generation of haematopoietic stem cells (HSCs) from human pluripotent stem cells (hPSCs) will depend on the accurate recapitulation of embryonic haematopoiesis. In the early embryo, HSCs develop from the haemogenic endothelium (HE) and are specified in a Notch-dependent manner through a process named endothelial-to-haematopoietic transition (EHT). As HE is associated with arteries, it is assumed that it represents a subpopulation of arterial vascular endothelium (VE). Here we demonstrate at a clonal level that hPSC-derived HE and VE represent separate lineages. HE is restricted to the CD34(+)CD73(-)CD184(-) fraction of day 8 embryoid bodies and it undergoes a NOTCH-dependent EHT to generate RUNX1C(+) cells with multilineage potential. Arterial and venous VE progenitors, in contrast, segregate to the CD34(+)CD73(med)CD184(+) and CD34(+)CD73(hi)CD184(-) fractions, respectively. Together, these findings identify HE as distinct from VE and provide a platform for defining the signalling pathways that regulate their specification to functional HSCs.


Subject(s)
Arteries/physiology , Cell Differentiation , Cell Lineage , Endothelial Progenitor Cells/physiology , Hematopoietic Stem Cells/physiology , Multipotent Stem Cells/physiology , Pluripotent Stem Cells/physiology , 5'-Nucleotidase/deficiency , Antigens, CD34/metabolism , Arteries/cytology , Arteries/metabolism , Biomarkers/metabolism , Cell Line , Cell Separation/methods , Coculture Techniques , Core Binding Factor Alpha 2 Subunit/metabolism , Endothelial Progenitor Cells/metabolism , GPI-Linked Proteins/deficiency , Hematopoietic Stem Cells/metabolism , Humans , Microscopy, Video , Multipotent Stem Cells/metabolism , Phenotype , Pluripotent Stem Cells/metabolism , Precursor Cells, T-Lymphoid/physiology , Receptors, CXCR5/deficiency , Receptors, Notch/metabolism , Signal Transduction , Time Factors , Veins/cytology , Veins/physiology
5.
Cell Stem Cell ; 14(2): 237-52, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24412311

ABSTRACT

Human pluripotent stem cell (hPSC) differentiation typically yields heterogeneous populations. Knowledge of signals controlling embryonic lineage bifurcations could efficiently yield desired cell types through exclusion of alternate fates. Therefore, we revisited signals driving induction and anterior-posterior patterning of definitive endoderm to generate a coherent roadmap for endoderm differentiation. With striking temporal dynamics, BMP and Wnt initially specified anterior primitive streak (progenitor to endoderm), yet, 24 hr later, suppressed endoderm and induced mesoderm. At lineage bifurcations, cross-repressive signals separated mutually exclusive fates; TGF-ß and BMP/MAPK respectively induced pancreas versus liver from endoderm by suppressing the alternate lineage. We systematically blockaded alternate fates throughout multiple consecutive bifurcations, thereby efficiently differentiating multiple hPSC lines exclusively into endoderm and its derivatives. Comprehensive transcriptional and chromatin mapping of highly pure endodermal populations revealed that endodermal enhancers existed in a surprising diversity of "pre-enhancer" states before activation, reflecting the establishment of a permissive chromatin landscape as a prelude to differentiation.


Subject(s)
Cell Lineage , Endoderm/embryology , Pluripotent Stem Cells/cytology , Signal Transduction , Animals , Base Sequence , Body Patterning/drug effects , Bone Morphogenetic Proteins/metabolism , Cell Lineage/drug effects , Chromatin/metabolism , Culture Media, Serum-Free/pharmacology , Digestive System/embryology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endoderm/cytology , Endoderm/drug effects , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic/drug effects , Fibroblast Growth Factors/metabolism , Humans , Liver/embryology , MAP Kinase Signaling System/drug effects , Mice , Molecular Sequence Data , Pancreas/embryology , Pluripotent Stem Cells/drug effects , Primitive Streak/cytology , Primitive Streak/embryology , Protein Binding/drug effects , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transforming Growth Factor beta/metabolism , Wnt Proteins/metabolism
6.
PLoS One ; 8(2): e55530, 2013.
Article in English | MEDLINE | ID: mdl-23424635

ABSTRACT

BACKGROUND: The production of human platelets from embryonic stem cells in a defined culture system is a prerequisite for the generation of platelets for therapeutic use. As an important step towards this goal, we report the differentiation of human embryonic stem cells (hESCs) towards the megakaryocyte (Mk) lineage using a 'spin embryoid body' method in serum-free differentiation medium. METHODOLOGY AND PRINCIPAL FINDINGS: Immunophenotypic analyses of differentiating hESC identified a subpopulation of cells expressing high levels of CD41a that expressed other markers associated with the Mk lineage, including CD110, CD42b and CD61. Differentiated cells were sorted on the basis of their expression of CD41a, CD34 and CD45 and assessed for Mk colony formation, expression of myeloid and Mk genes and ability to endoreplicate DNA. In a collagen-based colony assay, the CD41a⁺ cells sorted from these differentiation cultures produced 100-800 Mk progenitors at day 13 and 25-160 Mk progenitors at day 20 of differentiation per 100,000 cells assayed. Differentiated Mk cells produced platelet-like particles which expressed CD42b and were activated by ADP, similar to platelets generated from precursors in cord blood. These studies were complemented by real time PCR analyses showing that subsets of cells enriched for CD41a⁺ Mk precursors expressed high levels of Mk associated genes such as PF4 and MPL. Conversely, high levels of myeloid and erythroid related transcripts, such as GATA1, TAL1/SCL and PU.1, were detected in sorted fractions containing CD34⁺ and CD45⁺ cells. CONCLUSIONS: We describe a serum- and feeder-free culture system that enabled the generation of Mk progenitors from human embryonic stem cells. These cells formed colonies that included differentiated Mks that fragmented to form platelet-like particles. This protocol represents an important step towards the generation of human platelets for therapeutic use.


Subject(s)
Cell Culture Techniques/methods , Embryonic Stem Cells/cytology , Megakaryocytes/cytology , Adenosine Diphosphate/pharmacology , Antigens, CD/metabolism , Blood Platelets/metabolism , Cell Differentiation/drug effects , Culture Media, Serum-Free , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Feeder Cells , Gene Expression Regulation/drug effects , Humans , Megakaryocytes/drug effects , Platelet Factor 4/genetics , Polyploidy , Receptors, Thrombopoietin/genetics , Time Factors
7.
Nat Protoc ; 3(10): 1550-8, 2008.
Article in English | MEDLINE | ID: mdl-18802436

ABSTRACT

The first step in the generation of genetically tagged human embryonic stem cell (HESC) reporter lines is the isolation of cells that contain a stably integrated copy of the reporter vector. These cells are identified by their continued growth in the presence of a specific selective agent, usually conferred by a cassette encoding antibiotic resistance. In order to mitigate potential interference between the regulatory elements driving expression of the antibiotic resistance gene and those controlling the reporter gene, it is advisable to remove the positive selection cassette once the desired clones have been identified. This report describes a protocol for the removal of loxP-flanked selection cassettes from genetically modified HESCs by transient transfection with a vector expressing Cre recombinase. An integrated procedure for the clonal isolation of these genetically modified lines using single-cell deposition flow cytometry is also detailed. When performed sequentially, these protocols take approximately 1 month.


Subject(s)
Drug Resistance, Microbial/genetics , Embryonic Stem Cells/metabolism , Flow Cytometry/methods , Gene Transfer Techniques , Mutagenesis, Insertional/methods , Genetic Vectors/genetics , Humans
8.
Nat Protoc ; 2(4): 792-6, 2007.
Article in English | MEDLINE | ID: mdl-17446878

ABSTRACT

The ability to genetically modify human embryonic stem cells (HESCs) will be critical for their widespread use as a tool for understanding fundamental aspects of human biology and pathology and for their development as a platform for pharmaceutical discovery. Here, we describe a method for the genetic modification of HESCs using electroporation, the preferred method for introduction of DNA into cells in which the desired outcome is gene targeting. This report provides methods for cell amplification, electroporation, colony selection and screening. The protocol we describe has been tested on four different HESC lines, and takes approximately 4 weeks from electroporation to PCR screening of G418-resistant clones.


Subject(s)
Electroporation/methods , Embryonic Stem Cells/cytology , Cell Line , Gene Targeting/methods , Genetic Engineering/methods , Genetic Vectors , Humans
9.
Dev Biol ; 293(1): 90-103, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16546155

ABSTRACT

We analyzed the transcriptional profiles of differentiating mouse embryonic stem cells (mESCs) and show that embryoid bodies (EBs) sequentially expressed genes associated with the epiblast, primitive streak, mesoderm and endoderm of the developing embryo, validating ESCs as a model system for identifying cohorts of genes marking specific stages of embryogenesis. By comparing the transcriptional profiles of undifferentiated ESCs to those of their differentiated progeny, we identified 503 mESC and 983 hESC genes selectively expressed in undifferentiated ES cells. Over 75% of the mESC genes were expressed in hESC and vice versa, attesting to the underlying similarity of mESCs and hESCs. The expression of a cohort of 68 genes decreased greater than 2-fold during differentiation in both mESCs and hESCs. As well as containing many validated ESC genes such as Oct4 [Pou5f1], Nanog and Nodal, this cohort included an uncharacterised gene (FLJ30046), which we designated SLAIN1/Slain1. Slain1 was expressed at the stem cell and epiblast stages of ESC differentiation and in the epiblast, nervous system, tailbud and somites of the developing mouse embryo. SLAIN1 and its more widely expressed homologue SLAIN2 comprise a new family of structurally unique genes conserved throughout vertebrate evolution.


Subject(s)
Cell Differentiation/genetics , Gene Expression Profiling , Proteins/genetics , Stem Cells/metabolism , Amino Acid Sequence , Animals , Cell Line , Embryo, Mammalian , Gene Expression Regulation, Developmental , Humans , Mice , Molecular Sequence Data , Proteins/metabolism , Stem Cells/cytology
10.
Development ; 132(5): 873-84, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15673572

ABSTRACT

The homeobox gene Mixl1 is expressed in the primitive streak of the gastrulating embryo, and marks cells destined to form mesoderm and endoderm. The role of Mixl1 in development of haematopoietic mesoderm was investigated by analysing the differentiation of ES cells in which GFP was targeted to one (Mixl1(GFP/w)) or both (Mixl1(GFP/GFP)) alleles of the Mixl1 locus. In either case, GFP was transiently expressed, with over 80% of cells in day 4 embryoid bodies (EBs) being GFP(+). Up to 45% of Mixl1(GFP/w) day 4 EB cells co-expressed GFP and the haemangioblast marker FLK1, and this doubly-positive population was enriched for blast colony forming cells (BL-CFCs). Mixl1-null ES cells, however, displayed a haematopoietic defect characterised by reduced and delayed Flk1 expression and a decrease in the frequency of haematopoietic CFCs. These data indicated that Mixl1 was required for efficient differentiation of cells from the primitive streak stage to blood. Differentiation of ES cells under serum-free conditions demonstrated that induction of Mixl1- and Flk1-expressing haematopoietic mesoderm required medium supplemented with BMP4 or activin A. In conclusion, this study has revealed an important role for Mixl1 in haematopoietic development and demonstrates the utility of the Mixl1(GFP/w) ES cells for evaluating growth factors influencing mesendodermal differentiation.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Embryo, Mammalian/cytology , Hematopoiesis/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Mesoderm/metabolism , Stem Cells/cytology , Activins/metabolism , Alleles , Animals , Body Patterning , Bone Morphogenetic Protein 4 , Cadherins/metabolism , Cell Differentiation , Cell Proliferation , Culture Media, Serum-Free/pharmacology , DNA Primers/chemistry , Flow Cytometry , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/metabolism , Heterozygote , Inhibin-beta Subunits/metabolism , Mice , Mice, Knockout , Microscopy, Fluorescence , RNA/metabolism , Recombination, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
11.
Blood ; 106(5): 1601-3, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-15914555

ABSTRACT

To realize the therapeutic potential of human embryonic stem cells (hESCs), it is necessary to regulate their differentiation in a uniform and reproducible manner. We have developed a method in which known numbers of hESCs in serum-free medium were aggregated by centrifugation to foster the formation of embryoid bodies (EBs) of uniform size (spin EBs). These spin EBs differentiated efficiently and synchronously, as evidenced by the sequential expression of molecular markers representing stem cells, primitive streak, and mesoderm. In the presence of hematopoietic growth factors, reproducible differentiation was achieved with blood cells formed in more than 90% of EBs. Using chimeric EBs generated from mixtures of green fluorescence protein-positive (GFP(+)) and GFP(-) hESCs in a clonogenic assay, hematopoietic precursor frequency was estimated to be approximately 1:500 input cells. This method of EB formation provides a generally applicable means for modulating and objectively monitoring the directed differentiation of hESCs.


Subject(s)
Cell Differentiation/physiology , Embryo, Mammalian/cytology , Hematopoietic Stem Cells/cytology , Stem Cell Transplantation , Stem Cells/cytology , Cells, Cultured , Centrifugation/methods , Culture Media, Serum-Free , Embryo, Mammalian/physiology , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cells/physiology , Humans , Stem Cells/physiology
12.
J Gen Virol ; 85(Pt 12): 3627-3636, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15557235

ABSTRACT

The mature flavivirus particle comprises a nucleocapsid core surrounded by a lipid bilayer containing the membrane (M) (derived from the precursor prM) and envelope (E) proteins. The formation of intracellular prM/E heterodimers occurs rapidly after translation and is believed to be important for the assembly and secretion of immature virus particles. In this study, the role of the His residue at position 39 in the M protein (M39) of dengue virus type 2 (DENV-2) in the virus life cycle was investigated. Mutations encoding basic (Arg), non-polar (Leu and Pro) and uncharged polar (Asn, Gln and Tyr) amino acids at M39 were introduced into a DENV-2 genomic-length cDNA clone and their effects on virus replication were examined. Substitution of the His residue with non-polar amino acids abolished virus replication, whereas substitution with basic or uncharged polar amino acids decreased virus replication moderately ( approximately 2 log(10) p.f.u. ml(-1) decrease in viral titre for Arg and Asn) or severely (>3.5 log(10) p.f.u. ml(-1) decrease in viral titre for Gln and Tyr). Selected mutations were introduced into a prM-E gene cassette and expressed transiently in COS cells to investigate whether the mutations impaired prM/E association or secretion. None of the mutations was found to disrupt the formation of intracellular prM/E heterodimers. However, the mutations that abolished virus replication prevented secretion of prM/E complexes. The results of this study pinpoint a critical residue in the M protein that potentially plays a role in viral morphogenesis, secretion and entry.


Subject(s)
Dengue Virus/physiology , Viral Envelope Proteins/physiology , Virus Assembly , Animals , COS Cells , Cricetinae , Histidine
SELECTION OF CITATIONS
SEARCH DETAIL