Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
Add more filters

Publication year range
1.
Development ; 150(14)2023 07 15.
Article in English | MEDLINE | ID: mdl-37390294

ABSTRACT

Caudal developmental defects, including caudal regression, caudal dysgenesis and sirenomelia, are devastating conditions affecting the skeletal, nervous, digestive, reproductive and excretory systems. Defects in mesodermal migration and blood supply to the caudal region have been identified as possible causes of caudal developmental defects, but neither satisfactorily explains the structural malformations in all three germ layers. Here, we describe caudal developmental defects in transmembrane protein 132a (Tmem132a) mutant mice, including skeletal, posterior neural tube closure, genitourinary tract and hindgut defects. We show that, in Tmem132a mutant embryos, visceral endoderm fails to be excluded from the medial region of early hindgut, leading directly to the loss or malformation of cloaca-derived genitourinary and gastrointestinal structures, and indirectly to the neural tube and kidney/ureter defects. We find that TMEM132A mediates intercellular interaction, and physically interacts with planar cell polarity (PCP) regulators CELSR1 and FZD6. Genetically, Tmem132a regulates neural tube closure synergistically with another PCP regulator Vangl2. In summary, we have identified Tmem132a as a new regulator of PCP, and hindgut malformation as the underlying cause of developmental defects in multiple caudal structures.


Subject(s)
Neural Tube Defects , Mice , Animals , Neural Tube Defects/metabolism , Neural Tube/metabolism , Neurulation , Germ Layers/metabolism , Cell Polarity/physiology , Membrane Proteins/genetics , Membrane Proteins/metabolism
2.
Development ; 149(20)2022 10 15.
Article in English | MEDLINE | ID: mdl-35899604

ABSTRACT

Directed differentiation of pluripotent stem cells (PSCs) is a powerful model system for deconstructing embryonic development. Although mice are the most advanced mammalian model system for genetic studies of embryonic development, state-of-the-art protocols for directed differentiation of mouse PSCs into defined lineages require additional steps and generates target cell types with lower purity than analogous protocols for human PSCs, limiting their application as models for mechanistic studies of development. Here, we examine the potential of mouse epiblast stem cells cultured in media containing Wnt pathway inhibitors as a starting point for directed differentiation. As a proof of concept, we focused our efforts on two specific cell/tissue types that have proven difficult to generate efficiently and reproducibly from mouse embryonic stem cells: definitive endoderm and neural organoids. We present new protocols for rapid generation of nearly pure definitive endoderm and forebrain-patterned neural organoids that model the development of prethalamic and hippocampal neurons. These differentiation models present new possibilities for combining mouse genetic tools with in vitro differentiation to characterize molecular and cellular mechanisms of embryonic development.


Subject(s)
Endoderm , Pluripotent Stem Cells , Animals , Cell Differentiation/physiology , Endoderm/metabolism , Female , Germ Layers , Humans , Mammals , Mice , Organoids , Pregnancy , Prosencephalon
3.
FASEB J ; 38(4): e23463, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38334393

ABSTRACT

With self-renewal and pluripotency features, embryonic stem cells (ESCs) provide an invaluable tool to investigate early cell fate decisions. Pluripotency exit and lineage commitment depend on precise regulation of gene expression that requires coordination between transcription (TF) and chromatin factors in response to various signaling pathways. SET domain-containing 3 (SETD3) is a methyltransferase that can modify histones in the nucleus and actin in the cytoplasm. Through an shRNA screen, we previously identified SETD3 as an important factor in the meso/endodermal lineage commitment of mouse ESCs (mESC). In this study, we identified SETD3-dependent transcriptomic changes during endoderm differentiation of mESCs using time-course RNA-seq analysis. We found that SETD3 is involved in the timely activation of the endoderm-related gene network. The canonical Wnt signaling pathway was one of the markedly altered signaling pathways in the absence of SETD3. The assessment of Wnt transcriptional activity revealed a significant reduction in Setd3-deleted (setd3∆) mESCs coincident with a decrease in the nuclear pool of the key TF ß-catenin level, though no change was observed in its mRNA or total protein level. Furthermore, a proximity ligation assay (PLA) found an interaction between SETD3 and ß-catenin. We were able to rescue the differentiation defect by stably re-expressing SETD3 or activating the canonical Wnt signaling pathway by changing mESC culture conditions. Our results suggest that alterations in the canonical Wnt pathway activity and subcellular localization of ß-catenin might contribute to the endoderm differentiation defect of setd3∆ mESCs.


Subject(s)
Mouse Embryonic Stem Cells , beta Catenin , Animals , Mice , beta Catenin/metabolism , Cell Differentiation/genetics , Endoderm , Wnt Signaling Pathway/physiology
4.
Development ; 148(1)2021 01 07.
Article in English | MEDLINE | ID: mdl-33199445

ABSTRACT

Anterior mesoderm (AM) and definitive endoderm (DE) progenitors represent the earliest embryonic cell types that are specified during germ layer formation at the primitive streak (PS) of the mouse embryo. Genetic experiments indicate that both lineages segregate from Eomes-expressing progenitors in response to different Nodal signaling levels. However, the precise spatiotemporal pattern of the emergence of these cell types and molecular details of lineage segregation remain unexplored. We combined genetic fate labeling and imaging approaches with single-cell RNA sequencing (scRNA-seq) to follow the transcriptional identities and define lineage trajectories of Eomes-dependent cell types. Accordingly, all cells moving through the PS during the first day of gastrulation express Eomes AM and DE specification occurs before cells leave the PS from Eomes-positive progenitors in a distinct spatiotemporal pattern. ScRNA-seq analysis further suggested the immediate and complete separation of AM and DE lineages from Eomes-expressing cells as last common bipotential progenitor.


Subject(s)
Cell Lineage , Endoderm/cytology , Endoderm/metabolism , Gastrulation , Mesoderm/cytology , Mesoderm/metabolism , Alleles , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Germ Layers/cytology , Mice , Models, Biological , Primitive Streak/embryology , Primitive Streak/metabolism , Stem Cells/metabolism , T-Box Domain Proteins/metabolism , Time Factors , Transcription, Genetic
5.
Cell Commun Signal ; 22(1): 300, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816763

ABSTRACT

Optimizing the efficiency of definitive endoderm (DE) differentiation is necessary for the generation of diverse organ-like structures. In this study, we used the small molecule inhibitor saracatinib (SAR) to enhance DE differentiation of human embryonic stem cells and induced pluripotent stem cells. SAR significantly improved DE differentiation efficiency at low concentrations. The interaction between SAR and Focal Adhesion Kinase (FAK) was explored through RNA-seq and molecular docking simulations, which further supported the inhibition of DE differentiation by p-FAK overexpression in SAR-treated cells. In addition, we found that SAR inhibited the nuclear translocation of Yes-associated protein (YAP), a downstream effector of FAK, which promoted DE differentiation. Moreover, the addition of SAR enabled a significant reduction in activin A (AA) from 50 to 10 ng/mL without compromising DE differentiation efficiency. For induction of the pancreatic lineage, 10 ng/ml AA combined with SAR at the DE differentiation stage yielded a comparative number of PDX1+/NKX6.1+ pancreatic progenitor cells to those obtained by 50 ng/ml AA treatment. Our study highlights SAR as a potential modulator that facilitates the cost-effective generation of DE cells and provides insight into the orchestration of cell fate determination.


Subject(s)
Benzodioxoles , Cell Differentiation , Endoderm , Quinazolines , Signal Transduction , Humans , Cell Differentiation/drug effects , Endoderm/drug effects , Endoderm/cytology , Endoderm/metabolism , Benzodioxoles/pharmacology , Signal Transduction/drug effects , Quinazolines/pharmacology , Transcription Factors/metabolism , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Adaptor Proteins, Signal Transducing/metabolism , YAP-Signaling Proteins/metabolism , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Activins/metabolism , Molecular Docking Simulation
6.
Cell Biol Int ; 48(6): 835-847, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38419492

ABSTRACT

Pluripotent stem cells (PSCs) hold enormous potential for treating multiple diseases owing to their ability to self-renew and differentiate into any cell type. Albeit possessing such promising potential, controlling their differentiation into a desired cell type continues to be a challenge. Recent studies suggest that PSCs respond to different substrate stiffness and, therefore, can differentiate towards some lineages via Hippo pathway. Human PSCs can also differentiate and self-organize into functional cells, such as organoids. Traditionally, human PSCs are differentiated on stiff plastic or glass plates towards definitive endoderm and then into functional pancreatic progenitor cells in the presence of soluble growth factors. Thus, whether stiffness plays any role in differentiation towards definitive endoderm from human pluripotent stem cells (hPSCs) remains unclear. Our study found that the directed differentiation of human embryonic stem cells towards endodermal lineage on the varying stiffness did not differ from the differentiation on stiff plastic dishes. We also observed no statistical difference between the expression of yes-associated protein (YAP) and phosphorylated YAP. Furthermore, we demonstrate that lysophosphatidic acid, a YAP activator, enhanced definitive endoderm formation, whereas verteporfin, a YAP inhibitor, did not have the significant effect on the differentiation. In summary, our results suggest that human embryonic stem cells may not differentiate in response to changes in stiffness, and that such cues may not have as significant impact on the level of YAP. Our findings indicate that more research is needed to understand the direct relationship between biophysical forces and hPSCs differentiation.


Subject(s)
Cell Differentiation , Cell Lineage , Endoderm , Human Embryonic Stem Cells , Humans , Cell Differentiation/physiology , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Endoderm/cytology , Endoderm/metabolism , YAP-Signaling Proteins/metabolism , Transcription Factors/metabolism
7.
Arch Toxicol ; 97(10): 2625-2641, 2023 10.
Article in English | MEDLINE | ID: mdl-37612375

ABSTRACT

Fetal development is one of the most sensitive windows to methylmercury (MeHg) toxicity. Laboratory and epidemiological studies have shown a dose-response relationship between fetal MeHg exposure and neuro performance in different life stages from infants to adults. In addition, MeHg exposure has been reported to be associated with disorders in endoderm-derived organs, such as morphological changes in liver cells and pancreatic cell dysfunctions. However, the mechanisms of the effects of MeHg on non-neuronal organs or systems, especially during the early development of endoderm-derived organs, remain unclear. Here we determined the effects of low concentrations of MeHg exposure during the differentiation of definitive endoderm (DE) cells from human embryonic stem cells (hESCs). hESCs were exposed to MeHg (0, 10, 100, and 200 nM) that covers the range of Hg concentrations typically found in human maternal blood during DE cell induction. Transcriptomic analysis showed that sub-lethal doses of MeHg exposure could alter global gene expression patterns during hESC to DE cell differentiation, leading to increased expression of endodermal genes/proteins and the over-promotion of endodermal fate, mainly through disrupting calcium homeostasis and generating ROS. Bioinformatic analysis results suggested that MeHg exerts its developmental toxicity mainly by disrupting ribosome biogenesis during early cell lineage differentiation. This disruption could lead to aberrant growth or dysfunctions of the developing endoderm-derived organs, and it may be the underlying mechanism for the observed congenital diseases later in life. Based on the results, we proposed an adverse outcome pathway for the effects of MeHg exposure during human embryonic stem cells to definitive endoderm differentiation.


Subject(s)
Adverse Outcome Pathways , Human Embryonic Stem Cells , Methylmercury Compounds , Adult , Infant , Humans , Methylmercury Compounds/toxicity , Endoderm , Cell Differentiation
8.
Genes Dev ; 29(20): 2108-22, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26494787

ABSTRACT

Gene regulatory networks controlling functional activities of spatially and temporally distinct endodermal cell populations in the early mouse embryo remain ill defined. The T-box transcription factor Eomes, acting downstream from Nodal/Smad signals, directly activates the LIM domain homeobox transcription factor Lhx1 in the visceral endoderm. Here we demonstrate Smad4/Eomes-dependent Lhx1 expression in the epiblast marks the entire definitive endoderm lineage, the anterior mesendoderm, and midline progenitors. Conditional inactivation of Lhx1 disrupts anterior definitive endoderm development and impedes node and midline morphogenesis in part due to severe disturbances in visceral endoderm displacement. Transcriptional profiling and ChIP-seq (chromatin immunoprecipitation [ChIP] followed by high-throughput sequencing) experiments identified Lhx1 target genes, including numerous anterior definitive endoderm markers and components of the Wnt signaling pathway. Interestingly, Lhx1-binding sites were enriched at enhancers, including the Nodal-proximal epiblast enhancer element and enhancer regions controlling Otx2 and Foxa2 expression. Moreover, in proteomic experiments, we characterized a complex comprised of Lhx1, Otx2, and Foxa2 as well as the chromatin-looping protein Ldb1. These partnerships cooperatively regulate development of the anterior mesendoderm, node, and midline cell populations responsible for establishment of the left-right body axis and head formation.


Subject(s)
DNA-Binding Proteins/metabolism , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Germ Layers/embryology , DNA-Binding Proteins/genetics , Embryo, Mammalian , Enhancer Elements, Genetic/physiology , Gene Deletion , Gene Expression Profiling , Germ Layers/metabolism , Hepatocyte Nuclear Factor 3-beta/metabolism , LIM Domain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Otx Transcription Factors/metabolism , Protein Binding , Transcription Factors/genetics , Transcription Factors/metabolism , Wnt Signaling Pathway
9.
Development ; 145(19)2018 10 11.
Article in English | MEDLINE | ID: mdl-30232173

ABSTRACT

The definitive endoderm (DE) is the embryonic germ layer that forms the gut tube and associated organs, including thymus, lungs, liver and pancreas. To understand how individual DE cells furnish gut organs, genetic fate mapping was performed using the Rosa26lacZ Cre-reporter paired with a tamoxifen-inducible DE-specific Cre-expressing transgene. We established a low tamoxifen dose that infrequently induced heritable lacZ expression in a single cell of individual E8.5 mouse embryos and identified clonal cell descendants at E16.5. As expected, only a fraction of the E16.5 embryos contained lacZ-positive clonal descendants and a subset of these contained descendants in multiple organs, revealing novel ontogeny. Furthermore, immunohistochemical analysis was used to identify lacZ-positive hepatocytes and biliary epithelial cells, which are the cholangiocyte precursors, in each clonally populated liver. Together, these data not only uncover novel and suspected lineage relationships between DE-derived organs, but also illustrate the bipotential nature of individual hepatoblasts by demonstrating that single hepatoblasts contribute to both the hepatocyte and the cholangiocyte lineage in vivo.


Subject(s)
Chromosome Mapping , Endoderm/cytology , Hepatocytes/cytology , Organ Specificity/genetics , Single-Cell Analysis , Stem Cells/cytology , Animals , Bile Ducts/cytology , Cell Lineage , Clone Cells , Embryo, Mammalian/cytology , Epithelial Cells/cytology , Female , Hepatocytes/metabolism , Male , Mice, Inbred C57BL , Stem Cells/metabolism
10.
Biotechnol Bioeng ; 118(2): 979-991, 2021 02.
Article in English | MEDLINE | ID: mdl-33205831

ABSTRACT

Scalable processes are requisite for the robust biomanufacturing of human pluripotent stem cell (hPSC)-derived therapeutics. Toward this end, we demonstrate the xeno-free expansion and directed differentiation of human embryonic and induced pluripotent stem cells to definitive endoderm (DE) in a controlled stirred suspension bioreactor (SSB). Based on previous work on converting hPSCs to insulin-producing progeny, differentiation of two hPSC lines was optimized in planar cultures yielding up to 87% FOXA2+ /SOX17+ cells. Next, hPSCs were propagated in an SSB with controlled pH and dissolved oxygen. Cultures displayed a 10- to 12-fold increase in cell number over 5-6 days with the maintenance of pluripotency (>85% OCT4+ ) and viability (>85%). For differentiation, SSB cultures yielded up to 89% FOXA2+ /SOX17+ cells or ~ 8 DE cells per seeded hPSC. Specification to DE cell fate was consistently more efficient in the bioreactor compared to planar cultures. Hence, a tunable strategy is established that is suitable for the xeno-free manufacturing of DE cells from different hPSC lines in scalable SSBs. This study advances bioprocess development for producing a wide gamut of human DE cell-derived therapeutics.


Subject(s)
Bioreactors , Endoderm/metabolism , Human Embryonic Stem Cells/metabolism , Cell Line , Endoderm/cytology , Human Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology
11.
Int J Mol Sci ; 22(21)2021 Nov 08.
Article in English | MEDLINE | ID: mdl-34769484

ABSTRACT

Multiple endocrine neoplasia type 1 (MEN1) is a rare tumor syndrome that manifests differently among various patients. Despite the mutations in the MEN1 gene that commonly predispose tumor development, there are no obvious phenotype-genotype correlations. The existing animal and in vitro models do not allow for studies of the molecular genetics of the disease in a human-specific context. We aimed to create a new human cell-based model, which would consider the variability in genetic or environmental factors that cause the complexity of MEN1 syndrome. Here, we generated patient-specific induced pluripotent stem cell lines carrying the mutation c.1252G>T, D418Y in the MEN1 gene. To reduce the genetically determined variability of the existing cellular models, we created an isogenic cell system by modifying the target allele through CRISPR/Cas9 editing with great specificity and efficiency. The high potential of these cell lines to differentiate into the endodermal lineage in defined conditions ensures the next steps in the development of more specialized cells that are commonly affected in MEN1 patients, such as parathyroid or pancreatic islet cells. We anticipate that this isogenic system will be broadly useful to comprehensively study MEN1 gene function across different contexts, including in vitro modeling of MEN1 syndrome.


Subject(s)
Fibroblasts/metabolism , Gene Editing/methods , Induced Pluripotent Stem Cells/metabolism , Multiple Endocrine Neoplasia Type 1/pathology , Mutation , Proto-Oncogene Proteins/antagonists & inhibitors , Adult , CRISPR-Cas Systems , Cells, Cultured , Female , Humans , Multiple Endocrine Neoplasia Type 1/genetics , Multiple Endocrine Neoplasia Type 1/metabolism , Proto-Oncogene Proteins/genetics
12.
Development ; 144(7): 1249-1260, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28174238

ABSTRACT

The T-box transcription factor (TF) Eomes is a key regulator of cell fate decisions during early mouse development. The cis-acting regulatory elements that direct expression in the anterior visceral endoderm (AVE), primitive streak (PS) and definitive endoderm (DE) have yet to be defined. Here, we identified three gene-proximal enhancer-like sequences (PSE_a, PSE_b and VPE) that faithfully activate tissue-specific expression in transgenic embryos. However, targeted deletion experiments demonstrate that PSE_a and PSE_b are dispensable, and only VPE is required for optimal Eomes expression in vivo Embryos lacking this enhancer display variably penetrant defects in anterior-posterior axis orientation and DE formation. Chromosome conformation capture experiments reveal VPE-promoter interactions in embryonic stem cells (ESCs), prior to gene activation. The locus resides in a large (500 kb) pre-formed compartment in ESCs and activation during DE differentiation occurs in the absence of 3D structural changes. ATAC-seq analysis reveals that VPE, PSE_a and four additional putative enhancers display increased chromatin accessibility in DE that is associated with Smad2/3 binding coincident with transcriptional activation. By contrast, activation of the Eomes target genes Foxa2 and Lhx1 is associated with higher order chromatin reorganisation. Thus, diverse regulatory mechanisms govern activation of lineage specifying TFs during early development.


Subject(s)
Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Regulatory Sequences, Nucleic Acid/genetics , T-Box Domain Proteins/genetics , Animals , Cell Differentiation/genetics , Chromatin/metabolism , Endoderm/metabolism , Enhancer Elements, Genetic , Female , Forkhead Transcription Factors/metabolism , Gastrulation/genetics , Gene Deletion , Gene Targeting , Genes, Reporter , Genotype , Mice, Inbred C57BL , Models, Biological , Polycomb-Group Proteins/metabolism , Signal Transduction/genetics , Smad2 Protein/metabolism , T-Box Domain Proteins/metabolism
13.
Int J Mol Sci ; 21(23)2020 Nov 28.
Article in English | MEDLINE | ID: mdl-33260776

ABSTRACT

Aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor, which mediates the effects of a variety of environmental stimuli in multiple tissues. Recent advances in AHR biology have underlined its importance in cells with high developmental potency, including pluripotent stem cells. Nonetheless, there is little data on AHR expression and its role during the initial stages of stem cell differentiation. The purpose of this study was to investigate the temporal pattern of AHR expression during directed differentiation of human embryonic stem cells (hESC) into neural progenitor, early mesoderm and definitive endoderm cells. Additionally, we investigated the effect of the AHR agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on the gene expression profile in hESCs and differentiated cells by RNA-seq, accompanied by identification of AHR binding sites by ChIP-seq and epigenetic landscape analysis by ATAC-seq. We showed that AHR is differentially regulated in distinct lineages. We provided evidence that TCDD alters gene expression patterns in hESCs and during early differentiation. Additionally, we identified novel potential AHR target genes, which expand our understanding on the role of this protein in different cell types.


Subject(s)
Cell Differentiation/drug effects , Human Embryonic Stem Cells/cytology , Polychlorinated Dibenzodioxins/toxicity , Receptors, Aryl Hydrocarbon/metabolism , Biomarkers , Cell Differentiation/genetics , Cell Line , Cell Lineage/drug effects , Cell Lineage/genetics , Down-Regulation/drug effects , Down-Regulation/genetics , Embryoid Bodies/cytology , Embryoid Bodies/drug effects , Genome, Human , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/metabolism , Humans , Ligands
14.
J Mol Cell Cardiol ; 137: 132-142, 2019 12.
Article in English | MEDLINE | ID: mdl-31668971

ABSTRACT

Specification of the primary heart field in mouse embryos requires signaling from the anterior visceral endoderm (AVE). The nature of these signals is not known. We hypothesized that the TGFß-activated kinase (TAK1/Map3k7) may act as a cardiogenic factor, based on its expression in heart-inducing endoderm and its requirement for cardiac differentiation of p19 cells. To test this, mouse embryonic stem (ES) cells overexpressing Map3k7 were isolated and differentiated as embryoid bodies (EBs). Map3k7-overexpressing EBs showed increased expression of AVE markers but interestingly, showed little effect on mesoderm formation and had no impact on overall cardiomyocyte formation. To test whether the pronounced expansion of endoderm masks an expansion of cardiac lineages, chimeric EBs were made consisting of Map3k7-overexpressing ES and wild type ES cells harboring a cardiac reporter transgene, MHCα::GFP, allowing cardiac differentiation to be assessed specifically in wild type ES cells. Wild type ES cells co-cultured with Map3k7-overexpressing cells had a 4-fold increase in expression of the cardiac reporter, supporting the hypothesis that Map3k7 increases the formation of cardiogenic endoderm. To further examine the role of Map3k7 in early lineage specification, other endodermal markers were examined. Interestingly, markers that are expressed in both the VE and later in gut development were expanded, whereas transcripts that specifically mark the early definitive (streak-derived) endoderm (DE) were not. To determine if Map3k7 is necessary for endoderm differentiation, EBs were grown in the presence of the Map3k7 specific inhibitor 5Z-7-oxozeaenol. Endoderm differentiation was dramatically decreased in these cells. Western blot analysis showed that known downstream targets of Map3k7 (Jnk, Nemo-like kinase (NLK) and p38 MAPK) were all inhibited. By contrast, transcripts for another TGFß target, Sonic Hedgehog (Shh) were markedly upregulated, as were transcripts for Gli2 (but not Gli1 and Gli3). Together these data support the hypothesis that Map3k7 governs the formation, or proliferation of cardiogenic endoderm.


Subject(s)
Cell Differentiation , Endoderm/embryology , Endoderm/enzymology , Heart/embryology , MAP Kinase Kinase Kinases/metabolism , Mouse Embryonic Stem Cells/cytology , Organogenesis , Animals , Cell Line , Embryoid Bodies/cytology , Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , MAP Kinase Kinase Kinases/genetics , MAP Kinase Signaling System , Mesoderm/embryology , Mice , Myocytes, Cardiac/cytology , Up-Regulation/genetics , Zinc Finger Protein Gli2/metabolism
15.
Int J Mol Sci ; 21(1)2019 Dec 18.
Article in English | MEDLINE | ID: mdl-31861348

ABSTRACT

Endoderm-derived organs as liver and pancreas are potential targets for regenerative therapies, and thus, there is great interest in understanding the pathways that regulate the induction and specification of this germ layer. Currently, the knowledge of molecular mechanisms that guide the in vivo endoderm specification is restricted by the lack of early endoderm specific markers. Nephrocan (Nepn) is a gene whose expression characterizes the early stages of murine endoderm specification (E7.5-11.5) and encodes a secreted N-glycosylated protein. In the present study, we report the identification of a new transcript variant that is generated through alternative splicing. The new variant was found to have differential and tissue specific expression in the adult mouse. In order to better understand Nepn role during endoderm specification, we generated Nepn knock-out (KO) mice. Nepn-/- mice were born at Mendelian ratios and displayed no evident phenotype compared to WT mice. In addition, we produced nullizygous mouse embryonic stem cell (mESC) line lacking Nepn by applying (CRISPR)/CRISPR-associated systems 9 (Cas9) and employed a differentiation protocol toward endoderm lineage. Our in vitro results revealed that Nepn loss affects the endoderm differentiation impairing the expression of posterior foregut-associated markers.


Subject(s)
Body Patterning/genetics , Endoderm/embryology , Endoderm/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Animals , Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Editing , Gene Expression Regulation, Developmental , Gene Targeting , Genetic Loci , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Protein Isoforms/genetics
16.
J Biol Chem ; 292(23): 9840-9854, 2017 06 09.
Article in English | MEDLINE | ID: mdl-28298438

ABSTRACT

The mammalian post-implantation embryo has been extensively investigated at the tissue level. However, to unravel the molecular basis for the cell-fate plasticity and determination, it is essential to study the characteristics of individual cells. In particular, the individual definitive endoderm (DE) cells have not been characterized in vivo Here, we report gene expression patterns in single cells freshly isolated from mouse embryos on days 5.5 and 6.5. Initial transcriptome data from 124 single cells yielded signature genes for the epiblast, visceral endoderm, and extra-embryonic ectoderm and revealed a unique distribution pattern of fibroblast growth factor (FGF) ligands and receptors. Further analysis indicated that early-stage epiblast cells do not segregate into lineages of the major germ layers. Instead, some cells began to diverge from epiblast cells, displaying molecular features of the premesendoderm by expressing higher levels of mesendoderm markers and lower levels of Sox3 transcripts. Analysis of single-cell high-throughput quantitative RT-PCR data from 441 cells identified a late stage of the day 6.5 embryo in which mesoderm and DE cells emerge, with many of them coexpressing Oct4 and Gata6 Analysis of single-cell RNA-sequence data from 112 cells of the late-stage day 6.5 embryos revealed differentially expressed signaling genes and networks of transcription factors that might underlie the segregation of the mesoderm and DE lineages. Moreover, we discovered a subpopulation of mesoderm cells that possess molecular features of the extraembryonic mesoderm. This study provides fundamental insight into the molecular basis for lineage segregation in post-implantation mouse embryos.


Subject(s)
Antigens, Differentiation/biosynthesis , Cell Lineage/physiology , Embryo, Mammalian , Embryonic Development/physiology , Gene Expression Regulation, Developmental/physiology , Transcriptome/physiology , Animals , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , GATA6 Transcription Factor/biosynthesis , Mice , Octamer Transcription Factor-3/biosynthesis , SOXB1 Transcription Factors/biosynthesis
17.
J Cell Physiol ; 233(4): 3578-3589, 2018 04.
Article in English | MEDLINE | ID: mdl-29044512

ABSTRACT

Definitive endoderm (DE) is the first stage of human pluripotent stem cell (hPSC) differentiation into hepatocyte-like cells. Developing human liver cell models for pharmaceutical applications is highly demanding. Due to the vast number of existing protocols to generate DE cells from hPSCs, we aimed to compare the specificity and efficiency of selected published differentiation conditions. We differentiated two hPSC lines (induced PSC and embryonic stem cell) to DE cells on Matrigel matrix using growth factors (Activin A and Wnt-3a) and small molecules (sodium butyrate and IDE 1) in different combinations. By studying dynamic changes during 6 days in cell morphology and the expression of markers for pluripotency, DE, and other germ layer lineages, we found that Activin A is essential for DE differentiation, while Wnt-3a and sodium butyrate are dispensable. Although sodium butyrate exerted rapid DE differentiation kinetics, it caused massive cell death and could not generate sufficient cells for further differentiation and applications. We further discover that IDE 1 could not induce DE as reported previously. Hereby, we compared different conditions for DE induction and found an effective six day-protocol to obtain DE cells for the further differentiation and applications.


Subject(s)
Activins/pharmacology , Butyric Acid/pharmacology , Embryonic Stem Cells/drug effects , Endoderm/drug effects , Hepatocytes/drug effects , Cell Differentiation/drug effects , Cell Line , Cells, Cultured , Embryonic Stem Cells/cytology , Endoderm/cytology , Hepatocytes/metabolism , Humans , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects
18.
Am J Physiol Cell Physiol ; 312(5): C573-C582, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28298334

ABSTRACT

Artificially generated pancreatic ß-cells from pluripotent stem cells are expected for cell replacement therapy for type 1 diabetes. Several strategies are adopted to direct pluripotent stem cells toward pancreatic differentiation. However, a standard differentiation method for clinical application has not been established. It is important to develop more effective and safer methods for generating pancreatic ß-cells without toxic or mutagenic chemicals. In the present study, we screened several endogenous factors involved in organ development to identify the factor, which induced the efficiency of pancreatic differentiation and found that treatment with erythropoietin (EPO) facilitated the differentiation of mouse embryonic stem cells (ESCs) into definitive endoderm. At an early stage of differentiation, EPO treatment significantly increased Sox17 gene expression, as a marker of the definitive endoderm. Contrary to the canonical function of EPO, it did not affect the levels of phosphorylated JAK2 and STAT5, but stimulated the phosphorylation of ERK1/2 and Akt. The MEK inhibitor U0126 significantly inhibited EPO-induced Sox17 expression. The differentiation of ESCs into definitive endoderm is an important step for the differentiation into pancreatic and other endodermal lineages. This study suggests a possible role of EPO in embryonic endodermal development and a new agent for directing the differentiation into endodermal lineages like pancreatic ß-cells.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Endoderm/cytology , Erythropoietin/metabolism , Insulin-Secreting Cells/cytology , MAP Kinase Signaling System/physiology , Animals , Cell Differentiation/physiology , Cell Line , Endoderm/physiology , Gene Expression Regulation, Developmental/physiology , Insulin-Secreting Cells/physiology , Mice
19.
Development ; 141(19): 3772-81, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25209250

ABSTRACT

Although many regulatory networks involved in defining definitive endoderm have been identified, the mechanisms through which these networks interact to pattern the endoderm are less well understood. To explore the mechanisms involved in midgut patterning, we dissected the transcriptional regulatory elements of nephrocan (Nepn), the earliest known midgut specific gene in mice. We observed that Nepn expression is dramatically reduced in Sox17(-/-) and Raldh2(-/-) embryos compared with wild-type embryos. We further show that Nepn is directly regulated by Sox17 and the retinoic acid (RA) receptor via two enhancer elements located upstream of the gene. Moreover, Nepn expression is modulated by Activin signaling, with high levels inhibiting and low levels enhancing RA-dependent expression. In Foxh1(-/-) embryos in which Nodal signaling is reduced, the Nepn expression domain is expanded into the anterior gut region, confirming that Nodal signaling can modulate its expression in vivo. Together, Sox17 is required for Nepn expression in the definitive endoderm, while RA signaling restricts expression to the midgut region. A balance of Nodal/Activin signaling regulates the anterior boundary of the midgut expression domain.


Subject(s)
Body Patterning/physiology , Endoderm/physiology , Gastrointestinal Tract/embryology , Gene Expression Regulation, Developmental/physiology , Gene Regulatory Networks/physiology , Glycoproteins/metabolism , Signal Transduction/physiology , Activins/metabolism , Aldehyde Oxidoreductases/metabolism , Animals , Electrophoretic Mobility Shift Assay , Gene Regulatory Networks/genetics , Genetic Vectors/genetics , HMGB Proteins/metabolism , Intercellular Signaling Peptides and Proteins , Luciferases , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Receptors, Retinoic Acid/metabolism , SOXF Transcription Factors/metabolism
20.
Cell Tissue Res ; 368(3): 563-578, 2017 06.
Article in English | MEDLINE | ID: mdl-28283910

ABSTRACT

Exposure to maternal diabetes during fetal growth is a risk factor for the development of type II diabetes (T2D) in later life. Discovery of the mechanisms involved in this association should provide valuable background for therapeutic treatments. Early embryogenesis involves epigenetic changes including histone modifications. The bivalent histone methylation marks H3K4me3 and H3K27me3 are important for regulating key developmental genes during early fetal pancreas specification. We hypothesized that maternal hyperglycemia disrupted early pancreas development through changes in histone bivalency. A human embryonic stem cell line (VAL3) was used as the cell model for studying the effects of hyperglycemia upon differentiation into definitive endoderm (DE), an early stage of the pancreatic lineage. Hyperglycemic conditions significantly down-regulated the expression levels of DE markers SOX17, FOXA2, CXCR4 and EOMES during differentiation. This was associated with retention of the repressive histone methylation mark H3K27me3 on their promoters under hyperglycemic conditions. The disruption of histone methylation patterns was observed as early as the mesendoderm stage, with Wnt/ß-catenin signaling being suppressed during hyperglycemia. Treatment with Wnt/ß-catenin signaling activator CHIR-99021 restored the expression levels and chromatin methylation status of DE markers, even in a hyperglycemic environment. The disruption of DE development was also found in mouse embryos at day 7.5 post coitum from diabetic mothers. Furthermore, disruption of DE differentiation in VAL3 cells led to subsequent impairment in pancreatic progenitor formation. Thus, early exposure to hyperglycemic conditions hinders DE development with a possible relationship to the later impairment of pancreas specification.


Subject(s)
Cell Differentiation , Endoderm/cytology , Histones/metabolism , Hyperglycemia/embryology , Pancreas/embryology , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Azacitidine/pharmacology , Cell Line , Cell Lineage , DNA Modification Methylases/antagonists & inhibitors , DNA Modification Methylases/metabolism , Endoderm/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Developmental , Glucose/pharmacology , Humans , Hyperglycemia/metabolism , Male , Mesoderm/metabolism , Methylation , Mice , Mice, Inbred ICR , Pancreas/cytology , Pancreas/metabolism , Promoter Regions, Genetic , Signal Transduction , Wnt Proteins/metabolism , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL