Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
2.
Nat Cell Biol ; 19(11): 1313-1325, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29058721

ABSTRACT

Apicobasal polarity is known to affect epithelial morphogenesis and cell differentiation, but it remains unknown how these processes are mechanistically orchestrated. We find that ligand-specific EGFR signalling via PI(3)K and Rac1 autonomously modulates apicobasal polarity to enforce the sequential control of morphogenesis and cell differentiation. Initially, EGF controls pancreatic tubulogenesis by negatively regulating apical polarity induction. Subsequently, betacellulin, working via inhibition of atypical protein kinase C (aPKC), causes apical domain constriction within neurogenin3+ endocrine progenitors, which results in reduced Notch signalling, increased neurogenin3 expression, and ß-cell differentiation. Notably, the ligand-specific EGFR output is not driven at the ligand level, but seems to have evolved in response to stage-specific epithelial influences. The EGFR-mediated control of ß-cell differentiation via apical polarity is also conserved in human neurogenin3+ cells. We provide insight into how ligand-specific EGFR signalling coordinates epithelial morphogenesis and cell differentiation via apical polarity dynamics.


Subject(s)
Cell Polarity/physiology , ErbB Receptors/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/physiology , Organogenesis/physiology , Signal Transduction/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Epithelial Cells/metabolism , Mice , Mice, Knockout , Morphogenesis/physiology , Nerve Tissue Proteins/metabolism , Neuropeptides/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , rac1 GTP-Binding Protein/metabolism
3.
Elife ; 62017 07 21.
Article in English | MEDLINE | ID: mdl-28731406

ABSTRACT

Information remains scarce on human development compared to animal models. Here, we reconstructed human fetal pancreatic differentiation using cell surface markers. We demonstrate that at 7weeks of development, the glycoprotein 2 (GP2) marks a multipotent cell population that will differentiate into the acinar, ductal or endocrine lineages. Development towards the acinar lineage is paralleled by an increase in GP2 expression. Conversely, a subset of the GP2+ population undergoes endocrine differentiation by down-regulating GP2 and CD142 and turning on NEUROG3, a marker of endocrine differentiation. Endocrine maturation progresses by up-regulating SUSD2 and lowering ECAD levels. Finally, in vitro differentiation of pancreatic endocrine cells derived from human pluripotent stem cells mimics key in vivo events. Our work paves the way to extend our understanding of the origin of mature human pancreatic cell types and how such lineage decisions are regulated.


Subject(s)
Biomarkers/metabolism , Cell Differentiation , Cell Lineage , Fetus/cytology , Gene Expression Regulation, Developmental , Pancreas/cytology , Acinar Cells/cytology , Acinar Cells/metabolism , Cells, Cultured , Endocrine Cells/cytology , Endocrine Cells/metabolism , Female , Fetus/metabolism , Humans , Pancreas/metabolism , Pancreatic Ducts/cytology , Pancreatic Ducts/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Transcriptome
4.
Cell Rep ; 19(1): 36-49, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28380361

ABSTRACT

Stem cell-based therapy for type 1 diabetes would benefit from implementation of a cell purification step at the pancreatic endoderm stage. This would increase the safety of the final cell product, allow the establishment of an intermediate-stage stem cell bank, and provide a means for upscaling ß cell manufacturing. Comparative gene expression analysis revealed glycoprotein 2 (GP2) as a specific cell surface marker for isolating pancreatic endoderm cells (PECs) from differentiated hESCs and human fetal pancreas. Isolated GP2+ PECs efficiently differentiated into glucose responsive insulin-producing cells in vitro. We found that in vitro PEC proliferation declines due to enhanced expression of the cyclin-dependent kinase (CDK) inhibitors CDKN1A and CDKN2A. However, we identified a time window when reducing CDKN1A or CDKN2A expression increased proliferation and yield of GP2+ PECs. Altogether, our results contribute tools and concepts toward the isolation and use of PECs as a source for the safe production of hPSC-derived ß cells.


Subject(s)
Embryonic Stem Cells/metabolism , GPI-Linked Proteins/metabolism , Glucose/metabolism , Insulin-Secreting Cells/cytology , Biomarkers/metabolism , Cell Differentiation , Cell Proliferation , Cell Separation/methods , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinase Inhibitor p18/genetics , Cyclin-Dependent Kinase Inhibitor p18/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Diabetes Mellitus, Type 1/therapy , Embryonic Stem Cells/cytology , Endoderm/cytology , GPI-Linked Proteins/genetics , Gene Expression Regulation , Genome-Wide Association Study , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism
5.
PLoS One ; 5(9)2010 Sep 02.
Article in English | MEDLINE | ID: mdl-20824089

ABSTRACT

BACKGROUND: Pluripotency and self-renewal of human embryonic stem cells (hESCs) is mediated by a complex interplay between extra- and intracellular signaling pathways, which regulate the expression of pluripotency-specific transcription factors. The homeodomain transcription factor NANOG plays a central role in maintaining hESC pluripotency, but the precise role and regulation of NANOG are not well defined. METHODOLOGY/PRINCIPAL FINDINGS: To facilitate the study of NANOG expression and regulation in viable hESC cultures, we generated fluorescent NANOG reporter cell lines by gene targeting in hESCs. In these reporter lines, the fluorescent reporter gene was co-expressed with endogenous NANOG and responded to experimental induction or repression of the NANOG promoter with appropriate changes in expression levels. Furthermore, NANOG reporter lines facilitated the separation of hESC populations based on NANOG expression levels and their subsequent characterization. Gene expression arrays on isolated hESC subpopulations revealed genes with differential expression in NANOG(high) and NANOG(low) hESCs, providing candidates for NANOG downstream targets hESCs. CONCLUSION/SIGNIFICANCE: The newly derived NANOG reporter hESC lines present novel tools to visualize NANOG expression in viable hESCs. In future applications, these reporter lines can be used to elucidate the function and regulation of NANOG in pluripotent hESCs.


Subject(s)
Cell Line/metabolism , Embryonic Stem Cells/metabolism , Gene Targeting , Genes, Reporter , Homeodomain Proteins/genetics , Gene Expression Regulation , Homeodomain Proteins/metabolism , Humans , Nanog Homeobox Protein
6.
Stem Cells ; 28(1): 45-56, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19890880

ABSTRACT

Fibroblast growth factor (FGF) signaling controls axis formation during endoderm development. Studies in lower vertebrates have demonstrated that FGF2 primarily patterns the ventral foregut endoderm into liver and lung, whereas FGF4 exhibits broad anterior-posterior and left-right patterning activities. Furthermore, an inductive role of FGF2 during dorsal pancreas formation has been shown. However, whether FGF2 plays a similar role during human endoderm development remains unknown. Here, we show that FGF2 specifies hESC-derived definitive endoderm (DE) into different foregut lineages in a dosage-dependent manner. Specifically, increasing concentrations of FGF2 inhibits hepatocyte differentiation, whereas intermediate concentration of FGF2 promotes differentiation toward a pancreatic cell fate. At high FGF2 levels specification of midgut endoderm into small intestinal progenitors is increased at the expense of PDX1(+) pancreatic progenitors. High FGF2 concentrations also promote differentiation toward an anterior foregut pulmonary cell fate. Finally, by dissecting the FGF receptor intracellular pathway that regulates pancreas specification, we demonstrate for the first time to the best of our knowledge that induction of PDX1(+) pancreatic progenitors relies on FGF2-mediated activation of the MAPK signaling pathway. Altogether, these observations suggest a broader gut endodermal patterning activity of FGF2 that corresponds to what has previously been advocated for FGF4, implying a functional switch from FGF4 to FGF2 during evolution. Thus, our results provide new knowledge of how cell fate specification of human DE is controlled-facts that will be of great value for future regenerative cell therapies.


Subject(s)
Cell Differentiation , Cell Lineage , Digestive System/metabolism , Embryonic Stem Cells/metabolism , Endoderm/metabolism , Fibroblast Growth Factor 2/metabolism , Gastrula/metabolism , Activins/metabolism , Animals , Biological Evolution , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cell Lineage/drug effects , Cell Lineage/genetics , Digestive System/drug effects , Digestive System/embryology , Dose-Response Relationship, Drug , Embryonic Stem Cells/drug effects , Endoderm/cytology , Endoderm/drug effects , Gastrula/cytology , Gastrula/drug effects , Gene Expression Regulation, Developmental , Hepatocytes/metabolism , Homeodomain Proteins/metabolism , Humans , Intestine, Small/embryology , Intestine, Small/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Pancreas/embryology , Pancreas/metabolism , Protein Kinase Inhibitors/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/metabolism , Time Factors , Trans-Activators/metabolism , Wnt Proteins/metabolism , Wnt3 Protein
7.
PLoS One ; 4(3): e4794, 2009.
Article in English | MEDLINE | ID: mdl-19277121

ABSTRACT

BACKGROUND: Retinoic acid (RA) and fibroblast growth factor 4 (FGF4) signaling control endoderm patterning and pancreas induction/expansion. Based on these findings, RA and FGFs, excluding FGF4, have frequently been used in differentiation protocols to direct differentiation of hESCs into endodermal and pancreatic cell types. In vivo, these signaling pathways act in a temporal and concentration-dependent manner. However, in vitro, the underlying basis for the time of addition of growth and differentiation factors (GDFs), including RA and FGFs, as well as the concentration is lacking. Thus, in order to develop robust and reliable differentiation protocols of ESCs into mature pancreatic cell types, including insulin-producing beta cells, it will be important to mechanistically understand each specification step. This includes differentiation of mesendoderm/definitive endoderm into foregut endoderm--the origin of pancreatic endoderm. METHODOLOGY/PRINCIPAL FINDINGS: Here, we provide data on the individual and combinatorial role of RA and FGF4 in directing differentiation of ActivinA (AA)-induced hESCs into PDX1-expressing cells. FGF4's ability to affect endoderm patterning and specification in vitro has so far not been tested. By testing out the optimal concentration and timing of addition of FGF4 and RA, we present a robust differentiation protocol that on average generates 32% PDX1(+) cells. Furthermore, we show that RA is required for converting AA-induced hESCs into PDX1(+) cells, and that part of the underlying mechanism involves FGF receptor signaling. Finally, further characterization of the PDX1(+) cells suggests that they represent foregut endoderm not yet committed to pancreatic, posterior stomach, or duodenal endoderm. CONCLUSION/SIGNIFICANCE: In conclusion, we show that RA and FGF4 jointly direct differentiation of PDX1(+) foregut endoderm in a robust and efficient manner. RA signaling mediated by the early induction of RARbeta through AA/Wnt3a is required for PDX1 expression. Part of RA's activity is mediated by FGF signaling.


Subject(s)
Embryonic Stem Cells/drug effects , Endoderm/drug effects , Fibroblast Growth Factor 4/physiology , Homeodomain Proteins/biosynthesis , Signal Transduction/physiology , Trans-Activators/biosynthesis , Tretinoin/pharmacology , Activins/pharmacology , Cell Differentiation/drug effects , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Dose-Response Relationship, Drug , Drug Synergism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endoderm/cytology , Endoderm/metabolism , Gene Expression Regulation/drug effects , Homeodomain Proteins/genetics , Humans , Pancreas/cytology , Pancreas/embryology , Pyrroles/pharmacology , RNA, Messenger/biosynthesis , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/biosynthesis , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/physiology , Receptors, Retinoic Acid/biosynthesis , Receptors, Retinoic Acid/genetics , Signal Transduction/drug effects , Time Factors , Trans-Activators/genetics , Up-Regulation/drug effects , Wnt Proteins/physiology , Wnt3 Protein , Wnt3A Protein
8.
Proc Natl Acad Sci U S A ; 105(37): 14005-10, 2008 Sep 16.
Article in English | MEDLINE | ID: mdl-18779593

ABSTRACT

Rheumatoid factors (RF), autoantibodies that bind the Fc region of IgG, are one of the major diagnostic marker in rheumatoid arthritis (RA) but occur with lower frequency also in other infectious and inflammatory conditions. Through positional cloning of the previously described quantitative trait locus (QTL) Rf1 in congenic and advanced intercrossed rats, we identified the Ig lambda light chain locus as a locus that regulates the production of RF in rats. The congenic rats produce RF-Ig lambda and have significant higher levels of RF-IgG and RF-IgM in serum, while the DA rat has an impaired RF production and does not produces RF-Ig lambda. Thus, we could investigate the role of RF in pristane-induced arthritis (PIA) as well as ovalbumin-induced airway inflammation. We show that there was no difference in the development and severity of PIA between congenic and parental DA rats, suggesting that RF using lambda light chains have no impact on PIA. However, the RF producing congenic rats developed a more severe airway inflammation as indicated in the significantly increased number of eosinophils in bronchoalveolar lavage fluid as well as total IgE in serum. In addition, RF congenic rats had a significantly enhanced immune response toward OVA due to increased OVA-Igk but not OVA-Igl antibodies, suggesting a possible involvement of RF in the regulation of the humoral immune response.


Subject(s)
Alleles , Bronchitis/immunology , Bronchitis/metabolism , Hypersensitivity/immunology , Immunoglobulin lambda-Chains/genetics , Immunoglobulin lambda-Chains/immunology , Rheumatoid Factor/metabolism , Animals , Animals, Congenic , Bronchitis/genetics , Bronchitis/pathology , Cloning, Molecular , Genome/genetics , Hybridomas , Hypersensitivity/genetics , Hypersensitivity/metabolism , Hypersensitivity/pathology , Molecular Sequence Data , Rats , Rheumatoid Factor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL