Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Development ; 144(13): 2402-2414, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28576773

ABSTRACT

While much is known about the molecular pathways that regulate embryonic development and adult homeostasis of the endocrine pancreas, little is known about what regulates early postnatal development and maturation of islets. Given that birth marks the first exposure to enteral nutrition, we investigated how nutrient-regulated signaling pathways influence postnatal islet development in mice. We performed loss-of-function studies of mechanistic target of rapamycin (mTOR), a highly conserved kinase within a nutrient-sensing pathway known to regulate cellular growth, morphogenesis and metabolism. Deletion of Mtor in pancreatic endocrine cells had no significant effect on their embryonic development. However, within the first 2 weeks after birth, mTOR-deficient islets became dysmorphic, ß-cell maturation and function were impaired, and animals lost islet mass. Moreover, we discovered that these distinct functions of mTOR are mediated by separate downstream branches of the pathway, in that mTORC1 (with adaptor protein Raptor) is the main complex mediating the maturation and function of islets, whereas mTORC2 (with adaptor protein Rictor) impacts islet mass and architecture. Taken together, these findings suggest that nutrient sensing may be an essential trigger for postnatal ß-cell maturation and islet development.


Subject(s)
Islets of Langerhans/embryology , Islets of Langerhans/metabolism , Morphogenesis , Multiprotein Complexes/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Animals, Newborn , Cell Aggregation , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Models, Biological , Mutation/genetics
2.
Diabetologia ; 62(12): 2245-2251, 2019 12.
Article in English | MEDLINE | ID: mdl-31511930

ABSTRACT

AIMS/HYPOTHESIS: Type 1 diabetes is an autoimmune disorder characterised by loss of insulin-producing beta cells of the pancreas. Progress in understanding the cellular and molecular mechanisms underlying the human disease has been hampered by a dearth of appropriate human experimental models. We previously reported the characterisation of islet-infiltrating CD4+ T cells from a deceased organ donor who had type 1 diabetes. METHODS: Induced pluripotent stem cell (iPSC) lines derived from the above donor were differentiated into CD14+ macrophages and tested for their capacity to present antigen to T cell receptors (TCRs) derived from islet-infiltrating CD4+ T cells from the same donor. RESULTS: The iPSC macrophages displayed typical macrophage morphology, surface markers (CD14, CD86, CD16 and CD11b) and were phagocytic. In response to IFNγ treatment, iPSC macrophages upregulated expression of HLA class II, a characteristic that correlated with their capacity to present epitopes derived from proinsulin C-peptide to a T cell line expressing TCRs derived from islet-infiltrating CD4+ T cells of the original donor. T cell activation was specifically blocked by anti-HLA-DQ antibodies but not by antibodies directed against HLA-DR. CONCLUSIONS/INTERPRETATION: This study provides a proof of principle for the use of iPSC-derived immune cells for modelling key cellular interactions in human type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Induced Pluripotent Stem Cells/metabolism , Islets of Langerhans/metabolism , Macrophages/metabolism , Receptors, Antigen, T-Cell/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/physiology , Diabetes Mellitus, Type 1/immunology , Humans , Induced Pluripotent Stem Cells/immunology , Islets of Langerhans/immunology , Macrophages/immunology , Receptors, Antigen, T-Cell/immunology
3.
Blood ; 119(26): 6243-54, 2012 Jun 28.
Article in English | MEDLINE | ID: mdl-22611158

ABSTRACT

Transcriptional profiling of differentiating human embryonic stem cells (hESCs) revealed that MIXL1-positive mesodermal precursors were enriched for transcripts encoding the G-protein-coupled APELIN receptor (APLNR). APLNR-positive cells, identified by binding of the fluoresceinated peptide ligand, APELIN (APLN), or an anti-APLNR mAb, were found in both posterior mesoderm and anterior mesendoderm populations and were enriched in hemangioblast colony-forming cells (Bl-CFC). The addition of APLN peptide to the media enhanced the growth of embryoid bodies (EBs), increased the expression of hematoendothelial genes in differentiating hESCs, and increased the frequency of Bl-CFCs by up to 10-fold. Furthermore, APLN peptide also synergized with VEGF to promote the growth of hESC-derived endothelial cells. These studies identified APLN as a novel growth factor for hESC-derived hematopoietic and endothelial cells.


Subject(s)
Embryonic Stem Cells/drug effects , Hematopoiesis/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , Apelin , Apelin Receptors , Cells, Cultured , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/physiology , Endoderm/drug effects , Endoderm/metabolism , Endoderm/physiology , Gene Expression Profiling , Hemangioblasts/drug effects , Hemangioblasts/metabolism , Hemangioblasts/physiology , Hematopoiesis/genetics , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Mesoderm/cytology , Mesoderm/drug effects , Mesoderm/metabolism , Mesoderm/physiology , Microarray Analysis , Models, Biological , Protein Binding/drug effects , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
4.
Nat Commun ; 15(1): 7698, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39227582

ABSTRACT

Arterial endothelial cells (AECs) are the founder cells for intraembryonic haematopoiesis. Here, we report a method for the efficient generation of human haemogenic DLL4+ AECs from pluripotent stem cells (PSC). Time-series single-cell RNA-sequencing reveals the dynamic evolution of haematopoiesis and lymphopoiesis, generating cell types with counterparts present in early human embryos, including stages marked by the pre-haematopoietic stem cell genes MECOM/EVI1, MLLT3 and SPINK2. DLL4+ AECs robustly support lymphoid differentiation, without the requirement for exogenous NOTCH ligands. Using this system, we find IL7 acts as a morphogenic factor determining the fate choice between the T and innate lymphoid lineages and also plays a role in regulating the relative expression level of RAG1. Moreover, we document a developmental pathway by which human RAG1+ lymphoid precursors give rise to the natural killer cell lineage. Our study describes an efficient method for producing lymphoid progenitors, providing insights into their endothelial and haematopoietic ontogeny, and establishing a platform to investigate the development of the human blood system.


Subject(s)
Hematopoiesis , Lymphopoiesis , Humans , Hematopoiesis/genetics , Lymphopoiesis/genetics , Endothelial Cells/metabolism , Endothelial Cells/cytology , Cell Differentiation , Cell Lineage/genetics , Interleukin-7/metabolism , Interleukin-7/genetics , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Killer Cells, Natural/metabolism , Killer Cells, Natural/cytology , Hemangioblasts/metabolism , Hemangioblasts/cytology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Single-Cell Analysis/methods , Receptors, Notch/metabolism , Receptors, Notch/genetics
5.
Stem Cell Res ; 56: 102547, 2021 10.
Article in English | MEDLINE | ID: mdl-34619644

ABSTRACT

Insulin expressing beta cells and glucagon expressing alpha cells are the two most abundant endocrine cell types of the human pancreatic islet. Both alpha and beta cells can be generated in vitro via the differentiation of pluripotent stem cells (PSCs), affording the opportunity to study their ontogeny and to examine their developmental inter-relationship. To aid these analyses, we have generated a PSC line in which insulin expression is reported by GFP and glucagon expression is reported by mCherry. This cell line enables viable isolation of cells expressing each hormone and optimisation of methods that lead to their generation.


Subject(s)
Endocrine Cells , Islets of Langerhans , Pluripotent Stem Cells , Cell Differentiation , Glucagon , Green Fluorescent Proteins , Humans , Insulin , Islets of Langerhans/cytology , Luminescent Proteins , Pancreas , Red Fluorescent Protein
6.
Sci Rep ; 11(1): 8315, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33859325

ABSTRACT

The identification of cell surface markers specific to pancreatic beta cells is important for both the study of islet biology and for investigating the pathophysiology of diseases in which this cell type is lost or damaged. Following analysis of publicly available RNAseq data, we identified specific integrin subunits, integrin αv and integrin ß5, that were expressed in beta cells. This finding was further elaborated using immunofluorescence analysis of histological sections derived from donor human pancreas. Despite the broad expression of specific integrin subunits, we found that expression of integrin αvß5 heterodimers was restricted to beta cells and that this complex persisted in islet remnants of some type 1 diabetic individuals from which insulin expression had been lost. This study identifies αvß5 heterodimers as a novel cell surface marker of human pancreatic beta cells, a finding that will aid in the identification and characterisation of this important cell type.


Subject(s)
Diabetes Mellitus, Type 1/diagnosis , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Receptors, Vitronectin/metabolism , Adult , Biomarkers/metabolism , Female , Gene Expression , Humans , Insulin/genetics , Insulin/metabolism , Male , Middle Aged , Receptors, Vitronectin/genetics
7.
Nat Cell Biol ; 22(1): 60-73, 2020 01.
Article in English | MEDLINE | ID: mdl-31907413

ABSTRACT

Defining the ontogeny of the human adaptive immune system during embryogenesis has implications for understanding childhood diseases including leukaemias and autoimmune conditions. Using RAG1:GFP human pluripotent stem cell reporter lines, we examined human T-cell genesis from pluripotent-stem-cell-derived haematopoietic organoids. Under conditions favouring T-cell development, RAG1+ cells progressively upregulated a cohort of recognized T-cell-associated genes, arresting development at the CD4+CD8+ stage. Sort and re-culture experiments showed that early RAG1+ cells also possessed B-cell, myeloid and erythroid potential. Flow cytometry and single-cell-RNA-sequencing data showed that early RAG1+ cells co-expressed the endothelial/haematopoietic progenitor markers CD34, VECAD and CD90, whereas imaging studies identified RAG1+ cells within CD31+ endothelial structures that co-expressed SOX17+ or the endothelial marker CAV1. Collectively, these observations provide evidence for a wave of human T-cell development that originates directly from haemogenic endothelium via a RAG1+ intermediate with multilineage potential.


Subject(s)
Endothelium/cytology , Hemangioblasts/cytology , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/metabolism , Pluripotent Stem Cells/cytology , Cell Differentiation/physiology , Cell Line , Embryonic Development/physiology , Hematopoietic Stem Cell Transplantation/methods , Humans , Organoids/cytology
8.
Stem Cell Res ; 34: 101380, 2019 01.
Article in English | MEDLINE | ID: mdl-30605840

ABSTRACT

We describe the generation and characterization of 5 human induced pluripotent stem cell (iPSC) lines derived from peripheral blood mononuclear cells (PBMCs) of healthy adult individuals. The PBMCs were reprogrammed using non-integrating Sendai viruses containing the reprogramming factors POU5F1 (OCT4), SOX2, KLF4 and MYC. The iPSC lines exhibited a normal karyotype, expressed pluripotency markers and differentiated into cells representative of the three embryonic germ layers. These iPSC lines can be used as controls in studying disease mechanisms.


Subject(s)
Cell Culture Techniques/methods , Induced Pluripotent Stem Cells/pathology , Leukocytes, Mononuclear/pathology , Adult , Cell Line , Female , Humans , Kruppel-Like Factor 4 , Male , Middle Aged , Young Adult
9.
Nat Med ; 23(1): 49-59, 2017 01.
Article in English | MEDLINE | ID: mdl-27869805

ABSTRACT

The enteric nervous system (ENS) of the gastrointestinal tract controls many diverse functions, including motility and epithelial permeability. Perturbations in ENS development or function are common, yet there is no human model for studying ENS-intestinal biology and disease. We used a tissue-engineering approach with embryonic and induced pluripotent stem cells (PSCs) to generate human intestinal tissue containing a functional ENS. We recapitulated normal intestinal ENS development by combining human-PSC-derived neural crest cells (NCCs) and developing human intestinal organoids (HIOs). NCCs recombined with HIOs in vitro migrated into the mesenchyme, differentiated into neurons and glial cells and showed neuronal activity, as measured by rhythmic waves of calcium transients. ENS-containing HIOs grown in vivo formed neuroglial structures similar to a myenteric and submucosal plexus, had functional interstitial cells of Cajal and had an electromechanical coupling that regulated waves of propagating contraction. Finally, we used this system to investigate the cellular and molecular basis for Hirschsprung's disease caused by a mutation in the gene PHOX2B. This is, to the best of our knowledge, the first demonstration of human-PSC-derived intestinal tissue with a functional ENS and how this system can be used to study motility disorders of the human gastrointestinal tract.


Subject(s)
Enteric Nervous System/physiology , Induced Pluripotent Stem Cells , Intestines/physiology , Neural Crest , Organoids , Tissue Engineering/methods , Animals , Calcium/metabolism , Cell Line , Chick Embryo , Enteric Nervous System/physiopathology , Gastrointestinal Motility , Hirschsprung Disease/genetics , Hirschsprung Disease/physiopathology , Homeodomain Proteins/genetics , Humans , Immunohistochemistry , In Vitro Techniques , Interstitial Cells of Cajal/physiology , Intestines/physiopathology , Mice , Mice, SCID , Microscopy, Confocal , Models, Biological , Mutation , Myenteric Plexus/physiology , Myenteric Plexus/physiopathology , Neurogenesis/physiology , Neuroglia/physiology , Neurons/physiology , Permeability , Real-Time Polymerase Chain Reaction , Submucous Plexus/physiology , Submucous Plexus/physiopathology , Transcription Factors/genetics
10.
Stem Cell Reports ; 7(3): 518-526, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27594589

ABSTRACT

The ability to reliably express fluorescent reporters or other genes of interest is important for using human pluripotent stem cells (hPSCs) as a platform for investigating cell fates and gene function. We describe a simple expression system, designated GAPTrap (GT), in which reporter genes, including GFP, mCherry, mTagBFP2, luc2, Gluc, and lacZ are inserted into the GAPDH locus in hPSCs. Independent clones harboring variations of the GT vectors expressed remarkably consistent levels of the reporter gene. Differentiation experiments showed that reporter expression was reliably maintained in hematopoietic cells, cardiac mesoderm, definitive endoderm, and ventral midbrain dopaminergic neurons. Similarly, analysis of teratomas derived from GT-lacZ hPSCs showed that ß-galactosidase expression was maintained in a spectrum of cell types representing derivatives of the three germ layers. Thus, the GAPTrap vectors represent a robust and straightforward tagging system that enables indelible labeling of PSCs and their differentiated derivatives.


Subject(s)
Gene Expression , Genes, Reporter , Genetic Vectors/genetics , Pluripotent Stem Cells/metabolism , Transgenes , CRISPR-Cas Systems , Cell Differentiation , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats , Humans , Pluripotent Stem Cells/cytology , Transcription Activator-Like Effector Nucleases
11.
Ann N Y Acad Sci ; 1311: 124-37, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24611778

ABSTRACT

In 1998, the landmark paper describing the isolation and culture of human embryonic stem cells (ESCs) was published. Since that time, the main goal of many diabetes researchers has been to derive ß cells from ESCs as a renewable cell-based therapy for the treatment of patients with diabetes. In working toward this goal, numerous protocols that attempt to recapitulate normal pancreatic development have been published that result in the formation of pancreatic cell types from human pluripotent cells. This review examines stem cell differentiation methods and places them within the context of pancreatic development. We additionally compare strategies that are currently being used to generate pancreatic cell types and contrast them with approaches that have been used to generate functional cell types in different lineages. In doing this, we aim to identify how new approaches might be used to improve yield and functionality of in vitro-derived pancreatic ß cells as an eventual cell-based therapy for type 1 diabetes.


Subject(s)
Cell Differentiation , Insulin-Secreting Cells/cytology , Pancreas/cytology , Pluripotent Stem Cells/cytology , Humans
12.
Rev Diabet Stud ; 11(1): 6-18, 2014.
Article in English | MEDLINE | ID: mdl-25148364

ABSTRACT

Human embryonic stem cells have been advanced as a source of insulin-producing cells that could potentially replace cadaveric-derived islets in the treatment of type 1 diabetes. To this end, protocols have been developed that promote the formation of pancreatic progenitors and endocrine cells from human pluripotent stem cells, encompassing both embryonic stem cells and induced pluripotent stem cells. In this review, we examine these methods and place them in the context of the developmental and embryological studies upon which they are based. In particular, we outline the stepwise differentiation of cells towards definitive endoderm, pancreatic endoderm, endocrine lineages and the emergence of functional beta-cells. In doing so, we identify key factors common to many such protocols and discuss the proposed action of these factors in the context of cellular differentiation and ongoing development. We also compare strategies that entail transplantation of progenitor populations with those that seek to develop fully functional hormone expressing cells in vitro. Overall, our survey of the literature highlights the significant progress already made in the field and identifies remaining deficiencies in developing a pluripotent stem cell based treatment for type 1 diabetes.


Subject(s)
Cell Differentiation , Insulin-Secreting Cells/cytology , Models, Biological , Pluripotent Stem Cells/cytology , Animals , Cells, Cultured , Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Insulin-Secreting Cells/metabolism , Primary Cell Culture/trends
13.
Stem Cells Dev ; 21(10): 1662-74, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22034992

ABSTRACT

To develop methods for the generation of insulin-producing ß-cells for the treatment of diabetes, we have used GFP-tagged embryonic stem cells (ESCs) to elucidate the process of pancreas development. Using the reporter Pdx1(GFP/w) ESC line, we have previously described a serum-free differentiation protocol in which Pdx1-GFP(+) cells formed GFP bright (GFP(br)) epithelial buds that resembled those present in the developing mouse pancreas. In this study we extend these findings to demonstrate that these cells can undergo a process of branching morphogenesis, similar to that seen during pancreatic development of the mid-gestation embryo. These partially disaggregated embryoid bodies containing GFP(br) buds initially form epithelial ring-like structures when cultured in Matrigel. After several days in culture, these rings undergo a process of proliferation and form a ramified network of epithelial branches. Comparative analysis of explanted dissociated pancreatic buds from E13.5 Pdx1(GFP/w) embryos and ESC-derived GFP(br) buds reveal a similar process of proliferation and branching, with both embryonic Pdx1(GFP/w) branching pancreatic epithelium and ESC-derived GFP(br) branching organoids expressing markers representing epithelial (EpCAM and E-Cadherin), ductal (Mucin1), exocrine (Amylase and Carboxypeptidase 1A), and endocrine cell types (Glucagon and Somatostatin). ESC-derived branching structures also expressed a suite of genes indicative of ongoing pancreatic differentiation, paralleling gene expression within similar structures derived from the E13.5 fetal pancreas. In summary, differentiating mouse ESCs can generate pancreatic material that has significant similarity to the fetal pancreatic anlagen, providing an in vitro platform for investigating the cellular and molecular mechanisms underpinning pancreatic development.


Subject(s)
Embryoid Bodies/physiology , Embryonic Development , Organogenesis , Pancreas/embryology , Animals , Cell Differentiation , Cells, Cultured , Coculture Techniques , Embryoid Bodies/metabolism , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/physiology , Endoderm/cytology , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Pancreas/cytology , Pancreas/metabolism , Real-Time Polymerase Chain Reaction , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tissue Culture Techniques , Trans-Activators/genetics , Trans-Activators/metabolism
14.
PLoS One ; 5(5): e10706, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20502661

ABSTRACT

BACKGROUND: Mesendoderm induction during embryonic stem cell (ESC) differentiation in vitro is stimulated by the Transforming Growth Factor and Wingless (Wnt) families of growth factors. PRINCIPAL FINDINGS: We identified the periods during which Bone Morphogenetic Protein (BMP) 4, Wnt3a or Activin A were able to induce expression of the mesendoderm marker, Mixl1, in differentiating mouse ESCs. BMP4 and Wnt3a were required between differentiation day (d) 1.5 and 3 to most effectively induce Mixl1, whilst Activin A induced Mixl1 expression in ESC when added between d2 and d4, indicating a subtle difference in the requirement for Activin receptor signalling in this process. Stimulation of ESCs with these factors at earlier or later times resulted in little Mixl1 induction, suggesting that the differentiating ESCs passed through 'temporal windows' in which they sequentially gained and lost competence to respond to each growth factor. Inhibition of either Activin or Wnt signalling blocked Mixl1 induction by any of the three mesendoderm-inducing factors. Mixing experiments in which chimeric EBs were formed between growth factor-treated and untreated ESCs revealed that BMP, Activin and Wnt signalling all contributed to the propagation of paracrine mesendoderm inducing signals between adjacent cells. Finally, we demonstrated that the differentiating cells passed through 'exit gates' after which point they were no longer dependent on signalling from inducing molecules for Mixl1 expression. CONCLUSIONS: These studies suggest that differentiating ESCs are directed by an interconnected network of growth factors similar to those present in early embryos and that the timing of growth factor activity is critical for mesendoderm induction.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endoderm/metabolism , Paracrine Communication , Signal Transduction , Activins/metabolism , Animals , Bone Morphogenetic Protein 4/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Endoderm/cytology , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Time Factors , Wnt Proteins/metabolism , Wnt3 Protein , Wnt3A Protein
SELECTION OF CITATIONS
SEARCH DETAIL