Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(1): 113668, 2024 01 23.
Article in English | MEDLINE | ID: mdl-38198277

ABSTRACT

Perlecan (HSPG2), a heparan sulfate proteoglycan similar to agrin, is key for extracellular matrix (ECM) maturation and stabilization. Although crucial for cardiac development, its role remains elusive. We show that perlecan expression increases as cardiomyocytes mature in vivo and during human pluripotent stem cell differentiation to cardiomyocytes (hPSC-CMs). Perlecan-haploinsuffient hPSCs (HSPG2+/-) differentiate efficiently, but late-stage CMs have structural, contractile, metabolic, and ECM gene dysregulation. In keeping with this, late-stage HSPG2+/- hPSC-CMs have immature features, including reduced ⍺-actinin expression and increased glycolytic metabolism and proliferation. Moreover, perlecan-haploinsuffient engineered heart tissues have reduced tissue thickness and force generation. Conversely, hPSC-CMs grown on a perlecan-peptide substrate are enlarged and display increased nucleation, typical of hypertrophic growth. Together, perlecan appears to play the opposite role of agrin, promoting cellular maturation rather than hyperplasia and proliferation. Perlecan signaling is likely mediated via its binding to the dystroglycan complex. Targeting perlecan-dependent signaling may help reverse the phenotypic switch common to heart failure.


Subject(s)
Agrin , Heparan Sulfate Proteoglycans , Humans , Heparan Sulfate Proteoglycans/genetics , Heparan Sulfate Proteoglycans/metabolism , Agrin/metabolism , Myocytes, Cardiac/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism
2.
Cell Rep Methods ; 3(4): 100456, 2023 04 24.
Article in English | MEDLINE | ID: mdl-37159667

ABSTRACT

Decreased left ventricle (LV) function caused by genetic mutations or injury often leads to debilitating and fatal cardiovascular disease. LV cardiomyocytes are, therefore, a potentially valuable therapeutical target. Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) are neither homogeneous nor functionally mature, which reduces their utility. Here, we exploit cardiac development knowledge to instruct differentiation of hPSCs specifically toward LV cardiomyocytes. Correct mesoderm patterning and retinoic acid pathway blocking are essential to generate near-homogenous LV-specific hPSC-CMs (hPSC-LV-CMs). These cells transit via first heart field progenitors and display typical ventricular action potentials. Importantly, hPSC-LV-CMs exhibit increased metabolism, reduced proliferation, and improved cytoarchitecture and functional maturity compared with age-matched cardiomyocytes generated using the standard WNT-ON/WNT-OFF protocol. Similarly, engineered heart tissues made from hPSC-LV-CMs are better organized, produce higher force, and beat more slowly but can be paced to physiological levels. Together, we show that functionally matured hPSC-LV-CMs can be obtained rapidly without exposure to current maturation regimes.


Subject(s)
Cardiovascular Diseases , Pluripotent Stem Cells , Humans , Myocytes, Cardiac , Heart Ventricles , Action Potentials
3.
Stem Cell Reports ; 17(4): 894-910, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35334218

ABSTRACT

The spinal cord emerges from a niche of neuromesodermal progenitors (NMPs) formed and maintained by WNT/fibroblast growth factor (FGF) signals at the posterior end of the embryo. NMPs can be generated from human pluripotent stem cells and hold promise for spinal cord replacement therapies. However, NMPs are transient, which compromises production of the full range of rostrocaudal spinal cord identities in vitro. Here we report the generation of NMP-derived pre-neural progenitors (PNPs) with stem cell-like self-renewal capacity. PNPs maintain pre-spinal cord identity for 7-10 passages, dividing to self-renew and to make neural crest progenitors, while gradually adopting a more posterior identity by activating colinear HOX gene expression. The HOX clock can be halted through GDF11-mediated signal inhibition to produce a PNP and NC population with a thoracic identity that can be maintained for up to 30 passages.


Subject(s)
Neural Crest , Pluripotent Stem Cells , Body Patterning/genetics , Bone Morphogenetic Proteins/metabolism , Cell Differentiation/genetics , Fibroblast Growth Factors/metabolism , Growth Differentiation Factors/metabolism , Humans , Neural Crest/metabolism , Pluripotent Stem Cells/metabolism , Spinal Cord/metabolism
4.
Metabolites ; 12(1)2021 Dec 22.
Article in English | MEDLINE | ID: mdl-35050131

ABSTRACT

Cardiomyocytes rely on specialised metabolism to meet the high energy demand of the heart. During heart development, metabolism matures and shifts from the predominant utilisation of glycolysis and glutamine oxidation towards lactate and fatty acid oxidation. Iron deficiency (ID) leads to cellular metabolism perturbations. However, the exact alterations in substrate metabolism during ID are poorly defined. Using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM), the present study investigated changes in major metabolic substrate utilisation in the context of ID or upon transferrin rescue. Typically, during hiPSC-CM differentiation, the greatest increase in total metabolic output and rate was seen in fatty acid metabolism. When ID was induced, hiPSC-CMs displayed increased reliance on glycolytic metabolism, and six TCA cycle, five amino acid, and four fatty acid substrates were significantly impaired. Transferrin rescue was able to improve TCA cycle substrate metabolism, but the amino acid and fatty acid metabolism remained perturbed. Replenishing iron stores partially reverses the adverse metabolic changes that occur during ID. Understanding the changes in metabolic substrate utilisation and their modification may provide potential for discovery of new biomarkers and therapeutic targets in cardiovascular diseases.

5.
Epigenetics Chromatin ; 12(1): 14, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30767785

ABSTRACT

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


Subject(s)
Alleles , Genomic Imprinting , Mouse Embryonic Stem Cells/metabolism , RNA, Messenger/genetics , Animals , Cell Differentiation , Cell Line , Cells, Cultured , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Silencing , Germ Layers/cytology , Germ Layers/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mouse Embryonic Stem Cells/cytology
6.
Biol Open ; 7(8)2018 Aug 17.
Article in English | MEDLINE | ID: mdl-30026265

ABSTRACT

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

7.
Stem Cell Reports ; 7(4): 764-776, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27641648

ABSTRACT

Wnt signaling is a key regulator of vertebrate heart development; however, specific roles for human cardiomyocyte development remain uncertain. Here we use human embryonic stem cells (hESCs) to analyze systematically in human cardiomyocyte development the expression of endogenous Wnt signaling components, monitor pathway activity, and dissect stage-specific requirements for canonical and noncanonical Wnt signaling mechanisms using small-molecule inhibitors. Our analysis suggests that WNT3 and WNT8A, via FZD7 and canonical signaling, regulate BRACHYURY expression and mesoderm induction; that WNT5A/5B, via ROR2 and noncanonical signaling, regulate MESP1 expression and cardiovascular development; and that later in development WNT2, WNT5A/5B, and WNT11, via FZD4 and FZD6, regulate functional cardiomyocyte differentiation via noncanonical Wnt signaling. Our findings confirm in human development previously proposed roles for canonical Wnt signaling in sequential stages of vertebrate cardiomyogenesis, and identify more precise roles for noncanonical signaling and for individual Wnt signal and Wnt receptor genes in human cardiomyocyte development.


Subject(s)
Cell Differentiation , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Organogenesis , Wnt Signaling Pathway , Biomarkers , Cell Differentiation/genetics , Embryonic Development/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mesoderm/embryology , Mesoderm/metabolism , beta Catenin/metabolism
8.
Development ; 142(12): 2121-35, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-26015544

ABSTRACT

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


Subject(s)
Embryonic Stem Cells/cytology , Fetal Proteins/metabolism , Gene Expression Regulation, Developmental , Neurogenesis/physiology , Smad1 Protein/metabolism , T-Box Domain Proteins/metabolism , Animals , Bone Morphogenetic Protein 4/metabolism , Cell Line , Embryonic Stem Cells/metabolism , Endoderm/cytology , Gastrulation/physiology , Humans , Mesoderm/cytology , Mice , Mice, Transgenic , Smad2 Protein/metabolism , Smad3 Protein/metabolism
9.
Nat Protoc ; 9(4): 929-38, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24675733

ABSTRACT

Vascular smooth muscle cells (SMCs) arise from diverse developmental origins. Regional distribution of vascular diseases may, in part, be attributed to this inherent heterogeneity in SMC lineage. Therefore, systems for generating human SMC subtypes of distinct embryonic origins would represent useful platforms for studying the influence of SMC lineage on the spatial specificity of vascular disease. Here we describe how human pluripotent stem cells can be differentiated into distinct populations of SMC subtypes under chemically defined conditions. The initial stage (days 0-5 or 0-7) begins with the induction of three intermediate lineages: neuroectoderm, lateral plate mesoderm and paraxial mesoderm. Subsequently, these precursor lineages are differentiated into contractile SMCs (days 5-19+). At key stages, the emergence of lineage-specific markers confirms recapitulation of embryonic developmental pathways and generation of functionally distinct SMC subtypes. The ability to derive an unlimited supply of human SMCs will accelerate applications in regenerative medicine and disease modeling.


Subject(s)
Cell Culture Techniques/methods , Muscle, Smooth, Vascular/cytology , Pluripotent Stem Cells , Cell Differentiation , Cell Lineage , Humans , Mesoderm/cytology
10.
Reproduction ; 147(5): D1-12, 2014 May.
Article in English | MEDLINE | ID: mdl-24518070

ABSTRACT

It is imperative to unveil the full range of differentiated cell types into which human pluripotent stem cells (hPSCs) can develop. The need is twofold: it will delimit the therapeutic utility of these stem cells and is necessary to place their position accurately in the developmental hierarchy of lineage potential. Accumulated evidence suggested that hPSC could develop in vitro into an extraembryonic lineage (trophoblast (TB)) that is typically inaccessible to pluripotent embryonic cells during embryogenesis. However, whether these differentiated cells are truly authentic TB has been challenged. In this debate, we present a case for and a case against TB differentiation from hPSCs. By analogy to other differentiation systems, our debate is broadly applicable, as it articulates higher and more challenging standards for judging whether a given cell type has been genuinely produced from hPSC differentiation.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Trophoblasts/cytology , Cell Lineage , Cells, Cultured , Embryonic Stem Cells/physiology , Female , Humans , In Vitro Techniques , Morphogenesis/physiology , Placenta/cytology , Placenta/physiology , Pregnancy , Trophoblasts/physiology
11.
Nat Biotechnol ; 30(2): 165-73, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22252507

ABSTRACT

Heterogeneity of embryological origins is a hallmark of vascular smooth muscle cells (SMCs) and may influence the development of vascular disease. Differentiation of human pluripotent stem cells (hPSCs) into developmental origin-specific SMC subtypes remains elusive. Here we describe a chemically defined protocol in which hPSCs were initially induced to form neuroectoderm, lateral plate mesoderm or paraxial mesoderm. These intermediate populations were further differentiated toward SMCs (>80% MYH11(+) and ACTA2(+)), which displayed contractile ability in response to vasoconstrictors and invested perivascular regions in vivo. Derived SMC subtypes recapitulated the unique proliferative and secretory responses to cytokines previously documented in studies using aortic SMCs of distinct origins. Notably, this system predicted increased extracellular matrix degradation by SMCs derived from lateral plate mesoderm, which was confirmed using rat aortic SMCs from corresponding origins. This differentiation approach will have broad applications in modeling origin-dependent disease susceptibility and in developing bioengineered vascular grafts for regenerative medicine.


Subject(s)
Cell Differentiation , Muscle, Smooth, Vascular/embryology , Muscle, Smooth, Vascular/growth & development , Animals , Disease Susceptibility , Extracellular Matrix/metabolism , Humans , Mesoderm/growth & development , Neural Plate/growth & development , Pluripotent Stem Cells/cytology , Rats , Regenerative Medicine , Vasoconstrictor Agents/metabolism
12.
Cell Stem Cell ; 9(2): 144-55, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21816365

ABSTRACT

BMP is thought to induce hESC differentiation toward multiple lineages including mesoderm and trophoblast. The BMP-induced trophoblast phenotype is a long-standing paradox in stem cell biology. Here we readdressed BMP function in hESCs and mouse epiblast-derived cells. We found that BMP4 cooperates with FGF2 (via ERK) to induce mesoderm and to inhibit endoderm differentiation. These conditions induced cells with high levels of BRACHYURY (BRA) that coexpressed CDX2. BRA was necessary for and preceded CDX2 expression; both genes were essential for expression not only of mesodermal genes but also of trophoblast-associated genes. Maximal expression of the latter was seen in the absence of FGF but these cells coexpressed mesodermal genes and moreover they differed in cell surface and epigenetic properties from placental trophoblast. We conclude that BMP induces human and mouse pluripotent stem cells primarily to form mesoderm, rather than trophoblast, acting through BRA and CDX2.


Subject(s)
Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation/drug effects , Cell Lineage/drug effects , Embryonic Stem Cells/cytology , Fetal Proteins/metabolism , Homeodomain Proteins/metabolism , Pluripotent Stem Cells/cytology , T-Box Domain Proteins/metabolism , Animals , CDX2 Transcription Factor , Chromones/pharmacology , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Fetal Proteins/genetics , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation, Developmental/drug effects , Gene Regulatory Networks/genetics , Glycoprotein Hormones, alpha Subunit/genetics , Glycoprotein Hormones, alpha Subunit/metabolism , Homeodomain Proteins/genetics , Humans , Keratin-7/genetics , Keratin-7/metabolism , Mesoderm/cytology , Mesoderm/drug effects , Mesoderm/metabolism , Mice , Morpholines/pharmacology , Neuropeptides/genetics , Neuropeptides/metabolism , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Signal Transduction/drug effects , T-Box Domain Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Trophoblasts/cytology , Trophoblasts/drug effects , Trophoblasts/metabolism
13.
Stem Cells ; 27(2): 341-51, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19056911

ABSTRACT

Embryonic stem (ES) cells represent a possible source of islet tissue for the treatment of diabetes. Achieving this goal will require a detailed understanding of how the transcription factor cascade initiated by the homeodomain transcription factor Pdx1 culminates in pancreatic beta-cell development. Here we describe a genetic approach that enables fine control of Pdx1 transcriptional activity during endoderm differentiation of mouse and human ES cell. By activating an exogenous Pdx1VP16 protein in populations of cells enriched in definitive endoderm we show a distinct lineage-dependent requirement for this transcription factor's activity. Mimicking the natural biphasic pattern of Pdx1 expression was necessary to induce an endocrine pancreas-like cell phenotype, in which 30% of the cells were beta-cell-like. Cell markers consistent with the different beta-cell differentiation stages appeared in a sequential order following the natural pattern of pancreatic development. Furthermore, in mouse ES-derived cultures the differentiated beta-like cells secreted C-peptide (insulin) in response to KCl and 3-isobutyl-1-methylxanthine, suggesting that following a natural path of development in vitro represents the best approach to generate functional pancreatic cells. Together these results reveal for the first time a significant effect of the timed expression of Pdx1 on the non-beta-cells in the developing endocrine pancreas. Collectively, we show that this method of in vitro differentiation provides a template for inducing and studying ES cell differentiation into insulin-secreting cells.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/metabolism , Trans-Activators/metabolism , Activins/pharmacology , Amyloid/genetics , Animals , Blotting, Western , Bone Morphogenetic Protein 4/pharmacology , C-Peptide/metabolism , Cell Differentiation/drug effects , Cell Line , Embryonic Stem Cells/drug effects , Flow Cytometry , Gene Expression/drug effects , HeLa Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Humans , Immunohistochemistry , Insulin/genetics , Islet Amyloid Polypeptide , LIM-Homeodomain Proteins , Mice , Polymerase Chain Reaction , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Trans-Activators/genetics , Trans-Activators/physiology , Transcription Factors
14.
Mol Cell Endocrinol ; 294(1-2): 1-9, 2008 Nov 06.
Article in English | MEDLINE | ID: mdl-18687378

ABSTRACT

In recent years major progress has been made in understanding the role of transcription factors in the development of the endocrine pancreas in the mouse. Here we describe how a number of these transcription factors play a role in maintaining the differentiated phenotype of the beta cell, and in the mechanisms that allow the beta cell to adapt to changing metabolic demands that occur throughout life. Amongst these factors, Pdx1 plays a critical role in defining the region of the primitive gut that will form the pancreas, Ngn3 expression drives cells towards an endocrine lineage, and a number of additional proteins including Pdx1, in a second wave of expression, Pax4, NeuroD1/beta2, and MafA act as beta cell differentiation factors. In the mature beta cell Pdx1, MafA, beta2, and Nkx2.2 play important roles in regulating expression of insulin and to some extent other genes responsible for maintaining beta cell function. We emphasise here that data from gene expression studies in rodents seldom map on to the known structure of the corresponding human promoters. In the adult the beta cell is particularly susceptible to autoimmune-mediated attack and to the toxic metabolic milieu associated with over-eating, and utilises a number of these transcription factors in its defence. Pdx1 has anti-apoptotic and proliferative activities that help facilitate the maintenance of beta cell mass, while Ngn3 may be involved in the recruitment of progenitor cells, and Pax4 (and possibly HNF1alpha and Hnf4alpha) in the proliferation of beta cells in the adult pancreas. Other transcription factors with a more widespread pattern of expression that play a role in beta cell survival or proliferation include Foxo1, CREB family members, NFAT, FoxM1, Snail and Asc-2.


Subject(s)
Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Transcription Factors/metabolism , Animals , Cell Survival , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Humans , Nuclear Proteins , Regeneration
15.
Biochem Soc Trans ; 36(Pt 3): 363-5, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18481959

ABSTRACT

Obesity is a metabolic disorder, which has been recognized as a global epidemic. It contributes to insulin resistance, the major cause of Type 2 diabetes, as well as to the development of other related diseases. Our basic premise is that a better understanding of how adult stem cells of the pancreas contribute to the maintenance of the pancreatic beta-cell pool against the increased metabolic demands associated with obesity may provide new therapeutic targets for treating diabetes. At the same time, if we knew more about the biology of adipocyte formation, maintenance and deposition in obese individuals, perhaps some control over the adipocyte tissue mass of these individuals would be facilitated and treatment of obesity would become available. Many investigations in the field are therefore aimed at describing how adipocyte stem cells function in the various sites of fat deposition and the extent to which these stem cells contribute to both brown and white adipocytes. Studies on the differentiation of human embryonic stem cells along the pancreatic and adipocyte lineages may therefore better inform approaches to these studies.


Subject(s)
Metabolic Diseases/pathology , Stem Cells/cytology , Animals , Cell Differentiation , Cell Lineage , Diabetes Mellitus, Type 2/pathology , Humans , Obesity/pathology
16.
Semin Cell Dev Biol ; 18(6): 827-38, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17959396

ABSTRACT

There is a compelling need to develop novel therapies for diabetes mellitus. Recent successes in the transplantation of islets of Langerhans are seen as a major breakthrough. However, there is huge disparity between potential recipients and the availability of donor tissue. Human embryonic stem cells induced to form pancreatic beta cells could provide a replenishable supply of tissue. Early studies on the spontaneous differentiation of mouse embryonic stem cells have laid the foundation for a more directed approach based on recapitulating the events that occur during the development of the pancreas in the mouse. A high yield of definitive endoderm has been achieved, and although beta-like cells can be generated in a step-wise manner, the efficiency is still low and the final product is not fully differentiated. Future challenges include generating fully functional islet cells under Xeno-free and chemically defined conditions and circumventing the need for immunosuppression.


Subject(s)
Diabetes Mellitus/therapy , Embryonic Stem Cells/transplantation , Stem Cell Transplantation/methods , Animals , Embryonic Stem Cells/cytology , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/transplantation
17.
Mol Cell Endocrinol ; 253(1-2): 14-21, 2006 Jul 11.
Article in English | MEDLINE | ID: mdl-16698177

ABSTRACT

In order to purify and characterize nestin-positive cells in the developing pancreas a transgenic mouse was generated, in which the enhanced green fluorescent protein (EGFP) was driven by the nestin second intronic enhancer and upstream promoter. In keeping with previous studies on the distribution of nestin, EGFP was expressed in the developing embryo in neurones in the brain, eye, spinal cord, tail bud and glial cells in the small intestine. In the pancreas there was no detectable EGFP at embryonic day 11.5 (E11.5). EGFP expression appeared at E12.5 and increased in intensity through E14.5, E18.5 and post-natal day 1. Flow cytometry was used to quantify and purify the EGFP positive population in the E15.5 pancreas. The purified (96%) EGFP-expressing cells, which represent 20% of the total cell population, were shown by RT/PCR to express exocrine cell markers (amylase and P48) and endocrine cell markers (insulin 1, insulin 2, and Ngn3). They also expressed, at a lower level, PDX-1, Isl-1, and the islet hormones pancreatic polypeptide, glucagon and somatostatin as well as GLUT2, the stem cell marker ABCG2 and PECAM, a marker of endothelial cells. It was further shown by immunocytochemistry of the E15.5 pancreas that EGFP colocalised in separate subpopulations of cells that expressed nestin, insulin and amylase. These results support the conclusion that nestin expressing cells can give rise to both endocrine and exocrine cells. The ability to purify these putative progenitor cells may provide further insights into their properties and function.


Subject(s)
Green Fluorescent Proteins/genetics , Intermediate Filament Proteins/genetics , Islets of Langerhans/embryology , Nerve Tissue Proteins/genetics , Pancreas, Exocrine/enzymology , Amylases/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/metabolism , Insulin/metabolism , Islets of Langerhans/enzymology , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Nestin , Promoter Regions, Genetic , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...