46 research outputs found

    The role of ARX in human pancreatic endocrine specification

    Get PDF
    The in vitro differentiation of human embryonic stem cells (hESCs) offers a model system to explore human development. Humans with mutations in the transcription factor Aristaless Related Homeobox (ARX) often suffer from the syndrome X-linked lissencephaly with ambiguous genitalia (XLAG), affecting many cell types including those of the pancreas. Indeed, XLAG pancreatic islets lack glucagon and pancreatic polypeptide-positive cells but retain somatostatin, insulin, and ghrelin-positive cells. To further examine the role of ARX in human pancreatic endocrine development, we utilized genomic editing in hESCs to generate deletions in ARX. ARX knockout hESCs retained pancreatic differentiation capacity and ARX knockout endocrine cells were biased toward somatostatin-positive cells (94% of endocrine cells) with reduced pancreatic polypeptide (rarely detected), glucagon (90% reduced) and insulin-positive (65% reduced) lineages. ARX knockout somatostatin-positive cells shared expression patterns with human fetal and adult δ-cells. Differentiated ARX knockout cells upregulated PAX4, NKX2.2, ISL1, HHEX, PCSK1, PCSK2 expression while downregulating PAX6 and IRX2. Re-expression of ARX in ARX knockout pancreatic progenitors reduced HHEX and increased PAX6 and insulin expression following differentiation. Taken together these data suggest that ARX plays a key role in pancreatic endocrine fate specification of pancreatic polypeptide, somatostatin, glucagon and insulin positive cells from hESCs

    A Multi-Parameter, High-Content, High-Throughput Screening Platform to Identify Natural Compounds that Modulate Insulin and Pdx1 Expression

    Get PDF
    Diabetes is a devastating disease that is ultimately caused by the malfunction or loss of insulin-producing pancreatic beta-cells. Drugs capable of inducing the development of new beta-cells or improving the function or survival of existing beta-cells could conceivably cure this disease. We report a novel high-throughput screening platform that exploits multi-parameter high-content analysis to determine the effect of compounds on beta-cell survival, as well as the promoter activity of two key beta-cell genes, insulin and pdx1. Dispersed human pancreatic islets and MIN6 beta-cells were infected with a dual reporter lentivirus containing both eGFP driven by the insulin promoter and mRFP driven by the pdx1 promoter. B-score statistical transformation was used to correct systemic row and column biases. Using this approach and 5 replicate screens, we identified 7 extracts that reproducibly changed insulin and/or pdx1 promoter activity from a library of 1319 marine invertebrate extracts. The ability of compounds purified from these extracts to significantly modulate insulin mRNA levels was confirmed with real-time PCR. Insulin secretion was analyzed by RIA. Follow-up studies focused on two lead compounds, one that stimulates insulin gene expression and one that inhibits insulin gene expression. Thus, we demonstrate that multi-parameter, high-content screening can identify novel regulators of beta-cell gene expression, such as bivittoside D. This work represents an important step towards the development of drugs to increase insulin expression in diabetes and during in vitro differentiation of beta-cell replacements

    Finishing the euchromatic sequence of the human genome

    Get PDF
    The sequence of the human genome encodes the genetic instructions for human physiology, as well as rich information about human evolution. In 2001, the International Human Genome Sequencing Consortium reported a draft sequence of the euchromatic portion of the human genome. Since then, the international collaboration has worked to convert this draft into a genome sequence with high accuracy and nearly complete coverage. Here, we report the result of this finishing process. The current genome sequence (Build 35) contains 2.85 billion nucleotides interrupted by only 341 gaps. It covers ∼99% of the euchromatic genome and is accurate to an error rate of ∼1 event per 100,000 bases. Many of the remaining euchromatic gaps are associated with segmental duplications and will require focused work with new methods. The near-complete sequence, the first for a vertebrate, greatly improves the precision of biological analyses of the human genome including studies of gene number, birth and death. Notably, the human enome seems to encode only 20,000-25,000 protein-coding genes. The genome sequence reported here should serve as a firm foundation for biomedical research in the decades ahead

    Initial Cell Seeding Density Influences Pancreatic Endocrine Development During <i>in vitro</i> Differentiation of Human Embryonic Stem Cells

    Get PDF
    <div><p>Human embryonic stem cells (hESCs) have the ability to form cells derived from all three germ layers, and as such have received significant attention as a possible source for insulin-secreting pancreatic beta-cells for diabetes treatment. While considerable advances have been made in generating hESC-derived insulin-producing cells, to date <i>in vitro</i>-derived glucose-responsive beta-cells have remained an elusive goal. With the objective of increasing the <i>in vitro</i> formation of pancreatic endocrine cells, we examined the effect of varying initial cell seeding density from 1.3 x 10<sup>4</sup> cells/cm<sup>2</sup> to 5.3 x 10<sup>4</sup> cells/cm<sup>2</sup> followed by a 21-day pancreatic endocrine differentiation protocol. Low density-seeded cells were found to be biased toward the G2/M phases of the cell cycle and failed to efficiently differentiate into SOX17-CXCR4 co-positive definitive endoderm cells leaving increased numbers of OCT4 positive cells in day 4 cultures. Moderate density cultures effectively formed definitive endoderm and progressed to express PDX1 in approximately 20% of the culture. High density cultures contained approximately double the numbers of PDX1 positive pancreatic progenitor cells and also showed increased expression of <i>MNX1</i>, <i>PTF1a</i>, <i>NGN3</i>, <i>ARX</i>, and <i>PAX4</i> compared to cultures seeded at moderate density. The cultures seeded at high density displayed increased formation of polyhormonal pancreatic endocrine cell populations co-expressing insulin, glucagon and somatostatin. The maturation process giving rise to these endocrine cell populations followed the expected cascade of pancreatic progenitor marker (<i>PDX1</i> and <i>MNX1</i>) expression, followed by pancreatic endocrine specification marker expression (<i>BRN4</i>, <i>PAX4</i>, <i>ARX</i>, <i>NEUROD1</i>, <i>NKX6.1</i> and <i>NKX2.2</i>) and then pancreatic hormone expression (insulin, glucagon and somatostatin). Taken together these data suggest that initial cell seeding density plays an important role in both germ layer specification and pancreatic progenitor commitment, which precedes pancreatic endocrine cell formation. This work highlights the need to examine standard culture variables such as seeding density when optimizing hESC differentiation protocols. </p> </div

    High Cell Seeding Density Increases Pancreatic Progenitor Differentiation.

    No full text
    <p>(A) hESCs seeded at different densities were differentiated for 14 days and immunostained for PDX1 (green) and DNA (blue). (B) Single-cell quantification of PDX1 positive nuclei as a percentage of total nuclei (C) RT-qPCR of 21 day differentiated cells. Expression is shown relative to isolated human islets. Different superscripts (a, b, c) are significantly different from each other within each graph by one-way ANOVA with Bonferroni post-hoc test. Scale bars are 100 μm.</p

    Higher Cell Seeding Density Improves Definitive Endoderm Differentiation.

    No full text
    <p>(A) CA1S hESCs were differentiated using a protocol designed to mimic human development in a 21 day, 5 stage process. (B) hESCs were seeded onto matrigel-coated culture plates at the indicated density, yielding 30%-100% confluence as shown at 24 hours after seeding. (C) On day 4 of differentiation, markers of definitive endoderm induction were assessed by flow cytometry (CXCR4 and SOX17 expression) or RT-qPCR (<i>FOXA2</i> and Goosecoid, shown relative to undifferentiated hESC expression levels). (D) Expression of OCT4 (marker of pluripotent cells) was assessed by RT-qPCR and immunofluorescence as a percentage of the total number of nuclei (OCT4 is green, nuclei are blue). * represents significant difference from 1.3 x 10<sup>4</sup> cells/cm<sup>2</sup> by one-way ANOVA with Bonferroni post-hoc test. Different superscripts (a, b, c) are significantly different from each other within each graph by one-way ANOVA with Bonferroni post-hoc test. Scale bars are 100 μm. </p

    High Seeding Density Cultures Follow Expected Endocrine Developmental Timeline.

    No full text
    <p>(A) Media samples from multiple (N=12) high hESC cell seeding density differentiations contain reproducibly high levels of C-peptide and glucagon as measured by radioimmunoassay. (B) Over the differentiation time course expression of transcription factors and islet hormones was examined by RT-qPCR relative to adult human islet expression levels. (C) 21 day differentiated hESCs were immunostained as agarose-embedded, paraffinized sections for pancreatic hormones and key transcription factors involved in pancreatic endocrine induction and maturation. * represents p<0.05 comparing day 14 and 21 media content. Different superscripts (a, b, c) are significantly different from each other within each graph by one-way ANOVA with Bonferroni post-hoc test. Scale bars are 50 μm.</p

    Higher Cell Seeding Density Decreases Cell Cycle Progression.

    No full text
    <p>(A) A representative histogram (left) of low density (1.3 x 10<sup>4</sup> cells/cm<sup>2</sup>, black line) and high density (5.3 x 10<sup>4</sup> cells/cm<sup>2</sup>, red line) seeded CA1S hESCs stained for DNA content by propidium iodide to indicate cell cycle state within the depicted gates 24-hours after seeding. (B) Single cells gated for uniform DNA width were assessed in triplicate and quantified as either G0/G1, S or G2/M phases using the gates in (A) as a percentage of the total single cell population. Four cell seeding densities of CA1S cells (1.3, 2.6, 3.9 and 5.3 x 10<sup>4</sup> cells/cm<sup>2</sup>) along with 5.3 x 10<sup>4</sup> cells/cm<sup>2</sup> seeded cells treated overnight with 2% DMSO to induce cell cycle arrest (2% DMSO) were quantified. * represents significant difference from 1.3 x 10<sup>4</sup> cells/cm<sup>2</sup> by one-way ANOVA with Bonferroni post-hoc test within the same cell cycle population. (C) Representative images and quantification of immunocytochemistry of pRb S780 (green, nuclei are blue). pRb S780 positive mitotic cells were quantified as a percentage of the total cell populations in five randomly selected images. Different superscripts (a, b, c) are significantly different from each other by one-way ANOVA with Bonferroni post-hoc test. Scale bars are 100 μm.</p

    Higher Cell Seeding Density Enhances Pancreatic Endocrine Formation.

    No full text
    <p>(A) Insulin, glucagon, and somatostatin expression were assessed in 21 day differentiated hESCs using RT-qPCR (shown relative to human islets). (B) C-peptide and glucagon release were assayed in static 24 hour media samples take on the indicated culture, or during a sequential glucose and/or potassium chloride (KCl) stimulated hormone release assay performed on day 19 of culture. Following a 2 hour 2 mM glucose wash, cells were treated for 1 hour with 2 mM glucose (2G), 25 mM glucose (25G), then 30 mM KCl (30KCl). Diamonds, squares, triangles, and circles represent 1.3 x 10<sup>4</sup> cells/cm<sup>2</sup>, 2.6 x 10<sup>4</sup> cells/cm<sup>2</sup>, 3.9 x 10<sup>4</sup> cells/cm<sup>2</sup>, and 5.3 x 10<sup>4</sup> cells/cm<sup>2</sup> initial seeding density respectively. * represents p<0.05 comparing 5.3 x 10<sup>4</sup> cells/cm<sup>2</sup> with other cell densities. # represents p<0.05 comparing KCl stimulation versus other stimuli within the 5.3 x 10<sup>4</sup> cells/cm<sup>2</sup> seeding density. (C) hESCs seeded at different densities and differentiated for 21 days were agarose-embedded and immunostained for insulin (blue), glucagon (green), somatostatin (red) and DNA (cyan). Right panel shows hormone staining and left panel shows the same hormone image with DNA. See <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0082076#pone.0082076.s004" target="_blank">Figure S4</a> for single channel images of a larger field of view. (D) Single-cell quantification of hormone population showing the number of cells positive for insulin, glucagon, or somatostatin as a percentage of the total number of nuclei. (E) Single-cell polyhormonal analysis of the hormone positive population in C as a percentage of total hormone positive population. Triple indicates cells scored positive for all three hormones. * represents p<0.05 comparing triple positive populations of 5.3 x 10<sup>4</sup> cells/cm<sup>2</sup> vs 3.9 x 10<sup>4</sup> cells/cm<sup>2</sup>. In panels A and D, different superscripts (a, b) are significantly different from each other within each graph by one-way ANOVA with Bonferroni post-hoc test. Scale bars are 50 μm.</p
    corecore