Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2018 Jul 11;9(1):2681.
doi: 10.1038/s41467-018-04918-x.

Discovery of a drug candidate for GLIS3-associated diabetes

Affiliations

Discovery of a drug candidate for GLIS3-associated diabetes

Sadaf Amin et al. Nat Commun. .

Abstract

GLIS3 mutations are associated with type 1, type 2, and neonatal diabetes, reflecting a key function for this gene in pancreatic β-cell biology. Previous attempts to recapitulate disease-relevant phenotypes in GLIS3-/- β-like cells have been unsuccessful. Here, we develop a "minimal component" protocol to generate late-stage pancreatic progenitors (PP2) that differentiate to mono-hormonal glucose-responding β-like (PP2-β) cells. Using this differentiation platform, we discover that GLIS3-/- hESCs show impaired differentiation, with significant death of PP2 and PP2-β cells, without impacting the total endocrine pool. Furthermore, we perform a high-content chemical screen and identify a drug candidate that rescues mutant GLIS3-associated β-cell death both in vitro and in vivo. Finally, we discovered that loss of GLIS3 causes β-cell death, by activating the TGFβ pathway. This study establishes an optimized directed differentiation protocol for modeling human β-cell disease and identifies a drug candidate for treating a broad range of GLIS3-associated diabetic patients.

PubMed Disclaimer

Conflict of interest statement

The authors have filed a patent of the differentiation strategy. The authors declare no other competing interests.

Figures

Fig. 1
Fig. 1
Generation of mono-hormonal pancreatic β-like cells through the induction of late-stage pancreatic progenitors (PP2). a Schematic representation of the stepwise differentiation protocol. b qRT-PCR analysis of pancreatic progenitor markers in hESCs, definitive endoderm (DE), PP1 and PP2 cells (ES, DE n = 3, PP1 n = 6, PP2 n = 8). c Heatmap representing relative expression profiles of genes related to β-cell development in PP1 and PP2 cells (PP1 n = 2, PP2 n = 3). d Immunocytochemistry analysis of insulin (INS) and glucagon (GCG) expression at D16_E and D30_L, insets show a higher magnification image. Scale bar = 100 µm. Intracellular flow cytometry analysis (e) and quantification (f) of INS and GCG expression at D16_E and D30_L (D16_E n = 4, D30_L n = 8). g Immunocytochemistry analysis of cells at D30_L. Scale bar = 100 µm, scale bar of high magnification insets = 40 µm. h Heatmap representing the relative expression level of endocrine markers in PP1-β and PP2-β cells. i GSEA analysis shows that PP2-β cells are transcriptionally closer to human adult β-cells than PP1-β cells. The adult β cells_UP and adult β cells_DN gene sets consist of the top 1000 differentially expressed genes (higher expression for UP and lower expression for DN) in primary human β-cells compared to PP1-β cells. j FPKM values of GLIS3 in PH-β cells derived using Zhu et al.’s protocol and the purified INS-GFP+ PP1-β (n = 4) and PP2-β cells (n = 2). k Glucose-stimulated c-peptide secretion of cells at D30_L and human islets. The amount of c-peptide secretion in 20 mM (high) D-glucose condition was normalized to the amount of c-peptide secreted in 2 mM (low) D-glucose condition (n = 6 for cells at D30_L and n = 10 for islets). l Insulin secretion of cells at D30_L in response to other secretagogues, including 30 mM KCl (n = 7), 30 µM Forskolin (n = 5), or 10 mM Arginine (n = 5) relative to basal Krebs–Ringer bicarbonate HEPES (KRBH) buffer treatment (n = 7). The fold change was normalized to the amount of c-peptide secreted in KRBH condition. P values by unpaired two-tailed t-test were *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. Data are presented as individual biological replicates. The center value is “mean”. Error bar is SEM
Fig. 2
Fig. 2
Biallelic mutation of GLIS3 affects pancreatic differentiation and the generation of endocrine cells. a qRT-PCR analysis of pancreatic markers in the isogenic WT and GLIS3−/− PP2 cells. (PDX1, NEUROD1, NKX6.1 n = 4, MAFA n = 12). b Heatmap representing the relative expression levels of endocrine genes in WT and GLIS3−/− PP2 cells (n = 3). c GSEA analysis showing the decrease of endocrine pancreas-related genes in GLIS3−/− PP2 cells. Gene ontology (GO) analysis (d) and KEGG pathway analysis (e) of genes significantly downregulated (P < 0.01) in GLIS3−/− PP2 cells. f Flow cytometry analysis of WT and GLIS3−/− cells at D30_L. g Quantification of the percentage of INS-GFP+ cells of WT (n = 6) and GLIS3−/− (n = 9) cells at D30_L. The percentage of INS-GFP+ cells was quantified based on flow cytometry analysis of GFP+ cells. h Quantification of the percentage of INS+ cells of WT (n = 11) and GLIS3−/− (n = 14) cells at D30_L. The percentage was quantified based on intracellular flow cytometry analysis of INS+ cells. i Total percentage of endocrine cells in isogenic WT (n = 10) and GLIS3−/− (n = 12) cells at D30_L. The percentage of endocrine cells is calculated as the sum of the percentages of INS+, GCG+, SST+, and GHRL+ cells. j Plot representing the ratios of different endocrine subtypes in WT and GLIS3−/− cells at D30_L. k Immunocytochemistry analysis of pancreatic endocrine marker expression in WT and GLIS3−/− cells at D30_L. Scale bar = 100 μm. l, m Histogram showing fluorescence intensity (l) and quantification of median fluorescence values (m) of INS staining of WT and GLIS3−/− PP2-β cells (n = 4). n Insulin content of the purified INS-GFP+ WT and GLIS3−/− PP2-β cells (n = 4). Data are normalized to the WT mean value. P values by unpaired two-tailed t-test were *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. The center value is “mean”. Error bar is SEM
Fig. 3
Fig. 3
Loss of GLIS3 leads to increased cell death in PP2 and PP2-β cells. a Quantification of early apoptotic cells (the percentage of Annexin V+/DAPI cells) in WT and GLIS3−/− ES, DE, PP1, and PP2 cells (n = 3). b Representative flow cytometry analysis plots of Annexin V staining in WT and GLIS3−/− cells at D23_L. c Immunostaining for PDX1 and cleaved caspase-3 in WT and GLIS3−/− cells at D23_L. Scale bar = 40 μm. Annexin V staining (d) and quantification (e) of early apoptotic cells in the INS-GFP+ cells at D30_L (n = 6). Histogram showing fluorescence intensity (f) and quantification of median fluorescence values (g) of Annexin V staining of WT and GLIS3−/− INS+ DAPIPP2-β cells (WT n = 4, GLIS3−/− n = 6). h PI, cleaved caspase-3 and INS staining of WT and GLIS3−/− cells at D30_L. Scale bar = 40 μm. i Quantification of cell death rate (the percentage of PI+INS+ cells in INS+ cells) and apoptosis rate (the percentage of cleaved caspase-3+INS+ cells in INS+ cells) of WT and GLIS3−/− INS+ PP2-β cells (n = 3). j Schematic representation of the in vivo transplantation experiment. k Immunostaining for INS, cleaved caspase-3 and STEM121 in the grafts of mice transplanted with WT or GLIS3−/− cells. Scale bar = 100 μm. l Quantification of the apoptosis rate (the percentage of cleaved caspase-3+/ STEM121+ cells in STEM121+ cells) within WT and GLIS3−/− grafts (n = 7 for WT, n = 4 for GLIS3−/−). m Quantification of the percentage of apoptotic INS+ cells (CAS3+PDX1+STEM121+) in the INS+ population within the WT and GLIS3−/− grafts (INS+STEM121+, WT n = 7, GLIS3−/− n = 4). CAS3: cleaved caspase-3. P values by unpaired two-tailed Student’s t-test were *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. The center value is “mean”. Error bar is SEM
Fig. 4
Fig. 4
A high-content chemical screen identifies galunisertib as a drug candidate to rescue cell death induced by loss of GLIS3 both in vitro and in vivo. a Schematic representation of the high-content chemical screen. b Chemical structure of galunisertib. c Inhibitory curve of galunisertib. d Immunocyto-chemistry analysis of GLIS3−/− PP2-β cells treated with DMSO or 10 µM galunisertib. Scale bar = 100 μm, scale bar of high magnification insets = 40 μm. e, f Quantification of the cell death rate (e, the percentage of PI+INS+ cells in INS+ cells, n = 4) and apoptosis rate (f, the percentage of cleaved caspase-3+INS+ cells in INS+ cells, n = 3) of GLIS3−/− PP2-β cells treated with DMSO or 10 µM galunisertib. Flow cytometry analysis (g) and quantification (h) of early apoptotic cells (the percentage of Annexin V+/DAPI cells) in GLIS3−/− INS-GFP+ PP2-β cells treated with DMSO or 10 μM galunisertib (n = 6). i Relative number of INS+ cells in GLIS3−/− PP2-β cells treated with DMSO or 10 μM galunisertib. Data are normalized to DMSO-treated values (n = 4). j Schematic representation of the in vivo transplantation and drug treatment experiments. k Immunohistochemistry analysis of INS, cleaved caspase-3, and STEM121 in the grafts isolated from vehicle- or galunisertib-treated mice. Scale bar = 100 μm. l Quantification of immunohistochemistry data in j (n = 7). m Quantification of the percentage of apoptotic INS+ cells (CAS3+INS+ STEM121+) in the INS+ population within the grafts from vehicle- or galunisertib-treated mice (INS+STEM121+, n = 6). CAS3: cleaved caspase-3. P values by unpaired two-tailed t-test were *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. The center value is “mean”. Error bar is SEM
Fig. 5
Fig. 5
Galunisertib rescues loss of GLIS3 induced cell death by inhibiting TGFβ signaling. KEGG pathway analysis (a) and gene ontology (GO) analysis (b) of genes that are significantly (P < 0.01) upregulated in GLIS3−/− PP2 cells. Western blot analysis (c) and quantification (d, n = 3) of SMAD2/3 phosphorylation in WT and GLIS3−/− cells at D23_L. e Heatmap representing the relative expression levels of TGF-β-related genes upregulated at least two-fold in the purified WT and GLIS3−/− INS-GFP+ PP2-β cells. Western blotting analysis (f) and quantification (g, n = 3) of SMAD2/3 phosphorylation in GLIS3−/− cells at D30_L treated with DMSO or 10 μM galunisertib. h, i Quantification of the cell death rate (h, the percentage of PI+INS+ cells in INS+ cells) and apoptosis rate (i, the percentage of cleaved caspase-3+INS+ cells in INS+ cells) of GLIS3−/− PP2-β cells treated with different TGFβ inhibitors (n = 4). CAS3: cleaved caspase-3. P values by unpaired two-tailed t-test were *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. The center value is “mean”. Error bar is SEM

References

    1. Barrett JC, et al. Genome-wide association study and meta-analysis find that over 40 loci affect risk of type 1 diabetes. Nat. Genet. 2009;41:703–707. doi: 10.1038/ng.381. - DOI - PMC - PubMed
    1. Steck AK, et al. Improving prediction of type 1 diabetes by testing non-HLA genetic variants in addition to HLA markers. Pediatr. Diabetes. 2014;15:355–362. doi: 10.1111/pedi.12092. - DOI - PMC - PubMed
    1. Winkler C, et al. Feature ranking of type 1 diabetes susceptibility genes improves prediction of type 1 diabetes. Diabetologia. 2014;57:2521–2529. doi: 10.1007/s00125-014-3362-1. - DOI - PubMed
    1. Dupuis J, et al. New genetic loci implicated in fasting glucose homeostasis and their impact on type 2 diabetes risk. Nat. Genet. 2010;42:105–116. doi: 10.1038/ng.520. - DOI - PMC - PubMed
    1. Cho YS, et al. Meta-analysis of genome-wide association studies identifies eight new loci for type 2 diabetes in east Asians. Nat. Genet. 2011;44:67–72. doi: 10.1038/ng.1019. - DOI - PMC - PubMed

Publication types

MeSH terms