We used mice lacking Abcc8, a key component of the β-cell KATP-channel, to analyze the effects of a sustained elevation in the intracellular Ca2+ concentration ([Ca2+]i) on β-cell identity and gene expression. Lineage tracing analysis revealed the conversion of β-cells lacking Abcc8 into pancreatic polypeptide cells but not to α- or δ-cells. RNA-sequencing analysis of FACS-purified Abcc8−/− β-cells confirmed an increase in Ppy gene expression and revealed altered expression of more than 4,200 genes, many of which are involved in Ca2+ signaling, the maintenance of β-cell identity, and cell adhesion. The expression of S100a6 and S100a4, two highly upregulated genes, is closely correlated with membrane depolarization, suggesting their use as markers for an increase in [Ca2+]i. Moreover, a bioinformatics analysis predicts that many of the dysregulated genes are regulated by common transcription factors, one of which, Ascl1, was confirmed to be directly controlled by Ca2+ influx in β-cells. Interestingly, among the upregulated genes is Aldh1a3, a putative marker of β-cell dedifferentiation, and other genes associated with β-cell failure. Taken together, our results suggest that chronically elevated β-cell [Ca2+]i in Abcc8−/− islets contributes to the alteration of β-cell identity, islet cell numbers and morphology, and gene expression by disrupting a network of Ca2+-regulated genes.

In type 2 diabetes (T2D), pancreatic β-cells fail to respond appropriately to metabolic stresses brought on by age, obesity, and genetic risk factors. The mechanisms by which chronic metabolic stress, including insulin resistance, glucotoxicity, and lipotoxicity (13), impair β-cell function are not understood. Although metabolic stress is usually considered to be exogenous to the β-cell, chronic stimulation leads to changes within the β-cell, impairing function. One such factor is chronic elevation in the concentration of intracellular Ca2+ ([Ca2+]i), sometimes called excitotoxicity (4), which may be triggered by sustained β-cell depolarization resulting from chronic stimulation.

Ca2+ is a ubiquitous second messenger that is central to regulating cellular dynamics of many cell types, including β-cells. Genetic and pharmacological perturbations that stimulate or impair Ca2+signaling have dramatic effects on β-cell function. For instance, the disruption of calcineurin, a Ca2+-dependent phosphatase, or Ca2+/calmodulin-dependent protein kinase II or IV, two Ca2+-dependent kinases, profoundly impairs β-cell function, likely by modulating the activity of Ca2+-responsive transcription factors such as NFAT, CREB, and TORC2 (59). Conversely, the constitutive activation of calcineurin or calmodulin, a Ca2+ binding protein, also causes marked β-cell dysfunction (3,10,11).

Acutely, glucose metabolism induces ATP-sensitive potassium (KATP) channel closure, membrane depolarization, opening of voltage-gated Ca2+channels, a rise in [Ca2+]i, and insulin secretion. However, sustained elevation in [Ca2+]i has multiple effects on β-cell function that can be adaptive or maladaptive. β-Cell proliferation induced by glucose metabolism (12) is an example of an adaptive response to sustained elevations in [Ca2+]i. However, chronically elevated [Ca2+]i can also induce maladaptive responses because prevention of Ca2+ influx in the setting of insulin resistance prevents β-cell death (13). In either case, mice lacking KATP channels exhibit disrupted islet morphology, characterized by α-cells being located in the islet core (14,15), suggesting loss of β-cell identity or impairments in cell adhesion.

Here, we show that β-cells in Abcc8−/− mice exhibit chronic membrane depolarization and a sustained elevation in [Ca2+]i and dysregulation of more than 4,200 genes, many of which are involved in cell adhesion, Ca2+binding and Ca2+signaling, and maintenance of β-cell identity. We also report that Abcc8−/− mice exhibit β-cell to pancreatic polypeptide (PP)–cell trans-differentiation and have increased expression of Aldh1a3, a gene recently suggested as a marker of dedifferentiating β-cells. In addition, we show that S100a6 and S100a4, two EF-hand Ca2+ binding proteins, are acutely regulated in β-cells by membrane depolarization agents, suggesting that they may be markers for β-cell excitotoxicity. Finally, we performed a computational analysis to predict components of the gene regulatory network that may govern the observed gene expression changes and found that one of the predicted regulators, Ascl1, is directly regulated by Ca2+ influx in β-cells.

Mouse Lines

All animal experimentation was approved by the Vanderbilt Institutional Animal Care and Use Committee. Abcc8−/− (Abcc8tm1.1Mgn, MGI: 2388392), RIP-Cre (Tg.Ins2-cre25Mgn, MGI: 2176227), and R26LSL.YFP (Gt [ROSA]26Sortm[EYFP]Cos, MGI: 2449038) mice were congenic C57BL/6 and genotyped as previously described (1618). MIP-GFP mice (Tg[Ins1-EGFP/GH1]14Hara, MGI: 3583654) were congenic CD-1 and genotyped as previously described (19).

Glucose Tolerance Testing

After a 16-h fast, male Abcc8+/+ and Abcc8−/− C57BL/6 mice were given intraperitoneal injection of d-glucose (2 mg/g body weight). Blood glucose was measured using a BD Logic glucometer.

Verapamil Administration

Adult Abcc8+/+;MIP-GFP and Abcc8−/−;MIP-GFP mice were given Splenda (2%) or a combination of verapamil (1 mg/mL; Sigma-Aldrich, V4629) and Splenda in their drinking water for 3 weeks. Splenda was used to mask the taste of verapamil.

Immunofluorescence Microscopy

Pancreata were fixed in 4% paraformaldehyde, frozen, and sectioned at a depth of 8 μm. Immunofluorescence staining was performed as previously described (20). Antibodies are listed in the Supplementary Data. Images were acquired using an Olympus FV-1000 confocal microscope, pseudocolored using ImageJ, and are representative of the phenotype observed in at least three animals. Cell death was determined using the In Situ Cell Death Detection Kit (Roche, 11684795910).

Islet Isolation

Pancreata were injected with 0.6 mg/mL collagenase P (Roche, 11213865001) into the pancreatic bile duct. Dissociated tissue was fractionated using Histopaque-1077 (Sigma-Aldrich, 10771), followed by hand-picking of islets. For FACS and RNA sequencing (RNA-Seq), islets from four to seven mice were pooled per sample. For quantitative RT-PCR (qRT-PCR), islets from a single mouse were used per sample.

Resting Membrane Potential

Islets were isolated from pancreata of 7- to 10-week-old Abcc8+/+;MIP-GFP and Abcc8−/−;MIP-GFP mice, and electrophysiological recordings were performed as previously described (21).

Ca2+ Imaging

Islets were isolated from pancreata of 9- to 11-week-old Abcc8+/+and Abcc8−/− mice, and imaging of cytoplasmic Ca was performed as previously described (22).

Islet Culture

Wild-type islets were incubated for 24 h in DMEM (Gibco, 11966-025) containing 5.6 mmol/L glucose, 10% FBS (Gibco, 16140–071), and 1% penicillin-streptomycin (Gibco, 15140-122). Experimental media contained 100 μmol/L tolbutamide (Sigma-Aldrich, T0891) or 20 mmol/L KCl (Sigma-Aldrich, P5405), with or without 50 μmol/L verapamil.

Cell Isolation

Islets were dissociated in Accumax (Sigma-Aldrich, A7089) containing 10 units/mL DNase (Invitrogen, AM2222). After filtration (35-μm strainer) and centrifugation, cells were resuspended in Flow Cytometry Buffer (R&D Systems, FC001) containing 2 units/mL DNase, 0.5 mol/L EDTA, and 7-aminoactinomycin D (1:1,000; ThermoFisher, A1310). GFP+/7-aminoactinomycin D cells were analyzed and isolated using an Aria II (BD Biosciences) and collected in Homogenization Solution (Promega, TM351; containing 1-thioglycerol).

RNA Purification and Quality Control

RNA was isolated from FACS-purified β-cells and whole islets using Maxwell 16 LEV simplyRNA Tissue Kit (Promega, TM351). RNA samples were analyzed using the Agilent 2100 Bioanalyzer. Only samples with an RNA integrity number >7 were used.

Library Assembly, Sequencing, and Analysis

RNA samples from FACS-purified β-cells were amplified using SMART-Seq v4 Ultra Low Input RNA Kit for Sequencing (Clontech, 634888; eight PCR cycles). cDNA libraries were constructed using the Low Input Library Prep Kit (Clontech, 634947). Paired-end sequencing of four replicates each of Abcc8+/+;MIP-GFP and Abcc8−/−;MIP-GFP samples was performed on an Illumina NextSeq 500 (75-nucleotide reads). Approximately 900 million raw reads were processed using TrimGalore 0.4.0. STAR (23) was used to align sequences to mm10 (Genome Reference Consortium Mouse Reference 38 [GRCm38]) and GENCODE comprehensive gene annotations (Release M8). HTSeq was used for counting reads mapped to genomic features (24), and DESeq2 was used for differential gene expression analysis (25).

Upstream Regulator Prediction

iRegulon (26) was used to predict gene regulatory networks. Default search parameters were used (20 kb centered on the transcription start site, 7 species conservation). The enrichment score threshold was 3.0.

qRT-PCR

The High Capacity cDNA Reverse Transcription Kit (ThermoFisher, 4368814) was used to convert whole-islet RNA to cDNA. qRT-PCR was performed on an Applied Biosystems 7900HT using 2× SYBR Green PCR Master Mix (ThermoFisher, 4309155). Samples were analyzed in triplicate and normalized to Hprt expression. Primer sequences are listed in the Supplementary Data.

Statistical Analysis

Statistical significance was determined using the two-tailed Student t test. Data are represented as mean ± SEM. A threshold of P < 0.05 was used to declare significance.

Abcc8−/− β-Cells Exhibit Persistent Membrane Depolarization and Elevated [Ca2+]i

Previous studies have shown that single β-cells from mice lacking KATP channels, in contrast to normal mice, exhibit chronically elevated [Ca2+]i and continuous action potential firing (16,27,28), whereas intact islets exhibit electrical bursting and Ca2+ oscillations (29,30). To definitively establish Abcc8−/− mice as a model for chronically elevated β-cell [Ca2+]i, we measured membrane potential at high (11 mmol/L) and low (2 mmol/L) glucose concentrations (Fig. 1A and B). Abcc8−/− β-cells exhibit action potentials in high glucose but fail to undergo membrane potential polarization in low glucose (Fig. 1B). Quantification of the changes in Abcc8−/− β-cell membrane potentials confirms that Abcc8−/− β-cells show no difference in membrane potential between high and low glucose, contrary to Abcc8+/+ β-cells (Fig. 1C and D). However, Abcc8−/− β-cells exhibit a chronic elevation in [Ca2+]i, as shown by Ca2+-imaging at both low and high glucose (Fig. 1E). Quantification of the area under the curve shows that Abcc8−/− β-cells have significantly higher [Ca2+]i than Abcc8+/+ β-cells at each time interval measured (Fig. 1F). In addition, by examining individual traces rather than averaged data and imaging over a longer period, we observe Ca2+ oscillations in Abcc8−/− islets under low glucose conditions, as previously reported (29,30), whereas Abcc8+/+ islets exhibit consistently low Ca2+ levels (Supplementary Fig. 1). This indicates that although Abcc8−/− β-cells exhibit variable and oscillating [Ca2+]i under euglycemic conditions, their mean [Ca2+]i is elevated compared with Abcc8+/+ β-cells.

Figure 1

Abcc8−/− β-cells exhibit persistent membrane depolarization and elevated [Ca2+]i. A perforated patch recording technique was used to monitor the membrane potential of Abcc8+/+ and Abcc8−/− β-cells in whole islets. A: Abcc8+/+ β-cells display normal activity at 11 mmol/L glucose and become hyperpolarized in response to 2 mmol/L glucose (representative recording). B: Abcc8−/− β-cells display activity similar to Abcc8+/+ β-cells at 11 mmol/L glucose (with a trend toward a depolarized plateau potential); however, their activity remains statistically unchanged in response to 2 mmol/L glucose (representative recording). C: Quantification of the potentials of Abcc8+/+ and Abcc8−/− β-cells at 11 mmol/L and 2 mmol/L glucose. Abcc8+/+ β-cells become significantly more polarized in response to 2 mmol/L glucose, but there is no difference between Abcc8−/− β-cells at 11 mmol/L and 2 mmol/L glucose. D: Quantification of the percentage change in membrane potential between 11 mmol/L and 2 mmol/L glucose for Abcc8+/+ and Abcc8−/− β-cells. The membrane potential of Abcc8+/+ β-cells changes significantly more than Abcc8−/− β-cells. E: [Ca2+]i was monitored in Abcc8+/+ and Abcc8−/− islets with Fura-2 acetoxymethyl ester. Islets were equilibrated in 2 mmol/L glucose, stimulated with 11 mmol/L glucose, and returned to 2 mmol/L glucose, as indicated by the bars above the traces (overall averages are shown). F: Area under the curve of [Ca2+]i was quantified at intervals representing low glucose conditions (0–2 min), glucose-stimulation (10–12 min), and a return to low glucose (20–22 min). Data are an average of n ≥ 21 islets from 3 animals. **P < 0.01; ***P < 0.001.

Figure 1

Abcc8−/− β-cells exhibit persistent membrane depolarization and elevated [Ca2+]i. A perforated patch recording technique was used to monitor the membrane potential of Abcc8+/+ and Abcc8−/− β-cells in whole islets. A: Abcc8+/+ β-cells display normal activity at 11 mmol/L glucose and become hyperpolarized in response to 2 mmol/L glucose (representative recording). B: Abcc8−/− β-cells display activity similar to Abcc8+/+ β-cells at 11 mmol/L glucose (with a trend toward a depolarized plateau potential); however, their activity remains statistically unchanged in response to 2 mmol/L glucose (representative recording). C: Quantification of the potentials of Abcc8+/+ and Abcc8−/− β-cells at 11 mmol/L and 2 mmol/L glucose. Abcc8+/+ β-cells become significantly more polarized in response to 2 mmol/L glucose, but there is no difference between Abcc8−/− β-cells at 11 mmol/L and 2 mmol/L glucose. D: Quantification of the percentage change in membrane potential between 11 mmol/L and 2 mmol/L glucose for Abcc8+/+ and Abcc8−/− β-cells. The membrane potential of Abcc8+/+ β-cells changes significantly more than Abcc8−/− β-cells. E: [Ca2+]i was monitored in Abcc8+/+ and Abcc8−/− islets with Fura-2 acetoxymethyl ester. Islets were equilibrated in 2 mmol/L glucose, stimulated with 11 mmol/L glucose, and returned to 2 mmol/L glucose, as indicated by the bars above the traces (overall averages are shown). F: Area under the curve of [Ca2+]i was quantified at intervals representing low glucose conditions (0–2 min), glucose-stimulation (10–12 min), and a return to low glucose (20–22 min). Data are an average of n ≥ 21 islets from 3 animals. **P < 0.01; ***P < 0.001.

Close modal

Interestingly, despite having chronically elevated β-cell [Ca2+]i,Abcc8−/− mice have long been known to have surprisingly small disturbances in their plasma glucose concentrations (16). First, and as we previously reported (31), they exhibit outright hypoglycemia in the fasted state (Supplementary Fig. 3A). Second, they exhibit a trend toward mild hyperglycemia in the fed state (Supplementary Fig. 3B).

β-Cell Identity Becomes Compromised in Abcc8−/− Mice

During prolonged metabolic stress, β-cells can lose expression of functional markers and convert to other endocrine cell types (32). However, β-cell dedifferentiation has not been studied in the context of chronically elevated [Ca2+]i. Thus, we performed β-cell lineage tracing using Abcc8−/−; RIP-Cre; Rosa26LSL.YFP/+ mice. Although there was no evidence for β- to α-cell or β- to δ-cell trans-differentiation (Supplementary Fig. 2A and B), we observed an increase in yellow fluorescent protein (YFP)/PP coexpression (Fig. 2C). Most YFP/PP coexpressing cells are polyhormonal, expressing both PP and insulin (Fig. 2A), but a few cells (0.24% of all YFP-expressing cells and 10% of YFP/PP-coexpressing cells) (Supplementary Fig. 2C) no longer express insulin (Fig. 2B). Administration of a Ca2+channel blocker to Abcc8−/− mice resulted in a lower number of insulin/PP-coexpressing cells, but the difference was not significant (Supplementary Fig. 2D). The loss of β-cell identity correlates with impaired glucose tolerance not attributable to β-cell death (Supplementary Fig. 3A and C). Complete dedifferentiation (e.g., YFP expression without hormone immunostaining), was only observed at a very low rate and was similar in both wild-type and knockout animals (0.19% and 0.21%, respectively) (Supplementary Fig. 2E).

Figure 2

Loss of strict β-cell identity in Abcc8−/− mice. We performed β-cell lineage tracing using Abcc8−/−;RIP-Cre;R26LSL.YFP/+ animals and assessed β-cell dedifferentiation at 12 weeks of age. A: Representative examples of polyhormonal cells coexpressing YFP, PP, and insulin. B: Representative examples of YFP-labeled cells that are expressing PP but not expressing insulin. C: Quantification of PP/YFP double+ cells shows an increase in their occurrence in Abcc8−/−;RIP-Cre; R26LSL.YFP/+ mice compared with Abcc8+/+;RIP-Cre; R26LSL.YFP/+ mice at 12 weeks of age. n = 3–4 animals, 10–15 islets counted per animal. Scale bars = 5 μm. *P < 0.05.

Figure 2

Loss of strict β-cell identity in Abcc8−/− mice. We performed β-cell lineage tracing using Abcc8−/−;RIP-Cre;R26LSL.YFP/+ animals and assessed β-cell dedifferentiation at 12 weeks of age. A: Representative examples of polyhormonal cells coexpressing YFP, PP, and insulin. B: Representative examples of YFP-labeled cells that are expressing PP but not expressing insulin. C: Quantification of PP/YFP double+ cells shows an increase in their occurrence in Abcc8−/−;RIP-Cre; R26LSL.YFP/+ mice compared with Abcc8+/+;RIP-Cre; R26LSL.YFP/+ mice at 12 weeks of age. n = 3–4 animals, 10–15 islets counted per animal. Scale bars = 5 μm. *P < 0.05.

Close modal

RNA Expression Profiling

To determine how chronically elevated β-cell [Ca2+]i affects gene expression, we performed RNA-Seq using FACS-purified β-cells from Abcc8−/−;MIP-GFP mice at 8–9 weeks of age. Because the human growth hormone mini-gene within the MIP-GFP allele causes both human growth hormone expression and activation of STAT5 signaling (33), we used MIP-GFP–expressing mice as controls. Principal component and gene clustering analyses (Supplementary Fig. 4B and C), performed on RNA-Seq data from FACS-purified cells (Supplementary Fig. 4A), indicates that most of the top 500 differentially expressed genes cluster as expected. Differential expression analysis (Fig. 3B and Supplementary Table 1) revealed 4,208 differentially expressed genes (2,152 downregulated and 2,056 upregulated) in Abcc8−/− β-cells (adjusted P < 0.05). Approximately 90% are protein coding, 3% are noncoding RNAs, 2% are pseudogenes, and 3% are other types of processed transcripts (Fig. 3A).

Figure 3

RNA-sequencing of Abcc8−/− β-cells. We performed RNA-sequencing of FACS-purified β-cells from Abcc8−/−;MIP-GFP and Abcc8+/+;MIP-GFP animals at 8–9 weeks of age. A: Pie chart shows the percentage of differentially regulated genes that fall into biotype categories of protein-coding, noncoding RNA (ncRNA), other RNA, pseudogene, other processed transcripts, and other types of transcripts. B: Volcano plot shows the most differentially expressed genes in Abcc8−/−;MIP-GFP β-cells based on the −log10 (false discovery rate [FDR]–adjusted P value) and the log2 (fold change). Genes with a log2 (fold change) greater than three are labeled and grouped into categories by biotype characterization. C: Selected differentially expressed genes in Abcc8−/−;MIP-GFP β-cells. Abcc8−/−;MIP-GFP β-cells exhibit a decrease in expression of genes associated with β-cell maturation/function, an increase in expression of genes associated with Ca2+signaling, Ca2+binding, or Ca2+-dependent gene expression, a decrease in expression of genes associated with cell adhesion, and an increase in genes that are enriched in a population of dedifferentiated β-cells, as indicated by the fold change in selected genes from our RNA-Seq data set. All genes shown were manually selected and have FDR-adjusted P values <0.05.

Figure 3

RNA-sequencing of Abcc8−/− β-cells. We performed RNA-sequencing of FACS-purified β-cells from Abcc8−/−;MIP-GFP and Abcc8+/+;MIP-GFP animals at 8–9 weeks of age. A: Pie chart shows the percentage of differentially regulated genes that fall into biotype categories of protein-coding, noncoding RNA (ncRNA), other RNA, pseudogene, other processed transcripts, and other types of transcripts. B: Volcano plot shows the most differentially expressed genes in Abcc8−/−;MIP-GFP β-cells based on the −log10 (false discovery rate [FDR]–adjusted P value) and the log2 (fold change). Genes with a log2 (fold change) greater than three are labeled and grouped into categories by biotype characterization. C: Selected differentially expressed genes in Abcc8−/−;MIP-GFP β-cells. Abcc8−/−;MIP-GFP β-cells exhibit a decrease in expression of genes associated with β-cell maturation/function, an increase in expression of genes associated with Ca2+signaling, Ca2+binding, or Ca2+-dependent gene expression, a decrease in expression of genes associated with cell adhesion, and an increase in genes that are enriched in a population of dedifferentiated β-cells, as indicated by the fold change in selected genes from our RNA-Seq data set. All genes shown were manually selected and have FDR-adjusted P values <0.05.

Close modal

Abcc8−/− β-Cells Exhibit Changes in Expression of Genes Involved in β-Cell Maturation, Ca2+ Signaling, Cell Adhesion, and Dedifferentiation

To correlate gene expression with the functional abnormalities of Abcc8−/− islets, we examined genes known to be highly enriched in mature β-cells and observed that Ins1, Slc2a2, Neurod1, Gck, Glp1r, Npy, several synaptotagmins, Tph1, Tph2, and Egfr are all downregulated (Fig. 3C). Conversely, Ppy is upregulated in Abcc8−/− β-cells, consistent with our observation of polyhormonal cells.

Because Abcc8−/− β-cells have a persistent increase in [Ca2+]i, we examined genes with well-defined roles in Ca2+ binding or signaling, such as Myo7a, a Ca2+/calmodulin-binding myosin motor protein, Pcp4, a calmodulin binding protein known to protect neurons from Ca2+-induced toxicity (34), and Cacng3, a subunit of a voltage-dependent Ca2+ channel, and found their expression was elevated. Similarly, we observed that Nfatc1, Mef2c, and Mef2d, three calcium-regulated transcription factors; Camk1d, Camkk1, and Camkk2, three kinases involved in calcium-signaling; and S100a1, S100a3, S100a4, S100a6, and S100a13, five EF-hand Ca2+ binding proteins, were also upregulated (Fig. 3C).

Quantitation of islet cell types in Abcc8−/− mice revealed fewer β-cells, more α-, δ-, and PP cells, and a progressive redistribution of non–β-endocrine cells to the islet core (Supplementary Fig. 5). To correlate gene expression with this disrupted islet architecture, we examined expression of cell adhesion molecules, hypothesizing that a reduction in such genes could result in loss of islet structure. Consistently, we observed a reduction in multiple cell adhesion molecules, including Cldn1, Cldn3, Cldn8, Cldn23, and Ocln, genes involved in tight junctions; Cdh1, Cdh7, Cdh8, Cdh13, and Cdh18, encoding cadherins; Cdhr1, a cadherin-related protein; Pcdhb15 and Pcdhb22, encoding protocadherins; and Vcam1, a vascular cell adhesion molecule (Fig. 3C).

Aldh1a3, a retinaldehyde dehydrogenase recently suggested to be a marker for β-cell dedifferentiation (35), is 27-fold upregulated in Abcc8−/− β-cells by RNA-Seq (Fig. 3C). In addition, six other genes (Serpina7, Aass, Asb11, Penk, Fabp3, and Tcea1) enriched in dedifferentiated β-cells (35) are upregulated in Abcc8−/− β-cells (Fig. 3C). Coimmunostaining with insulin indicates that although ALDH1A3 is expressed in only 0.4% of Abcc8+/+ β-cells, it is present in 29.1% of Abcc8−/− β-cells (Fig. 4A and B). However, this fraction was reduced to 11.4% in Abcc8−/− mice administered the Ca2+ channel blocker verapamil (Fig. 4A and B), further suggesting that a chronic increase in [Ca2+]i impairs the maintenance of cell identity.

Figure 4

Heterogeneous expression of S100A6 and ALDH1A3. A: Coimmunostaining of ALDH1A3, a potent marker of dedifferentiated β-cells, and insulin in pancreatic sections from Abcc8+/+, Abcc8−/−, and Abcc8−/− mice given verapamil. B: Quantification of A showing the percentage of insulin (Ins) and ALDH1A3 coexpressing cells in each group. Verapamil treatment partially rescues the percentage of ALDH1A3-expressing β-cells in Abcc8−/− mice. C: Coimmunostaining of S100A6 and insulin in Abcc8+/+ and Abcc8−/− pancreatic sections. D: Quantification of C showing the percentage of insulin-expressing cells that coexpress S100A6 in Abcc8+/+ and Abcc8−/− mice. E: Coimmunostaining of ALDH1A3 and S100A6 shows that there is not strict colocalization of these two factors, again emphasizing the heterogeneous nature of β-cell failure. The arrows indicate cells that coexpress ALDH1A3 and S100A6. n = 3 animals, 8–12 islets counted per animal. Scale bars = 50 μm. *P < 0.05; **P < 0.01; ***P < 0.001.

Figure 4

Heterogeneous expression of S100A6 and ALDH1A3. A: Coimmunostaining of ALDH1A3, a potent marker of dedifferentiated β-cells, and insulin in pancreatic sections from Abcc8+/+, Abcc8−/−, and Abcc8−/− mice given verapamil. B: Quantification of A showing the percentage of insulin (Ins) and ALDH1A3 coexpressing cells in each group. Verapamil treatment partially rescues the percentage of ALDH1A3-expressing β-cells in Abcc8−/− mice. C: Coimmunostaining of S100A6 and insulin in Abcc8+/+ and Abcc8−/− pancreatic sections. D: Quantification of C showing the percentage of insulin-expressing cells that coexpress S100A6 in Abcc8+/+ and Abcc8−/− mice. E: Coimmunostaining of ALDH1A3 and S100A6 shows that there is not strict colocalization of these two factors, again emphasizing the heterogeneous nature of β-cell failure. The arrows indicate cells that coexpress ALDH1A3 and S100A6. n = 3 animals, 8–12 islets counted per animal. Scale bars = 50 μm. *P < 0.05; **P < 0.01; ***P < 0.001.

Close modal

S100a6 and S100a4 Are Markers of Excitotoxicity in β-Cells

To identify genes that could serve as markers of β-cell excitotoxicity, we analyzed S100a6 and S100a4. Both genes are appealing targets because of their functions as EF-hand Ca2+ binding proteins, the association of S100a6 with insulin secretion (36), and the increased expression of members of the S100 gene family in islets from humans with hyperglycemia (37). Moreover, they are among the most highly upregulated genes in Abcc8−/− β-cells, with S100a6 and S100a4 increasing 37- and 5-fold, respectively. S100A6 is only expressed in 2.8% of Abcc8+/+ β-cells but is expressed in 38.4% of Abcc8−/− β-cells (Fig. 4C and D). In addition, qRT-PCR using whole-islet RNA confirms the upregulation of S100a6 and S1004 in Abcc8−/− islets (Fig. 5A).

Figure 5

S100a6 and S100a4 serve as markers of excitotoxicity in β-cells. A: qRT-PCR using whole-islet RNA confirms upregulation of S100a6 and S100a4 in Abcc8−/− islets compared with Abcc8+/+ islets. BE: qRT-PCR using whole-islet RNA from wild-type islets treated with 100 μmol/L tolbutamide or 20 mmol/L KCl, with or without 50 μmol/L verapamil, for 24 h. S100a6 (B and C) and S100a4 (D and E) are significantly upregulated in response to membrane depolarization, but this effect is negated when Ca2+ influx is blocked. F and G: qRT-PCR using whole-islet RNA from animals administered verapamil (1 mg/mL) in the drinking water for 3 weeks indicates that expression of both S100a6 and S100a4 is significantly downregulated when Ca2+ influx is inhibited. n.s. = not significant. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001.

Figure 5

S100a6 and S100a4 serve as markers of excitotoxicity in β-cells. A: qRT-PCR using whole-islet RNA confirms upregulation of S100a6 and S100a4 in Abcc8−/− islets compared with Abcc8+/+ islets. BE: qRT-PCR using whole-islet RNA from wild-type islets treated with 100 μmol/L tolbutamide or 20 mmol/L KCl, with or without 50 μmol/L verapamil, for 24 h. S100a6 (B and C) and S100a4 (D and E) are significantly upregulated in response to membrane depolarization, but this effect is negated when Ca2+ influx is blocked. F and G: qRT-PCR using whole-islet RNA from animals administered verapamil (1 mg/mL) in the drinking water for 3 weeks indicates that expression of both S100a6 and S100a4 is significantly downregulated when Ca2+ influx is inhibited. n.s. = not significant. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001.

Close modal

To determine whether the observed changes in S100a6 and S100a4 expression in Abcc8−/− β-cells are the result of chronically elevated [Ca2+]i and not some other mechanism, we treated wild-type islets with KCl or tolbutamide and found that membrane depolarization is associated with an increase in both S100a6 and S100a4 expression (Fig. 5B–E), mirroring the expression pattern in Abcc8−/− β-cells. These changes were reversed when islets were treated with both a depolarizing agent and the Ca2+channel inhibitor verapamil (Fig. 5B–E). Moreover, the expression of S100a6 and S100a4 was decreased when Ca2+ influx is pharmacologically inhibited in Abcc8−/− mice (Fig. 5F and G). These results suggest that S100a6 and S100a4 expression in Abcc8−/− β-cells is tightly correlated with [Ca2+]i. Finally, coimmunostaining with S100A6 and ALDH1A3 revealed that 15.4 ± 2.1% of insulin-expressing cells in Abcc8−/− mice express both S100A6 and ALDH1A3 (Fig. 4E).

Prediction of Upstream Regulators

To determine whether differentially expressed genes in Abcc8−/− β-cells might share common upstream regulators, we used iRegulon, a program that identifies common DNA binding motifs in coexpressed genes (26). Analysis of the top 500 upregulated genes revealed binding sites for ASCL1, CEBPG, and RARG in many genes, with some genes, including Aldh1a3, containing binding sites for all three of these transcriptional regulators (Fig. 6A, Supplementary Table 2, and Supplementary Fig. 6). Interestingly, all three of these predicted regulators are upregulated in Abcc8−/− β-cells (Fig. 6C). Conversely, analysis of the top 500 downregulated genes predicts binding sites for TEAD1, HNF1A, and ZFP647 (Fig. 6B, Supplementary Table 3, and Supplementary Fig. 7). However, the expression of these regulators is unchanged in Abcc8−/− β-cells (Fig. 6C).

Figure 6

Upstream regulator prediction using iRegulon. Using the top 500 up- and downregulated genes in Abcc8−/− β-cells, we used iRegulon to predict common upstream regulators based on enriched DNA-binding motifs. Tables summarize the top three predicted regulators of upregulated (A) and downregulated (B) genes, their enrichment scores, and the number of predicted targets. C: Expression of the predicted regulators in Abcc8−/− β-cells, as determined by RNA-Seq. D: qRT-PCR using whole-islet RNA confirms upregulation of Ascl1 in Abcc8−/− islets compared with Abcc8+/+ islets. E: qRT-PCR for Ascl1 using whole-islet RNA from wild-type islets treated with 20 mmol/L KCl, with or without 50 μmol/L verapamil, for 24 h. F: qRT-PCR using whole-islet RNA from animals administered verapamil. n.s. = not significant. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001.

Figure 6

Upstream regulator prediction using iRegulon. Using the top 500 up- and downregulated genes in Abcc8−/− β-cells, we used iRegulon to predict common upstream regulators based on enriched DNA-binding motifs. Tables summarize the top three predicted regulators of upregulated (A) and downregulated (B) genes, their enrichment scores, and the number of predicted targets. C: Expression of the predicted regulators in Abcc8−/− β-cells, as determined by RNA-Seq. D: qRT-PCR using whole-islet RNA confirms upregulation of Ascl1 in Abcc8−/− islets compared with Abcc8+/+ islets. E: qRT-PCR for Ascl1 using whole-islet RNA from wild-type islets treated with 20 mmol/L KCl, with or without 50 μmol/L verapamil, for 24 h. F: qRT-PCR using whole-islet RNA from animals administered verapamil. n.s. = not significant. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001.

Close modal

Ascl1 Is Regulated by [Ca2+]i in β-Cells

Because Ascl1 is 22-fold upregulated in Abcc8−/−;MIP-GFP β-cells (Fig. 3B) and 24-fold upregulated in Abcc8−/− islets (Fig. 6D) and has been shown by chromatin immunoprecipitation (ChIP) sequencing or ChIP PCR to bind near 51% of the predicted targets (3840), we studied its responsiveness to changes in [Ca2+]i in isolated islets. Consistent with Ascl1 being regulated by [Ca2+]i, its expression increases in response to membrane depolarization, an effect that is reversed when Ca2+ influx is inhibited by verapamil, both in isolated islets (Fig. 6E) and in mice (Fig. 6F). These results support the idea that ASCL1 could play a central role in regulating gene expression in β-cells with chronically elevated [Ca2+]i.

Abcc8−/− β-Cells Exhibit Elevated Basal [Ca2+]i

Abcc8−/− β-cells have been previously shown to have altered [Ca2+]i homeostasis, as would be expected in the absence of KATP channels (16,2730). We similarly observed that Abcc8−/− β-cells exhibit both persistent depolarization and an elevated mean [Ca2+]i compared with Abcc8+/+ islets (Fig. 1). Interestingly, although Abcc8−/− β-cells exhibit Ca2+ oscillations in substimulatory glucose (Supplementary Fig. 1), as has been previously reported (29,30), we did not consistently observe oscillations in membrane potential, as has been observed in β-cells lacking KATP channels. This may be the result of the short duration of the recordings or to our use of the patch-clamp technique rather than intracellular microelectrodes, which limits the detection of oscillations in membrane potential (29). Regardless, Abcc8−/− β-cells experience more persistent membrane depolarization and elevated mean [Ca2+]i compared with wild-type β-cells, and the downstream effects on gene expression are likely to be the same. We also observed a transient drop in [Ca2+]i in Abcc8−/− islets after glucose stimulation (Fig. 1E) that was not seen in Abcc8+/+ islets. This drop is the result of brief membrane hyperpolarization caused by transient activation of the sodium potassium ATPase (29) and has been observed previously in islets treated with KATP channel inhibitors (41) as well as in other KATP channel–deficient mice (29).

Abcc8−/− β-Cells Exhibit Dysregulated Gene Expression

By performing RNA-Seq on purified pancreatic β-cell populations from both Abcc8−/− and Abcc8+/+ mice, we found that the expression of 4,208 genes was perturbed. Some of these genes, including S100a6, Myo7a, Pcp4, Cacng3, Mef2c, and Camk1d, are known to be involved in calcium binding and signaling (Fig. 3C). The S100 gene family, for instance, modulates the activity of other proteins on calcium binding (42). Moreover, S100A6 specifically promotes Ca2+-stimulated insulin release (36). Camk1d, Camkk1, and Camkk2, three protein kinases, Myo7a, a myosin motor protein, and Mef2d and Mef2c, are involved in Ca2+/calmodulin-dependent signaling or are regulated by [Ca2+]i (43,44). However, the alteration of some Ca2+-regulated genes does not imply that all of the gene expression changes we observed are caused by changes in [Ca2+]i. Although we have demonstrated that changes in S100a6, S100a4, and Ascl1 expression are tightly linked to membrane potential (Figs. 5B and E and 6E), a similar analysis has not been performed for the remaining genes. Thus, although it is likely that a substantial fraction of the dysregulated genes are the result of altered [Ca2+]i homeostasis, other mechanisms, such as paracrine and/or neuronal signaling, may also be involved. Additional studies are necessary to determine which changes are directly attributable to chronically elevated [Ca2+]i and which are the result of other signaling mechanisms and/or compensatory adaptations.

Compromised β-Cell Identity in Abcc8−/− Mice

Recent studies have suggested that β-cell dedifferentiation contributes to the development of T2D (32,35). Among the genes affected in Abcc8−/− β-cells are many known to be involved in maintaining β-cell identity and/or function, such as Ins1, Slc2a2, Neurod1, Gck, and Syt10 (Fig. 3C). However, some of the transcription factors previously associated with β-cell dedifferentiation (32), including Mafa, Pdx1, Nkx6.1, FoxO1, Ngn3, Oct4, and Nanog, are unchanged. This difference may explain the modest loss of β-cell identity observed in Abcc8−/− mice, with only 2.21% of β-cells undergoing trans-differentiation to insulin/PP polyhormonal or PP monohormonal cells (Fig. 2C). Although treatment with a Ca2+ channel blocker did not statistically reverse the insulin/PP polyhormonal cells, its use resulted in a decrease in the percentage of Abcc8−/− β-cells expressing the dedifferentiation marker ALDH1A3 (Fig. 4B), further suggesting that a chronic increase in [Ca2+]i may contribute to loss of β-cell identity. In this regard, it is important to distinguish our results, which reflect the loss of KATP channels, from studies using KATP channel gain-of-function mutants (45,46). The current study examines the effects of elevated [Ca2+]i in a nearly euglycemic setting, whereas the latter reflects decreased [Ca2+]i in a hyperglycemic setting.

Two Potential Genetic Markers for Elevated [Ca2+]i

Our studies identified two genes, S100a6 and S100a4, that were highly upregulated in Abcc8−/− β-cells and were also acutely regulated by treatment with depolarizing agents and a Ca2+channel blocker in isolated islets (Fig. 5B and E). The administration of a Ca2+ channel blocker to Abcc8−/− mice only partially rescued the expression of S100a6 or S100a4 (Fig. 5F and G), but this may be the result of inadequate dosing or a treatment duration that was too short to overcome the effects of the Abcc8 gene knockout. However, it is also possible that these genes are only partially regulated by Ca2+ influx in vivo. Regardless, our data strongly suggest that S100a6 and S100a4 are regulated by Ca2+ in the β-cell and that they may serve as markers for β-cell excitotoxicity. In support of this, members of the S100 gene family, including S100A3, S100A6, S100A10, S100A11, and S100A16, have been associated with hyperglycemia in human islets (37), and S100A6 was specifically shown to be positively correlated with BMI in β-cells from donors with T2D (47). Neither gene was altered in two recent single-cell sequencing studies of people with T2D (47,48), but the sequencing depth was very shallow.

Effects of a Sustained Increase in [Ca2+]i on Islet Morphology

Perturbations in Ca2+ signaling also provide a compelling explanation for the disrupted islet morphology observed in KATP-channel knockout mice (Supplementary Fig. 5). [Ca2+]i is required for maintenance of tight junctions and adherens junctions, and focal adhesion disassembly occurs in response to elevated [Ca2+]i (49). Accordingly, among the downregulated genes are many that encode cell adhesion molecules, such as Ocln, Tln1, and Cdh1 (Fig. 3C), suggesting that a Ca2+-dependent gradual breakdown of cell-to-cell contacts may be responsible for the altered islet morphology.

Identification of Putative Upstream Regulators

Bioinformatics analysis of genes whose expression was increased in the Abcc8−/− mice identified ASCL1, CEBPG, and RARG as possible upstream regulators (Fig. 6A). ASCL1, also known as MASH1, is important for neuronal commitment and differentiation (50). Retinoic acid receptors, including RARG, play a role in both endocrine cell development and maintenance of proper insulin secretion and β-cell mass (51). Finally, CEBPB, a closely related protein to CEBPG, represses the insulin promoter under conditions of chronically elevated glucose, and its accumulation in β-cells increases vulnerability to endoplasmic reticulum stress (52,53). Although our computational analysis is purely predictive, it is partially validated by the fact that 51% of the ASCL1 target genes and 60% of the RARG binding sites were established by ChIP-Seq or ChIP-PCR (3840,54). Importantly, S100a4 is among the ASCL1 targets that have been experimentally validated (3840), and our in-depth analysis of Ascl1 strongly supports its regulation by [Ca2+]i, consistent with our model. Conversely, analysis of genes that are downregulated by [Ca2+]i predicts binding sites for TEAD1, HNF1A, and ZFP647 (Fig. 6B). Although Tead1 gene expression is unaffected in Abcc8−/− β-cells, the activity of TEAD1, a member of the Hippo pathway that interacts with YAP/TAZ to promote proliferation, is directly inhibited by Ca2+ (55). Furthermore, because a gene knockout of Hnf1a results in β-cell failure and diabetes (56), a predicted decrease in its activity could explain the decrease in genes involved in β-cell function in Abcc8−/− β-cells.

Does Excitotoxicity Contribute to β-Cell Failure?

Our studies, together with findings of others (15,16,2730), indicate that the absence of functional KATP channels in Abcc8−/− mice causes a chronic elevation of β-cell [Ca2+]i, an impairment of β-cell identity, alterations in islet cell numbers and morphology, and very significant perturbations in β-cell gene expression. Although the elevation of [Ca2+]i in Abcc8−/− β-cells, caused by the total absence of KATP-channels, is persistent and severe, a less severe or persistent elevation in mean [Ca2+]i may also occur in patients with prediabetes or T2D, when hyperglycemia chronically stimulates insulin secretion, or when a sulfonylurea is given as therapy for T2D. Interestingly, although Abcc8−/− mice exhibit impaired glucose tolerance at 12 weeks of age, they do not go on to develop T2D, at least in the time frame studied here. This suggests that the impairments in β-cell identity and islet morphology are insufficient themselves to cause β-cell failure. However, β-cell excitotoxicity, acting in combination with other metabolic stresses, may accelerate the loss of functional β-cells, perhaps by dedifferentiation or glucolipotoxicity. Regardless, our findings provide new insights into transcriptional responses that occur within β-cells when they are subjected to chronically elevated [Ca2+]i and suggest a model (Fig. 7) that involves the coordinated interactions of a network of Ca2+-responsive genes and upstream transcriptional modulators. This network may exist to balance both the positive and negative effects that [Ca2+]i is known to have on β-cell function and mass.

Figure 7

Model shows the effects of chronically elevated [Ca2+]i in the β-cell. Our results suggest that there is a putative gene regulatory network mediating the effects of chronically elevated [Ca2+]i in the β-cell. First, the effects of elevated [Ca2+]i may be at least partly mediated through the predicted regulator Ascl1, causing an elevation in Ca2+-dependent gene expression changes. Second, an elevated [Ca2+]i may also contribute to the loss of β-cell function and stable β-cell identity, as evidenced by elevation in Aldh1a3 expression as well as the presence of polyhormonal cells. These effects mediated by chronically elevated [Ca2+]i combined with other metabolic stresses may contribute to β-cell failure in T2D.

Figure 7

Model shows the effects of chronically elevated [Ca2+]i in the β-cell. Our results suggest that there is a putative gene regulatory network mediating the effects of chronically elevated [Ca2+]i in the β-cell. First, the effects of elevated [Ca2+]i may be at least partly mediated through the predicted regulator Ascl1, causing an elevation in Ca2+-dependent gene expression changes. Second, an elevated [Ca2+]i may also contribute to the loss of β-cell function and stable β-cell identity, as evidenced by elevation in Aldh1a3 expression as well as the presence of polyhormonal cells. These effects mediated by chronically elevated [Ca2+]i combined with other metabolic stresses may contribute to β-cell failure in T2D.

Close modal

RNA-Seq data are available in ArrayExpress (www.ebi.ac.uk/arrayexpress) under accession number E-MTAB-4726.

Acknowledgments. The authors thank the Vanderbilt Islet Procurement and Analysis Core for performing multiple islet isolations, Vanderbilt Technologies for Advanced Genomics (VANTAGE) for performing cell sorting and RNA-Seq, the Vanderbilt Cell Imaging Shared Resource for helping to acquire images, the Vanderbilt Tissue Pathology Shared Resource for processing and sectioning paraffin-embedded tissues, and the Vanderbilt Transgenic Mouse/ESC Shared Resource for reconstituting mice with the Abcc8 null allele. The authors also thank Guoqiang Gu and Roland Stein of Vanderbilt University for reading the manuscript and Jody Peters, Katherine Boyer, and Lauren Stiehle, also of Vanderbilt University, for technical assistance.

Funding. These studies were partly supported by National Institutes of Health/National Institute of Diabetes and Digestive and Kidney Diseases grants DK-72473 and DK-89523 to M.A.M. and DK-020593 to Vanderbilt Islet Procurement and Analysis Core, Vanderbilt Cell Imaging Shared Resource, and Vanderbilt Transgenic Mouse/ESC Shared Resource and by National Cancer Institute grant CA-68485 to Vanderbilt Cell Imaging Shared Resource and Vanderbilt Transgenic Mouse/ESC Shared Resource.

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. J.S.S. and M.A.M. designed the study and wrote the manuscript. J.S.S., H.W.C., and J.T.O. performed experiments and analyzed data. J.-P.C. performed RNA-Seq data processing, alignment, and differential gene expression analysis and edited the manuscript. M.T.D., P.K.D., and D.A.J. performed membrane potential measurements and calcium imaging and analyzed the data. A.B.O. contributed to study design and edited the manuscript. M.A.M. supervised and funded the experiments. M.A.M. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

1.
Muoio
DM
,
Newgard
CB
.
Mechanisms of disease: molecular and metabolic mechanisms of insulin resistance and beta-cell failure in type 2 diabetes
.
Nat Rev Mol Cell Biol
2008
;
9
:
193
205
[PubMed]
2.
Poitout
V
,
Robertson
RP
.
Glucolipotoxicity: fuel excess and beta-cell dysfunction
.
Endocr Rev
2008
;
29
:
351
366
[PubMed]
3.
Tornovsky-Babeay
S
,
Dadon
D
,
Ziv
O
, et al
.
Type 2 diabetes and congenital hyperinsulinism cause DNA double-strand breaks and p53 activity in β cells
.
Cell Metab
2014
;
19
:
109
121
[PubMed]
4.
Arundine
M
,
Tymianski
M
.
Molecular mechanisms of calcium-dependent neurodegeneration in excitotoxicity
.
Cell Calcium
2003
;
34
:
325
337
[PubMed]
5.
Heit
JJ
,
Apelqvist
AA
,
Gu
X
, et al
.
Calcineurin/NFAT signalling regulates pancreatic beta-cell growth and function
.
Nature
2006
;
443
:
345
349
[PubMed]
6.
Soleimanpour
SA
,
Crutchlow
MF
,
Ferrari
AM
, et al
.
Calcineurin signaling regulates human islet beta-cell survival
.
J Biol Chem
2010
;
285
:
40050
40059
[PubMed]
7.
Dadi
PK
,
Vierra
NC
,
Ustione
A
,
Piston
DW
,
Colbran
RJ
,
Jacobson
DA
.
Inhibition of pancreatic β-cell Ca2+/calmodulin-dependent protein kinase II reduces glucose-stimulated calcium influx and insulin secretion, impairing glucose tolerance
.
J Biol Chem
2014
;
289
:
12435
12445
[PubMed]
8.
Screaton
RA
,
Conkright
MD
,
Katoh
Y
, et al
.
The CREB coactivator TORC2 functions as a calcium- and cAMP-sensitive coincidence detector
.
Cell
2004
;
119
:
61
74
[PubMed]
9.
Blanchet
E
,
Van de Velde
S
,
Matsumura
S
, et al
.
Feedback inhibition of CREB signaling promotes beta cell dysfunction in insulin resistance
.
Cell Reports
2015
;
10
:
1149
1157
[PubMed]
10.
Epstein
PN
,
Overbeek
PA
,
Means
AR
.
Calmodulin-induced early-onset diabetes in transgenic mice
.
Cell
1989
;
58
:
1067
1073
[PubMed]
11.
Bernal-Mizrachi
E
,
Cras-Méneur
C
,
Ye
BR
,
Johnson
JD
,
Permutt
MA
.
Transgenic overexpression of active calcineurin in beta-cells results in decreased beta-cell mass and hyperglycemia
.
PLoS One
2010
;
5
:
e11969
[PubMed]
12.
Porat
S
,
Weinberg-Corem
N
,
Tornovsky-Babaey
S
, et al
.
Control of pancreatic β cell regeneration by glucose metabolism
.
Cell Metab
2011
;
13
:
440
449
[PubMed]
13.
Xu
G
,
Chen
J
,
Jing
G
,
Shalev
A
.
Preventing β-cell loss and diabetes with calcium channel blockers
.
Diabetes
2012
;
61
:
848
856
[PubMed]
14.
Shiota
C
,
Rocheleau
JV
,
Shiota
M
,
Piston
DW
,
Magnuson
MA
.
Impaired glucagon secretory responses in mice lacking the type 1 sulfonylurea receptor
.
Am J Physiol Endocrinol Metab
2005
;
289
:
E570
E577
[PubMed]
15.
Winarto
A
,
Miki
T
,
Seino
S
,
Iwanaga
T
.
Morphological changes in pancreatic islets of KATP channel-deficient mice: the involvement of KATP channels in the survival of insulin cells and the maintenance of islet architecture
.
Arch Histol Cytol
2001
;
64
:
59
67
[PubMed]
16.
Shiota
C
,
Larsson
O
,
Shelton
KD
, et al
.
Sulfonylurea receptor type 1 knock-out mice have intact feeding-stimulated insulin secretion despite marked impairment in their response to glucose
.
J Biol Chem
2002
;
277
:
37176
37183
[PubMed]
17.
Postic
C
,
Shiota
M
,
Niswender
KD
, et al
.
Dual roles for glucokinase in glucose homeostasis as determined by liver and pancreatic beta cell-specific gene knock-outs using Cre recombinase
.
J Biol Chem
1999
;
274
:
305
315
[PubMed]
18.
Srinivas
S
,
Watanabe
T
,
Lin
CS
, et al
.
Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus
.
BMC Dev Biol
2001
;
1
:
4
[PubMed]
19.
Hara
M
,
Wang
X
,
Kawamura
T
, et al
.
Transgenic mice with green fluorescent protein-labeled pancreatic beta -cells
.
Am J Physiol Endocrinol Metab
2003
;
284
:
E177
E183
[PubMed]
20.
Burlison
JS
,
Long
Q
,
Fujitani
Y
,
Wright
CV
,
Magnuson
MA
.
Pdx-1 and Ptf1a concurrently determine fate specification of pancreatic multipotent progenitor cells
.
Dev Biol
2008
;
316
:
74
86
[PubMed]
21.
Vierra
NC
,
Dadi
PK
,
Jeong
I
,
Dickerson
M
,
Powell
DR
,
Jacobson
DA
.
Type 2 diabetes-associated K+ channel TALK-1 modulates β-cell electrical excitability, second-phase insulin secretion, and glucose homeostasis
.
Diabetes
2015
;
64
:
3818
3828
[PubMed]
22.
Dadi
PK
,
Vierra
NC
,
Jacobson
DA
.
Pancreatic β-cell-specific ablation of TASK-1 channels augments glucose-stimulated calcium entry and insulin secretion, improving glucose tolerance
.
Endocrinology
2014
;
155
:
3757
3768
[PubMed]
23.
Dobin
A
,
Davis
CA
,
Schlesinger
F
, et al
.
STAR: ultrafast universal RNA-seq aligner
.
Bioinformatics
2013
;
29
:
15
21
[PubMed]
24.
Anders
S
,
Pyl
PT
,
Huber
W
.
HTSeq--a Python framework to work with high-throughput sequencing data
.
Bioinformatics
2015
;
31
:
166
169
[PubMed]
25.
Love
MI
,
Huber
W
,
Anders
S
.
Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2
.
Genome Biol
2014
;
15
:
550
[PubMed]
26.
Janky
R
,
Verfaillie
A
,
Imrichová
H
, et al
.
iRegulon: from a gene list to a gene regulatory network using large motif and track collections
.
PLoS Comput Biol
2014
;
10
:
e1003731
[PubMed]
27.
Miki
T
,
Nagashima
K
,
Tashiro
F
, et al
.
Defective insulin secretion and enhanced insulin action in KATP channel-deficient mice
.
Proc Natl Acad Sci U S A
1998
;
95
:
10402
10406
[PubMed]
28.
Seghers
V
,
Nakazaki
M
,
DeMayo
F
,
Aguilar-Bryan
L
,
Bryan
J
.
Sur1 knockout mice. A model for K(ATP) channel-independent regulation of insulin secretion
.
J Biol Chem
2000
;
275
:
9270
9277
[PubMed]
29.
Düfer
M
,
Haspel
D
,
Krippeit-Drews
P
,
Aguilar-Bryan
L
,
Bryan
J
,
Drews
G
.
Oscillations of membrane potential and cytosolic Ca(2+) concentration in SUR1(-/-) beta cells
.
Diabetologia
2004
;
47
:
488
498
[PubMed]
30.
Nenquin
M
,
Szollosi
A
,
Aguilar-Bryan
L
,
Bryan
J
,
Henquin
JC
.
Both triggering and amplifying pathways contribute to fuel-induced insulin secretion in the absence of sulfonylurea receptor-1 in pancreatic beta-cells
.
J Biol Chem
2004
;
279
:
32316
32324
[PubMed]
31.
De León
DD
,
Li
C
,
Delson
MI
,
Matschinsky
FM
,
Stanley
CA
,
Stoffers
DA
.
Exendin-(9-39) corrects fasting hypoglycemia in SUR-1-/- mice by lowering cAMP in pancreatic beta-cells and inhibiting insulin secretion
.
J Biol Chem
2008
;
283
:
25786
25793
[PubMed]
32.
Talchai
C
,
Xuan
S
,
Lin
HV
,
Sussel
L
,
Accili
D
.
Pancreatic β cell dedifferentiation as a mechanism of diabetic β cell failure
.
Cell
2012
;
150
:
1223
1234
[PubMed]
33.
Brouwers
B
,
de Faudeur
G
,
Osipovich
AB
, et al
.
Impaired islet function in commonly used transgenic mouse lines due to human growth hormone minigene expression
.
Cell Metab
2014
;
20
:
979
990
[PubMed]
34.
Kanazawa
Y
,
Makino
M
,
Morishima
Y
,
Yamada
K
,
Nabeshima
T
,
Shirasaki
Y
.
Degradation of PEP-19, a calmodulin-binding protein, by calpain is implicated in neuronal cell death induced by intracellular Ca2+ overload
.
Neuroscience
2008
;
154
:
473
481
[PubMed]
35.
Kim-Muller
JY
,
Fan
J
,
Kim
YJ
, et al
.
Aldehyde dehydrogenase 1a3 defines a subset of failing pancreatic β cells in diabetic mice
.
Nat Commun
2016
;
7
:
12631
[PubMed]
36.
Okazaki
K
,
Niki
I
,
Iino
S
,
Kobayashi
S
,
Hidaka
H
.
A role of calcyclin, a Ca(2+)-binding protein, on the Ca(2+)-dependent insulin release from the pancreatic beta cell
.
J Biol Chem
1994
;
269
:
6149
6152
[PubMed]
37.
Fadista
J
,
Vikman
P
,
Laakso
EO
, et al
.
Global genomic and transcriptomic analysis of human pancreatic islets reveals novel genes influencing glucose metabolism
.
Proc Natl Acad Sci U S A
2014
;
111
:
13924
13929
[PubMed]
38.
Castro
DS
,
Martynoga
B
,
Parras
C
, et al
.
A novel function of the proneural factor Ascl1 in progenitor proliferation identified by genome-wide characterization of its targets
.
Genes Dev
2011
;
25
:
930
945
[PubMed]
39.
Webb
AE
,
Pollina
EA
,
Vierbuchen
T
, et al
.
FOXO3 shares common targets with ASCL1 genome-wide and inhibits ASCL1-dependent neurogenesis
.
Cell Reports
2013
;
4
:
477
491
[PubMed]
40.
Raposo
AA
,
Vasconcelos
FF
,
Drechsel
D
, et al
.
Ascl1 coordinately regulates gene expression and the chromatin landscape during neurogenesis
.
Cell Reports
2015
;
9
:
1544
1556
[PubMed]
41.
Mourad
NI
,
Nenquin
M
,
Henquin
JC
.
Metabolic amplification of insulin secretion by glucose is independent of β-cell microtubules
.
Am J Physiol Cell Physiol
2011
;
300
:
C697
C706
[PubMed]
42.
Kligman
D
,
Hilt
DC
.
The S100 protein family
.
Trends Biochem Sci
1988
;
13
:
437
443
[PubMed]
43.
Mellström
B
,
Savignac
M
,
Gomez-Villafuertes
R
,
Naranjo
JR
.
Ca2+-operated transcriptional networks: molecular mechanisms and in vivo models
.
Physiol Rev
2008
;
88
:
421
449
[PubMed]
44.
Udovichenko
IP
,
Gibbs
D
,
Williams
DS
.
Actin-based motor properties of native myosin VIIa
.
J Cell Sci
2002
;
115
:
445
450
[PubMed]
45.
Wang
Z
,
York
NW
,
Nichols
CG
,
Remedi
MS
.
Pancreatic β cell dedifferentiation in diabetes and redifferentiation following insulin therapy
.
Cell Metab
2014
;
19
:
872
882
[PubMed]
46.
Brereton
MF
,
Iberl
M
,
Shimomura
K
, et al
.
Reversible changes in pancreatic islet structure and function produced by elevated blood glucose
.
Nat Commun
2014
;
5
:
4639
[PubMed]
47.
Segerstolpe
Å
,
Palasantza
A
,
Eliasson
P
, et al
.
Single-cell transcriptome profiling of human pancreatic islets in health and type 2 diabetes
.
Cell Metab
2016
;
24
:
593
607
[PubMed]
48.
Xin
Y
,
Kim
J
,
Okamoto
H
, et al
.
RNA sequencing of single human islet cells reveals type 2 diabetes genes
.
Cell Metab
2016
;
24
:
608
615
[PubMed]
49.
Giannone
G
,
Rondé
P
,
Gaire
M
, et al
.
Calcium rises locally trigger focal adhesion disassembly and enhance residency of focal adhesion kinase at focal adhesions
.
J Biol Chem
2004
;
279
:
28715
28723
[PubMed]
50.
Pang
ZP
,
Yang
N
,
Vierbuchen
T
, et al
.
Induction of human neuronal cells by defined transcription factors
.
Nature
2011
;
476
:
220
223
[PubMed]
51.
Brun
PJ
,
Grijalva
A
,
Rausch
R
, et al
.
Retinoic acid receptor signaling is required to maintain glucose-stimulated insulin secretion and β-cell mass
.
FASEB J
2015
;
29
:
671
683
[PubMed]
52.
Lu
M
,
Seufert
J
,
Habener
JF
.
Pancreatic beta-cell-specific repression of insulin gene transcription by CCAAT/enhancer-binding protein beta. Inhibitory interactions with basic helix-loop-helix transcription factor E47
.
J Biol Chem
1997
;
272
:
28349
28359
[PubMed]
53.
Matsuda
T
,
Kido
Y
,
Asahara
S
, et al
.
Ablation of C/EBPbeta alleviates ER stress and pancreatic beta cell failure through the GRP78 chaperone in mice
.
J Clin Invest
2010
;
120
:
115
126
[PubMed]
54.
Moutier
E
,
Ye
T
,
Choukrallah
MA
, et al
.
Retinoic acid receptors recognize the mouse genome through binding elements with diverse spacing and topology
.
J Biol Chem
2012
;
287
:
26328
26341
[PubMed]
55.
Thompson
M
,
Andrade
VA
,
Andrade
SJ
, et al
.
Inhibition of the TEF/TEAD transcription factor activity by nuclear calcium and distinct kinase pathways
.
Biochem Biophys Res Commun
2003
;
301
:
267
274
[PubMed]
56.
Pontoglio
M
,
Sreenan
S
,
Roe
M
, et al
.
Defective insulin secretion in hepatocyte nuclear factor 1alpha-deficient mice
.
J Clin Invest
1998
;
101
:
2215
2222
[PubMed]
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at http://www.diabetesjournals.org/content/license.

Supplementary data