Mutations in the gene encoding the transcription factor regulatory factor X-box binding 6 (RFX6) are associated with human diabetes. Within pancreatic islets, RFX6 expression is most abundant in islet α-cells, and α-cell RFX6 expression is altered in diabetes. However, the roles of RFX6 in regulating gene expression, glucagon output, and other crucial human adult α-cell functions are not yet understood. We developed a method for selective genetic targeting of human α-cells and assessed RFX6-dependent α-cell function. RFX6 suppression with RNA interference led to impaired α-cell exocytosis and dysregulated glucagon secretion in vitro and in vivo. By contrast, these phenotypes were not observed with RFX6 suppression across all islet cells. Transcriptomics in α-cells revealed RFX6-dependent expression of genes governing nutrient sensing, hormone processing, and secretion, with some of these exclusively expressed in human α-cells. Mapping of RFX6 DNA-binding sites in primary human islet cells identified a subset of direct RFX6 target genes. Together, these data unveil RFX6-dependent genetic targets and mechanisms crucial for regulating adult human α-cell function.

Article Highlights
  • RFX6 is expressed in all islet endocrine cell types and is dysregulated in multiple forms of diabetes, but its function has not yet been delineated in α-cells.

  • We used specific targeting of shRNA-mediated suppression of RFX6 in primary human α-cells to unveil glucagon secretion phenotypes.

  • RFX6 is required in adult human α-cells to maintain gene regulation and hallmark functions, including regulated glucagon secretion.

  • RNA-sequencing and cleavage under targets and release using nuclease studies reveal distinct RFX6 genetic targets in adult human α- and β-cells.

Dysfunction of pancreatic islet cells is a hallmark of diabetes. Previous studies have identified islet-enriched transcription factors (TFs) crucial for regulation and maintenance of islet cell subtypes, especially insulin-secreting β-cells and glucagon-secreting α-cells. One such TF is regulatory factor X-box binding 6 (RFX6), a conserved winged helix TF expressed in all pancreatic endocrine cells (18).

RFX6 is required for pancreatic islet cell development and insulin production in zebrafish, mice, and humans (3,58). Human RFX6 mutations can result in monogenic diseases, including Mitchell-Riley syndrome, neonatal diabetes, and a subset of maturity onset diabetes of the young (RFX6-MODY) (3,912). RFX6 expression is also dysregulated in human β- and α-cells in type 1 diabetes and type 2 diabetes (T2D) (13,14).

Despite higher expression in human α-cells than in β-cells, the function of RFX6 in α-cells remains poorly understood (15). Homozygous RFX6-null mice exhibit phenotypes consistent with human Mitchell-Riley syndrome and patients with RFX6-MODY, including reduction of islet cells expressing insulin, glucagon, somatostatin, or ghrelin. However, these mice die soon after birth, precluding studies of RFX6 function in mature α-cells (6). Heterozygous RFX6-null mice do not develop diabetes, despite the association of heterozygous RFX6 mutations with MODY in humans (6,12). Previous studies have investigated the effects of β-cell–specific Rfx6 knockout in mice, revealing dysregulated insulin secretion and calcium channel activity (3,5). However, analogous studies of selective α-cell RFX6 targeting have not been reported. Moreover, differences between mouse and human islets in cytoarchitecture, gene regulation, and function necessitate research in human models to comprehensively understand diabetes pathogenesis (1618). While RFX6 function has been studied in the human β-cell line EndoC-βH2, human α-cell lines have not yet been generated (3). These challenges and knowledge gaps motivated our studies of RFX6 function in α-cells from primary human islets. Here we innovated an approach for selective lentiviral targeting of adult human α-cells to study the impact of acute RFX6 loss using transplantation-based assays, RNA-sequencing (RNA-seq), cleavage under targets and release using nuclease (CUT&RUN) genomic mapping, and patch clamp electrophysiology (2,4,1921). These studies reveal that RFX6 is required in human α-cells to maintain gene regulation and hallmark functions.

See Supplementary Table 1 for all reagent catalog numbers.

Analysis of Single-Cell RNA-Seq Data From Human Pancreas Analysis Program

Human Pancreas Analysis Program (HPAP-T2D) organ procurement and processing were performed as previously described (22). SmartSeq2 single-cell RNA-seq fastq files from 21 donors (10 T2D, 11 non-T2D) were obtained from PANC-DB. Alignment to the human genome (Genome Reference Consortium Human Build 38 with External RNA Controls Consortium sequences) was performed using STAR, and gene counts were determined using htseq-count (intersection-nonempty, Geographical Topic Phrases annotation) with Ensembl 97 release genes (23,24). Cells with <1,000 genes detected or <80,000 mapped reads were excluded. Gene expression was normalized to counts per million after removal of counts corresponding to External RNA Controls Consortium spike-ins and log-transformed after addition of a pseudocount. Data analyses, including normalization, scaling, and clustering, used Seurat v3.1.1 following the developer's manual (https://github.com/satijalab/seurat), unless otherwise noted (25,26).

Human Islet Procurement

Islets from previously healthy subjects were procured via the Integrated Islet Distribution Program, Alberta Disease Institute IsletCore, University of San Francisco Islet Core, Stanford Diabetes Center Islet Core, National Diabetes Research Institute, and International Institute for the Advancement of Medicine. See Supplementary Table 2 for donor information.

Lentivirus Production

Commercial GIPZ human lentiviral RFX6 shRNA, scramble control, and RFX6-FLAG overexpression constructs were obtained from Dharmacon. Lentiviruses were produced by transient transfection of HEK293T cells with the vectors and packaging constructs. Supernatants were collected and concentrated using PEG-it and then stored at −80°C prior to transduction of primary human cells.

Whole Pseudoislet Generation

Pseudoislet generation was performed as previously described in detail (4).

MagSep Pseudoislet Generation

Islets were dissociated as with whole pseudoislet generation. Prior to lentiviral transfection, α-cells were isolated via the EasySep Release Human PE Positive Selection kit using a phycoerythrin (PE) anti-human CD26 antibody, following the manufacturer’s protocol. CD26+ cells were transduced with 1 × 109 viral units/mL lentivirus and cultured separately from untransduced CD26neg cells for 4 days in an ultra-low attachment 96-well plate. Cells were then dissociated and seeded into a 24-well AggreWell400 plate at ∼2 × 105 cells/well, following the manufacturer’s protocol. MagSep pseudoislets were cultured overnight before further analysis.

RNA Extraction and Quantitative RT-PCR

RNA extraction and quantitative RT-PCR (qRT-PCR) were performed as previously described in detail (2,4). TaqMan probes are listed in Supplementary Table 1.

Patch Clamp Electrophysiology

MagSep pseudoislets shipped overnight from Stanford were dissociated into single cells and cultured in 35-mm dishes in DMEM with 10% FBS and 1% penicillin-streptomycin for up to 2 days. Only cells positive for green fluorescent protein (GFP) were patched to confirm knockdown, while cell identity was confirmed by post hoc immunostaining for glucagon. Electrophysiological measurements of voltage-dependent exocytosis were performed as previously described in detail (27).

For measurement of calcium channel activity using previously described methods (27), barium was used as a charge carrier. The extracellular solution contained (in mmol/L): 105 NaCl, 5.6 KCl, 20 BaCl2, 1.2 MgCl2, 10 HEPES, and 5 tetraethylammonium-Cl (pH 7.4 with NaOH). The intracellular solution contained (in mmol/L): 130 CsCl, 5 tetraethylammonium-Cl, 1 MgCl2, 10 HEPES, 10 EGTA, 4 MgATP, and 1 Li guanosine-5′-triphosphate (pH 7.15 with CsOH). Currents were measured 30 s after obtaining the whole-cell configuration by 100-ms depolarizations from −50 mV with 10 mV increases up to 60 mV. Currents were normalized to the cell size (pico amperes/pico farad [pA/pF]). Data were analyzed using FitMaster (SmartEphys HEKA) and Prism (GraphPad Software).

In Vitro Glucose-Stimulated Glucagon and Insulin Secretion Assays

Batches of 25 pseudoislets (Supplementary Table 2) were used for in vitro secretion assays, as previously described (2,4). Pseudoislets were equilibrated at 7 mmol/L glucose (glucagon secretion) or 2.8 mmol/L glucose (insulin secretion), then incubated at 7 mmol/L and 1 mmol/L glucose (glucagon secretion) or 2.8 mmol/L, 16.7 mmol/L, and 16.7 mmol/L glucose plus 3-isobutyl-1-methylxanthine (insulin secretion) for 60 min each. Supernatants collected at the end of each incubation and pseudoislet lysates were quantified by glucagon or human insulin ELISA kits. Results are presented as a percentage of total glucagon or insulin content. Assays were conducted in RPMI 1640 media supplemented with 1% FBS and the above glucose concentrations.

Mice

We performed experiments using sex- and age-matched mice. Sex was not considered a factor in the statistical analysis of the data. GKO-NSG mice were generated and raised in the Stanford Animal Facility and genotyped as previously described (21).

Human Islet Transplantation

Human pseudoislets (n = 1,000) (Supplementary Table 2) suspended in Matrigel were transplanted into the left renal capsular space of host mice, as previously described (2,4). Transplant recipients were 8-week-old GKO-NSG mice (sex- and age-matched within experiments) and were anesthetized using ketamine/xylazine.

Insulin Tolerance Tests

Mice received intraperitoneal insulin injections at 0.5 units insulin/kg body weight at least 1 month after transplantation to allow for engraftment and detection of glucagon. Tail vein blood samples were collected at 0, 15, 30, 45, and 60 min after injection. Serum glucagon levels were measured by glucagon ELISA.

Cell Surface Staining and FACS of Human Islet Cells

Pseudoislets were dispersed into single cells and stained with LIVE/DEAD Fixable Near-IR Dead Cell Stain Kit. Cells were then labeled with the following conjugated antibodies: Alexa Fluor 647-ectonucleoside triphosphate diphosphohydrolase-3 (AF647-NTPDase3; 1:50), PE-CD26 (1:100), PE-Texas Red-CD45 (1:100), and PE-Cy7-HPi2 (1:100). Incubations were conducted at 4°C for 30 min. Labeled cells were sorted on a special order five-laser FACS Aria II using a 100-µm nozzle with appropriate controls and doublet removal. Sorted cells were collected into low retention tubes containing cell staining buffer supplemented with Ribolock RNase inhibitor. Cytometry data were analyzed using FlowJo 10.8 software.

Bulk RNA-Seq and Data Analysis

RNA from ∼5,000 sorted live α- or β-cells having RNA integrity number >7 were used for RNA-seq library construction. SMART-seq v4 Ultra Low input RNA kit amplified cDNA, and Nextera XT DNA Library Preparation kit generated RNA-seq libraries. Barcoded libraries were multiplexed and sequenced as paired-end 150-base pair reads on the Illumina NovaSeq 6000 platform. Sixteen libraries were generated from four different donors for αRFX6 knockdown (αRFX6kd), βRFX6kd, and respective controls.

RNA-seq analysis used FastQC v0.11.9 for quality control. All libraries had >30 million reads and Trim Galore! v0.6.5 trimmed barcodes. STAR v2.7.9a aligned reads to the human genome (Genome Reference Consortium human build 38) (23). RSEM v1.3.1 quantified transcripts per million (TPM) (28). DESeq2 identified differentially expressed genes (P-adjusted cutoff: 0.1) (29). Gene set enrichment analysis used DAVID v2002q3 and g:Profiler e106_eg_p16_65fcd96 (30,31).

CUT&RUN and Data Analysis

Extracted nuclei from 500,000 dispersed RFX6-FLAG pseudoislet cells per condition were processed using CUTANA ChIC/CUT&RUN protocol v3.1 (10-min RT incubation with proteins pAG-MNase, 2-h nutation at 4°C with 100 mmol/L CaCl2). Libraries were prepared with the CUTANA CUT&RUN Library Prep Kit, according to manual instructions. Sequencing on the NovaSeq 6000 platform generated paired-end 150-base pair reads (Novogene; >25 million reads). The CUT&RUNtools pipeline (32) performed read trimming (Trimmomatic), alignment (Bowtie2), and peak calling (HOMER’s macs2.narrow outputs) (3335). Genome browser tracks, peak calling, and motif identification (using “makeUCSCfile,” “findPeaks,” and “findMotifs” commands, respectively) were performed. Motif enrichment P values were generated by HOMER, and gene annotation was generated with the GREAT algorithm using the default parameters (36).

Other Statistical Analyses and Data Visualization

Figure legends provide information on the number of replicates, standard deviation, and statistical analyses for qRT-PCR, glucose-stimulated insulin secretion (GSIS)/glucose-suppression of glucagon secretion (GSGS), intraperitoneal insulin tolerance test (IP-ITT), and area under the curve. GraphPad Prism v9.1 and R v4.1.1 were used for graphing and statistical analyses. ComplexHeatmap in R was used to generate heat maps. Browser tracks were generated with the University of California Santa Cruz genome browser (37). Biorender.com was used for method graphics.

Study Approvals

All studies were conducted with deidentified human islets and are therefore not considered as human subject research by Stanford University Institutional Review Board guidelines. All animal experiments and methods were approved by the Stanford Administrative Panel on Laboratory Animal Care (APLAC 29985).

Data and Resource Availability

The data sets generated and analyzed during the current study were uploaded to the National Center for Biotechnology Information’s Gene Expression Omnibus (GSE248854) (38). Our studies used GKO-NSG mice from The Jackson Laboratory (Research Resource Identifier: International Mouse Strain Resource [IMSR]_JAX: 029819).

Selective Genetic Targeting Reveals RFX6 Roles in Adult Human Islet α-Cells

RFX6 is expressed in all endocrine islet cells and most abundantly in α-cells, as determined from single-cell RNA-seq (Research Design and Methods) (Fig. 1A). These data demonstrate that the proportion of islet α- and β-cells expressing RFX6 is also reduced from organ donors with T2D (Fig. 1B). To investigate RFX6 function in human islet cells, we used lentiviral vectors coexpressing a GFP reporter to achieve shRNA-mediated suppression of RFX6 (hereafter, RFX6kd); a nontargeting lentivirus was used as a control (hereafter, “control”) (Fig. 1C, Supplementary Fig. 1, and Supplementary Table 2). After dispersion, primary islet cells were infected with lentivirus, then reaggregated to form pseudoislets (hereafter, whole RFX6kd pseudoislets), allowing subsequent molecular, cellular, and functional analyses (Fig. 1D) (2,4). We assessed in vitro GSGS in whole RFX6kd pseudoislets 5 days after lentiviral transduction. GSGS data were normalized to total pseudoislet glucagon content. Whole RFX6kd pseudoislets did not exhibit dysregulated glucagon secretion compared with controls, despite clear suppression of RFX6 mRNA and protein (Fig. 1E and Supplementary Fig. 1A and B).

Figure 1

RFX6 shRNA-mediated knockdown can be selectively targeted to CD26+ α-cells. A: Violin plot of single-cell RFX6 expression and densities in the different cell types in adult pancreata (α-cells, n = 3,267 cells; β-cells, n = 2,647 cells, δ-cells, n = 153 cells; pancreatic polypeptide [PP] cells, n = 23 cells). B: RFX6 single-cell expression in both nondiabetic (ND) and T2D samples. C: Schematics of the lentiviral constructs coding for a shRNA against RFX6 and a nontargeted control (Ctrl) with a GFP reporter. hCMV, human cytomegalovirus; IRES, internal ribosome entry site; WPRE, woodchuck hepatitis virus posttranscriptional regulatory element. D: Schematic of whole pseudoislet technique (Research Design and Methods). E: Static batch glucagon secretion of whole control and RFX6kd pseudoislets (duplicates from n = 4 donors, normalized to total pseudoislet glucagon content). F: Schematic of MagSep pseudoislet technique (Research Design and Methods). G: Assessment of hormone expression in the MagSep CD26+ α-cell fraction by qRT-PCR (n = 7). H: Assessment of RFX6 expression in both the MagSep CD26+ (n = 8) and CD26neg (n = 4) fractions, normalized to the scramble control of the appropriate fraction. I: Assessment of putative RFX6 α-cell targets in CD26+ cells by qRT-PCR (n = 4). Data are presented as mean values ± SEM. Two-tailed t tests were used to generate P values: ns, not significant; *P < 0.05, **P < 0.01, ***P < 0.001.

Figure 1

RFX6 shRNA-mediated knockdown can be selectively targeted to CD26+ α-cells. A: Violin plot of single-cell RFX6 expression and densities in the different cell types in adult pancreata (α-cells, n = 3,267 cells; β-cells, n = 2,647 cells, δ-cells, n = 153 cells; pancreatic polypeptide [PP] cells, n = 23 cells). B: RFX6 single-cell expression in both nondiabetic (ND) and T2D samples. C: Schematics of the lentiviral constructs coding for a shRNA against RFX6 and a nontargeted control (Ctrl) with a GFP reporter. hCMV, human cytomegalovirus; IRES, internal ribosome entry site; WPRE, woodchuck hepatitis virus posttranscriptional regulatory element. D: Schematic of whole pseudoislet technique (Research Design and Methods). E: Static batch glucagon secretion of whole control and RFX6kd pseudoislets (duplicates from n = 4 donors, normalized to total pseudoislet glucagon content). F: Schematic of MagSep pseudoislet technique (Research Design and Methods). G: Assessment of hormone expression in the MagSep CD26+ α-cell fraction by qRT-PCR (n = 7). H: Assessment of RFX6 expression in both the MagSep CD26+ (n = 8) and CD26neg (n = 4) fractions, normalized to the scramble control of the appropriate fraction. I: Assessment of putative RFX6 α-cell targets in CD26+ cells by qRT-PCR (n = 4). Data are presented as mean values ± SEM. Two-tailed t tests were used to generate P values: ns, not significant; *P < 0.05, **P < 0.01, ***P < 0.001.

Close modal

Multiple factors may contribute to the observed lack of glucagon secretion phenotype, including the possibility that suppression of RFX6 expression in multiple islet cell types could mask α-cell phenotypes through nonautonomous mechanisms. To test this possibility, we developed a method for α-cell–selective gene targeting. An enriched primary human α-cell population was obtained by labeling dispersed islet cells with an antibody recognizing CD26, a known α-cell–specific surface protein (39,40). PE-conjugated α-CD26+ cells were isolated from CD26neg non–α-islet cells using magnetic beads (MagSep), then infected with lentivirus expressing shRNA targeting RFX6 and a GFP transgene or with control lentivirus (Research Design and Methods) (Fig. 1C). We cultured infected CD26+ cells separately from CD26neg cells for 4 days to allow for shRNA suppression of RFX6 mRNA, then reaggregated CD26+ and CD26neg cells for functional studies (hereafter, MagSep RFX6kd pseudoislets; Research Design and Methods) (Fig. 1F).

Five days after lentiviral infection, we used qRT-PCR and immunostaining to verify that the CD26+ fraction was enriched for α-cells (expressing glucagon [GCG]) and depleted of β-cells (expressing insulin [INS]) or δ-cells (expressing somatostatin [SST]) (Fig. 1G and Supplementary Fig. 2). In CD26+ cell fractions assessed by qRT-PCR, there was a marked reduction in RFX6 mRNA; by contrast, the CD26neg fraction had no change in RFX6 mRNA (Fig. 1H). Thus, we suppressed RFX6 expression selectively in CD26+ cells. Next, we assessed imputed targets of RFX6 in these cells by qRT-PCR. Compared with control CD26+ cells, we observed a marked reduction in mRNA levels of CACNA1A, SLC5A1, RFX3, and CACNB2, but not of GCG, CACNA1C, ABCC8, or GCK (Fig. 1I). Static batch assays with MagSep RFX6kd pseudoislets revealed a modest but significant reduction of glucagon secretion at a low glucose concentration (1 mmol/L glucose) compared with MagSep pseudoislets infected with control lentivirus (Fig. 2A). By contrast, batch assays did not reveal differences in insulin secretion by MagSep RFX6kd pseudoislets (Fig. 2B). Thus, development of selective gene targeting in human islet α-cells revealed a requirement for RFX6 in maintaining α-cell gene regulation and function.

Figure 2

RFX6kd MagSep pseudoislets exhibit dysregulated glucagon secretion and blunted exocytosis in vitro. A: In vitro low glucose-stimulated glucagon secretion from human MagSep RFX6kd pseudoislets (replicates from n = 6 donors). Secreted glucagon normalized to glucagon content. B: In vitro high glucose and IBMX-stimulated insulin secretion from human MagSep RFX6kd pseudoislets (replicates from n = 6 donors). Secreted insulin normalized to insulin content. C: Schematic of patch clamp technique with MagSep RFX6kd pseudoislets. D: Exocytosis representative trace, control, and RFX6kd. E: Cumulative capacitance per depolarization, control and RFX6kd (n = 4 donors). fF, femtofarad; pF, picofarad. Exocytosis at 1st depolarization (F) and average of 2nd–10th depolarizations (G), control and RFX6kd (n = 4 donors). H: Calcium channel activity, using Ba2+ as a charge carrier, in control and RFX6kd (n = 3 donors). Data are presented as mean values ± SEM. Two-tailed t tests were used to generate P values: ns, not significant; *P < 0.05, **P < 0.01.

Figure 2

RFX6kd MagSep pseudoislets exhibit dysregulated glucagon secretion and blunted exocytosis in vitro. A: In vitro low glucose-stimulated glucagon secretion from human MagSep RFX6kd pseudoislets (replicates from n = 6 donors). Secreted glucagon normalized to glucagon content. B: In vitro high glucose and IBMX-stimulated insulin secretion from human MagSep RFX6kd pseudoislets (replicates from n = 6 donors). Secreted insulin normalized to insulin content. C: Schematic of patch clamp technique with MagSep RFX6kd pseudoislets. D: Exocytosis representative trace, control, and RFX6kd. E: Cumulative capacitance per depolarization, control and RFX6kd (n = 4 donors). fF, femtofarad; pF, picofarad. Exocytosis at 1st depolarization (F) and average of 2nd–10th depolarizations (G), control and RFX6kd (n = 4 donors). H: Calcium channel activity, using Ba2+ as a charge carrier, in control and RFX6kd (n = 3 donors). Data are presented as mean values ± SEM. Two-tailed t tests were used to generate P values: ns, not significant; *P < 0.05, **P < 0.01.

Close modal

To assess α-cell functional defects after RFX6 suppression, we performed patch clamp electrophysiology on GFP+ cells isolated from control and MagSep RFX6kd pseudoislets (Fig. 2C). GFP+ cells were confirmed to be GCG+ by immunostaining (27). In line with our GSGS data, MagSep RFX6kd α-cells had significantly reduced exocytosis to a series of membrane depolarizations compared with control α-cells (Fig. 2D and E). Average exocytotic differences between RFX6kd and control cells were larger after the first depolarization event, compared with subsequent depolarization events, suggesting an effect on docked or “readily releasable” glucagon granules (Fig. 2F and G). In prior studies in β-cells, loss of RFX6 led to perturbations of calcium channel activity (3). However, we found no significant differences in calcium channel activity between RFX6kd and control α-cells (Fig. 2H). These findings complement prior studies of RFX6 in β-cells and suggest that RFX6-dependent regulation of exocytosis may differ in α- and β-cells (3,5).

RFX6 Suppression Durably Impairs α-Cell Function of Transplanted Pseudoislets

Glucagon secretion was modestly reduced after RFX6kd in α-cells for 5 days in vitro, and we next investigated the durability of this glucagon secretion defect in vivo using a transplantation model (Figs. 2A and 3,A). Because mature glucagon is identical in mice and humans, we previously developed immunocompromised NOD scid IL2Rγnull (NSG) mice harboring an in-frame deletion of Glucagon1-29 (GKO) to permit ELISA-based measures of glucagon output from transplanted human islets (hereafter, GKO-NSG) (21). Prior studies have shown that in GKO-NSG mice transplanted with human islets or pseudoislets, hypoglycemia elicits glucagon secretion (21). We transplanted 1) whole RFX6kd and 2) MagSep RFX6kd pseudoislets and their respective controls in GKO-NSG mice and assessed glucagon secretion at least 1 month after transplantation in response to hypoglycemia through IP-ITT (Fig. 3). All transplanted GKO-NSG mice experienced acute hypoglycemia upon insulin injection (Fig. 3B and E and Supplementary Figs. 3A and B and 4A and B). Mice transplanted with whole RFX6kd or control pseudoislets had comparable glucagon excursions after IP-ITT (Fig. 3C and D and Supplementary Fig. 3C and D). By contrast, average glucagon secretion was significantly blunted in mice transplanted with MagSep RFX6kd pseudoislets (Fig. 3F and G and Supplementary Fig. 4C and D), consistent with our in vitro GSGS findings (Fig. 2A). In summary, these data demonstrate that targeted RFX6kd in human adult α-cells durably impaired glucagon secretion after transplantation in vivo.

Figure 3

RFX6kd MagSep pseudoislets exhibit blunted glucagon secretion in vivo. A: Schematic of MagSep or whole RFX6kd pseudoislet transplant and IP-ITT. BG: Blood glucose, plasma glucagon, and area under the curve (AUC) of glucagon secretion upon insulin injection of control and RFX6kd whole (BD) (ctrl and KD, n = 5 donors) and MagSep (EG) (ctrl, n = 7 mice; KD, n = 8 mice; n = 7 donors) donor-matched pseudoislet transplanted mice. The data are presented as mean values ± SEM. Two-tailed paired t tests were used to generate P values: ns, not significant; *P < 0.05.

Figure 3

RFX6kd MagSep pseudoislets exhibit blunted glucagon secretion in vivo. A: Schematic of MagSep or whole RFX6kd pseudoislet transplant and IP-ITT. BG: Blood glucose, plasma glucagon, and area under the curve (AUC) of glucagon secretion upon insulin injection of control and RFX6kd whole (BD) (ctrl and KD, n = 5 donors) and MagSep (EG) (ctrl, n = 7 mice; KD, n = 8 mice; n = 7 donors) donor-matched pseudoislet transplanted mice. The data are presented as mean values ± SEM. Two-tailed paired t tests were used to generate P values: ns, not significant; *P < 0.05.

Close modal

RNA-seq Reveals the RFX6-Dependent Transcriptome in Adult Human α-Cells

To identify genetic mechanisms underlying phenotypes after RFX6kd, we performed transcriptome analysis on FACS-purified CD26+ GFP+ α-cells (hereafter, αRFX6kd cells: n = 4 independent donors) (Fig. 4A). qRT-PCR analysis confirmed depletion of RFX6 mRNA and enrichment of GCG mRNA in this α-cell–enriched subset. As expected, INS mRNA levels were also depleted (Supplementary Fig. 5A). We generated and sequenced bulk RNA-seq libraries of RFX6kd and control α-cells and confirmed significant RFX6 suppression in the αRFX6kd-cell subset (Fig. 4B). Principal component analysis revealed interdonor variability and separation of αRFX6kd and control cells, consistent with previous studies (Supplementary Fig. 5B) (2). We used the DESeq2 algorithm to identify 915 differentially expressed genes (DEGs) after αRFX6kd, with 558 showing reduced expression, and 357 showing increased expression (Fig. 4C and D and Supplementary Tables 3 and 4).

Figure 4

RNA-seq of αRFX6kd reveals genes regulated by RFX6 in primary human α-cells. A: Schematic of islet cell processing to seed the RNA-seq pipeline. B: Normalized transcript levels of RFX6 in CD26+ control and αRFX6kd cell fractions (n = 4). C: Volcano plot of DEGs in the αRFX6kd cell fraction. D: Heat map of DEGs in αRFX6kd. Significantly downregulated (E) and upregulated (F) gene ontology (GO) pathways after RFX6kd in α-cells. MAPK, mitogen-activated protein kinase. Data are presented as mean values ± SEM. *P < 0.05.

Figure 4

RNA-seq of αRFX6kd reveals genes regulated by RFX6 in primary human α-cells. A: Schematic of islet cell processing to seed the RNA-seq pipeline. B: Normalized transcript levels of RFX6 in CD26+ control and αRFX6kd cell fractions (n = 4). C: Volcano plot of DEGs in the αRFX6kd cell fraction. D: Heat map of DEGs in αRFX6kd. Significantly downregulated (E) and upregulated (F) gene ontology (GO) pathways after RFX6kd in α-cells. MAPK, mitogen-activated protein kinase. Data are presented as mean values ± SEM. *P < 0.05.

Close modal

Gene ontology pathway analysis of downregulated genes suggested that αRFX6kd impacted known regulators of endocrine function and identity. Prior studies have identified a subset of genes with human α-cell–enriched or α-cell–specific expression, including GPR119, GPR148, LOXL4, KLHL41, PLCE1, LDB2, ALDH1A1, PEMT, PTPRT, SPOCK3, and ARRDC4 (15,27,4143), and αRFX6kd resulted in reduced expression of these genes in α-cells (Fig. 4D). We also observed reduced expression of genes known to regulate islet development and fate (ISL1, IRF6, MAFB, LDHA, and JAK2), peptide hormone secretion (RFX6, GPR119, UCN3, SLC30A8, ISL1, RFX3, and GLUL), cell-cell signaling (UCN3, GPR119, TMEM64, FGF18, KCNA5, CACNA1A, NCAM1, SSTR2, EPCAM, and RAPGEF4), and vesicle-mediated transport (LGI3, SYTL2, BAIAP3, and EPG5) (Fig. 4E). Conversely, we observed increased expression of genes governing stress responses (HIF1A, DDR1, IFIH1, and IFIT3) and proliferation (ABCG1 and TRIB3) in RFX6kd α-cells (Fig. 4F). We conclude that RFX6 is required in mature human α-cells to maintain regulation of genes governing α-cell identity and function.

Comparison of RFX6-Dependent Transcriptomes in Adult α-Cells and β-Cells

We next identified DEGs in FACS-purified β-cells after RFX6kdRFX6kd). To isolate β-cells, we stained cells from transduced whole pseudoislets with an antibody specific for the human β-cell surface marker NTPDase3, then FACS-separated NTPDase3+ GFP+ cells (n = 4 independent donors) (Fig. 5A) (44). Principal component analysis revealed appropriate separation of RFX6kd and control NTPDase3+ cells and INS mRNA enrichment (Supplementary Fig. 5C and D). Compared with controls, the βRFX6kd-cell fraction also had significant RFX6 suppression (Fig. 5B). Knockdown of RFX6 in β-cells (63%) was slightly reduced compared with knockdown in donor-matched α-cells (77%) (Fig. 5C). DESeq2 analysis revealed 155 DEGs in βRFX6kd, with 96 downregulated and 59 upregulated genes (Fig. 5D and Supplementary Tables 5 and 6).

Figure 5

RNA-seq of βRFX6kd reveals both distinct and overlapping genes regulated by RFX6 in primary human β-cells compared with α-cells. A: Schematic of islet cell processing to seed the RNA-seq pipeline for human β-cells. B: Normalized transcript levels of RFX6 in NTPDase3+ control and β-RFX6kd cell fractions (n = 4). C: Normalized transcript levels of control and RFX6kd fractions of the CD26+ (green) and NTPDase3+ (yellow) fractions. D: Volcano plot of DEGs in αRFX6kd. E: Venn diagram showing the overlap between αRFX6kd and βRFX6kd DEGs. Significant gene ontology pathways of overlapping αRFX6kd and βRFX6kd DEGs (F) and β-cell specific DEGs (G). The data are presented as mean values ± SEM. *P < 0.05.

Figure 5

RNA-seq of βRFX6kd reveals both distinct and overlapping genes regulated by RFX6 in primary human β-cells compared with α-cells. A: Schematic of islet cell processing to seed the RNA-seq pipeline for human β-cells. B: Normalized transcript levels of RFX6 in NTPDase3+ control and β-RFX6kd cell fractions (n = 4). C: Normalized transcript levels of control and RFX6kd fractions of the CD26+ (green) and NTPDase3+ (yellow) fractions. D: Volcano plot of DEGs in αRFX6kd. E: Venn diagram showing the overlap between αRFX6kd and βRFX6kd DEGs. Significant gene ontology pathways of overlapping αRFX6kd and βRFX6kd DEGs (F) and β-cell specific DEGs (G). The data are presented as mean values ± SEM. *P < 0.05.

Close modal

We found that 45% (71 of 155) of βRFX6kd DEGs overlapped with αRFX6kd DEGs (Fig. 5E). This overlap included genes consistent with previous studies, including those encoding factors with well-characterized roles in peptide hormone secretion and ion channel function (CACNA1A, CACNA1C, SLC30A8, SLC5A1, KCNA5, and BAIAP3) and regulation of cell communication (NCAM1, TMEM64, STAT1, and IFI6) (Fig. 5F) (3). In addition, we observed 55% of DEGs (84 of 155) were altered after βRFX6kd but not αRFX6kd (Fig. 5G). This included ABCC8 and GCK, crucial regulators of β-cell stimulus-secretion coupling (4547). Our finding corroborates prior work in mice showing that Abcc8 and Gck are regulated by Rfx6 in β-cells (5). Other DEGs identified in βRFX6kd cells but not αRFX6kd cells include genes encoding β-cell–enriched factors (C1orf127, IGSF1, and CAPN13), TFs (SOX9, STAT6, and FOS), regulators of hormone secretion (LRP5, CASR, CFTR, SPP1, and ROBO1), and cell adhesion (CXCR4, NEO1, CDON, ROBO1, PCDHA4, SPON1, and VCAM1). Thus, RFX6 regulates distinct molecular functions and biological processes in α- and β-cells, while also coordinately regulating an overlapping subset of genes with orthologous roles.

Direct Targets of RFX6 Identified by CUT&RUN

To identify islet cell genomic DNA bound by RFX6, we used CUT&RUN, a high-resolution chromatin profiling strategy (20). Since available antibodies for endogenous islet RFX6 did not meet standards for chromatin assays, we misexpressed RFX6 in human islet cells with a lentiviral construct encoding human RFX6 tagged with a FLAG immunoepitope (n = 3 independent donors) (Fig. 6A and Supplementary Fig. 6AC). DNA bound by RFX6-FLAG protein was enriched with an anti-FLAG antibody and sequenced (Research Design and Methods). HOMER analysis was used, as previously described (2,35), to identify motifs bound by RFX6-FLAG. Peak centers of RFX6-FLAG sample libraries had enriched read densities compared with IgG controls, where there was minimal enrichment at the peak centers (Fig. 6B). The most enriched RFX6-FLAG–bound DNA-binding motif identified by HOMER was RFX6, demonstrating high specificity of the CUT&RUN assay (Fig. 6C). Other enriched motifs include sequences binding other RFX family members, including RFX5, and other TFs critical for islet cell identity and function like ISL1, NKX6.1, and MAFB (Fig. 6C).

Figure 6

Identification of putative RFX6 genetic targets in primary human islet cells using CUT&RUN. A: Schematic for CUT&RUN: pseudoislets were transduced with lentiviral constructs to overexpress RFX6-FLAG under the human cytomegalovirus (hCMV) promoter and used for CUT&RUN experiments with anti-FLAG antibody (Ab) (n = 3 donors). WPRE, woodchuck hepatitis virus posttranscriptional regulatory element. B: Heat maps showing peak enrichment of CUT&RUN sample libraries generated with IgG control and FLAG antibodies. C: HOMER analysis identified enriched binding motifs. D: Venn diagram showing the overlap between RFX6-associated genomic regions and RFX6kd DEGs. Tracks showing RFX6-FLAG genomic regions associated with GPR148 (E) and LOXL4 (F): differential peaks from RFX6-FLAG samples were analyzed using the GREAT algorithm and mapped at the top of each track. Human islet accessible chromatin regions are shown by assay for transposase-accessible chromatin (ATAC)-seq tracks.

Figure 6

Identification of putative RFX6 genetic targets in primary human islet cells using CUT&RUN. A: Schematic for CUT&RUN: pseudoislets were transduced with lentiviral constructs to overexpress RFX6-FLAG under the human cytomegalovirus (hCMV) promoter and used for CUT&RUN experiments with anti-FLAG antibody (Ab) (n = 3 donors). WPRE, woodchuck hepatitis virus posttranscriptional regulatory element. B: Heat maps showing peak enrichment of CUT&RUN sample libraries generated with IgG control and FLAG antibodies. C: HOMER analysis identified enriched binding motifs. D: Venn diagram showing the overlap between RFX6-associated genomic regions and RFX6kd DEGs. Tracks showing RFX6-FLAG genomic regions associated with GPR148 (E) and LOXL4 (F): differential peaks from RFX6-FLAG samples were analyzed using the GREAT algorithm and mapped at the top of each track. Human islet accessible chromatin regions are shown by assay for transposase-accessible chromatin (ATAC)-seq tracks.

Close modal

RFX6-FLAG–bound genomic regions were analyzed using GREAT, which identified 10,610 associated genes (36). To prioritize candidate genes likely directly regulated by RFX6, we intersected the gene list generated by GREAT with the DEGs identified after transcriptome analysis of αRFX6kd and βRFX6kd. This data integration identified 715 total “overlapping genes” (Fig. 6D and Supplementary Table 7). This concordance between RFX6-FLAG–bound regions and DEGs after RFX6kd provides evidence that our approach identified direct RFX6 targets.

Direct targets of RFX6 imputed by our analysis included ion channel subunits (CACNA1A, CACNA1C, KCNA5, and KCTD12), transmembrane transporters and receptors (SLC30A8, SLC5A1, GPR119, and ABCC8), enzymes (PCSK1, PCSK2, and GCK), and TFs (RFX6 and PAX6) previously shown to govern islet function (Fig. 6D). A subset of genes expressed in human α-cells (but not expressed in islet α-cells from other species such as mice) were identified as putative direct targets of RFX6, including GPR148, LOXL4, PLCE1, SPOCK3, and ARRDC4 (Fig. 6D and Supplementary Table 7) (15,27,4143). Likewise, our analysis revealed direct interaction of RFX6 with a subset of genes expressed in human β-cells (but not in β-cells from mice) (43) such as ROBO1 and CASR (Fig. 6D and Supplementary Table 7). Genomic regions of presumptive RFX6 target genes in α-cells, such as GPR148 and LOXL4, bound by RFX6-FLAG also corresponded with regions of transcriptionally active chromatin, as identified by previous assay for transposase-accessible chromatin-seq studies (Fig. 6E and F) (4850). These findings support the view that RFX6 directly activates genes encoding factors essential for mature α-cell function in human islets.

Work here fills significant knowledge gaps regarding RFX6 roles in mature adult pancreatic islet α-cells. Using a novel cell-selective lentiviral shRNA targeting approach to investigate functions of RFX6 in primary human adult islet cells, we have revealed a requirement for RFX6 in maintaining adult human α-cell gene regulation and glucagon secretion. By combining transcriptome analysis with CUT&RUN studies, we identified direct targets of RFX6, including genes crucial in endocrine cell fate and hormone secretion in addition to previously unrecognized RFX6-dependent targets. These findings elucidate a novel requirement for RFX6 in regulating mature human α-cell function.

Dysregulated RFX6 expression has been noted in islet cells from subjects with diabetes, and RFX6 loss was found to impair β-cell function in animal models and immortalized human β-cell lines (3,57,14). Far less is known about RFX6 functions in human α-cells compared with β-cells (13,27). Our in vitro studies, in vivo transplantation analysis, and patch clamp studies using MagSep RFX6kd pseudoislets revealed a marked reduction of glucose-dependent glucagon secretion and impaired α-cell exocytosis. Physiological studies of glucagon secretion are challenged by unavoidable variation between individual human donors (Supplementary Fig. 3B and D); thus, our conclusions are based on multiple samplings and the average of our studies (e.g., Fig. 3F). Transcriptomic and chromatin analyses identified downstream targets of RFX6 known to regulate exocytosis and glucagon output, including known ion channels, transport proteins, and vesicular fusion proteins. Reduced human α-cell glucagon output has not been previously linked to targeted RFX6 loss, although a recent study of type 1 diabetes islets correlated reduced α-cell expression of RFX6 (and multiple other factors) with impaired glucagon secretion (13). Unexpectedly, we did not detect impairment of α-cell glucagon secretion without selective RFX6 suppression, suggesting that compensatory effects from RFX6 loss in β-cells (Fig. 5E) or other non–α-islet cells may offset autonomous effects of RFX6 loss in α-cells, although further studies are needed to explore this possibility.

To identify direct genetic targets of RFX6 in human islet cells, we used CUT&RUN after misexpression of FLAG-tagged RFX6. While caution should be exercised in interpreting results with a tagged RFX6 due to possible excessive binding and false positive associations with target genes, we limited calling of RFX6 direct target genes by integrating the CUT&RUN analysis with α- and β-cell DEG analysis after RFX6 suppression. This approach enhanced the reliability of identifying direct genetic targets of RFX6 imputed by CUT&RUN. Among these targets were genes previously associated with RFX6-dependent regulation in β-cells, including ABCC8 and GCK (3,5). We also observed distinct α- and β-cell DEGs. For example, while expression of α-cell ABCC8 or GCK after RFX6kd remained unchanged, expression of genes regulating vesicle-mediated transport, nutrient processing, exocytosis, and ion channel activity were changed in α-cells, but not in β-cells, after RFX6kd. These data indicate differential regulation of α- and β-cells by RFX6. For example, our findings with patch clamp suggest that RFX6 loss impaired exocytotic machinery but not calcium channel activity (Fig. 2H) (3,5).

In summary, our study reveals that RFX6 is required for maintenance of genetic regulation and function in mature adult human α-cells, particularly in preserving genetic pathways responsible for regulated hormone secretion. Many islet transcriptional regulators, such as RFX6, PDX1, and MAFB, are expressed in multiple islet cell types, highlighting the potential of cell type-specific genetic targeting in deciphering cell autonomous and nonautonomous functions of these TFs. These findings advance our understanding of RFX6-dependent mechanisms that maintain adult human α- and β-cell functions. Moreover, they emphasize the importance of further exploring human α-cell biology to enhance our understanding of diabetes and pancreatic cancer. Our innovative approaches for targeted genetic manipulation in human primary islet cells can be extended to investigate cell-specific functions of other factors relevant to these diseases.

This article contains supplementary material online at https://doi.org/10.2337/figshare.24615675.

R.J.B. is currently affiliated with Diabetes, Obesity, and Metabolism Institute at Icahn School of Medicine, Mount Sinai, New York, NY.

Acknowledgments. The authors thank Drs. S. Park, R.L. Whitener, M.A. Miranda, S.H. Kim, M. Neukam, H. Peiris, and Mr. J. Lam in the Kim group, as well as Professors A. Gloyn, R. Nusse, and W. Talbot (Stanford) for their technical and conceptual guidance, advice, and encouragement. The authors thank organ donors and their families, and the Alberta Diabetes Institute IsletCore, Integrated Islet Distribution Program, the National Disease Research Interchange, and the International Institute for the Advancement of Medicine, the University of San Francisco Islet Core, and the Stanford Diabetes Center Islet Core. In Alberta, we thank the Human Organ Procurement and Exchange (HOPE) program and Trillium Gift of Life Network (TGLN), and J Lyon for his efforts in human islet isolation.

Funding. The IsletCore was supported by the Alberta Diabetes Institute, and the Integrated Islet Distribution Program was supported by National Institutes of Health (NIH), National Institute of Diabetes and Digestive and Kidney Diseases (UC4 DK098085). V.M.N.C. was supported by a NIH National Institute of General Medical Sciences T32 training grant (5T32GM007790). M.F.Q. was previously supported by the Stanford Medical Scholars, Howard Hughes Medical Institute (HHMI) Medical Research Fellows Programs, Stanford Medical Scientist Training Program, and a Knight-Hennessy Scholars award. K.T. was supported by a graduate research fellowship award from the National Science Foundation (DGF-114747). R.J.B. was supported by a postdoctoral fellowship from JDRF (3-PDF-2018-584-A-N). RNA-seq data were generated using instrumentation purchased with NIH funds (S10OD025212 and 1S10OD021763). This manuscript also used data acquired from the Human Pancreas Analysis Program (HPAP-Research Resource Identifier [RRID]: SCR_016202) Database (https://hpap.pmacs.upenn.edu), a Human Islet Research Network (RRID: SCR_014393) consortium supported by National Institute of Diabetes and Digestive and Kidney Diseases grants (UC4 DK112217, U01 DK123594, UC4 DK112232, and U01 DK123716). Work here was supported by NIH National Institute of Diabetes and Digestive and Kidney Diseases awards (R01 DK107507, R01 DK108817, and U01 DK123743 to S.K.K., and R01DK126482 to S.K.K. and P.E.M.), a JDRF Center of Excellence Award (to S.K.K.), and NIH National Institute of Diabetes and Digestive and Kidney Diseases P30 DK116074 (S.K.K.), which supports the Stanford Islet Research Core and Diabetes Genomics and Analysis Core. P.E.M. holds the Canada Research Chair (Tier I) in Islet Biology. S.K.K. holds the KM Mulberry Chair at Stanford.

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

Author Contributions. V.M.N.C., M.F.Q., K.T., A.B., R.J.B., X.G., Y.H., M.N., W.Z., and C.C. performed experiments. V.M.N.C., M.F.Q., K.T., R.J.B., P.E.M., and S.K.K. acquired the funding. V.M.N.C., K.T., and S.K.K. conceptualized the study. V.M.N.C. and S.K.K. wrote the manuscript with input from all coauthors. S.K.K. supervised the study. V.M.N.C. and S.K.K. are the guarantors of this work and, as such, had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.

Prior Presentation. Parts of this study were presented in abstract form and as an oral presentation at the 2021 Western Region Islet Study Group, Stevenson, WA, 3–5 November 2021.

1.
Aftab
S
,
Semenec
L
,
Chu
JSC
,
Chen
N.
Identification and characterization of novel human tissue-specific RFX transcription factors
.
BMC Evol Biol
2008
;
8
:
226
2.
Bevacqua
RJ
,
Lam
JY
,
Peiris
H
, et al
SIX2 and SIX3 coordinately regulate functional maturity and fate of human pancreatic β cells
.
Genes Dev
2021
;
35
:
234
249
3.
Chandra
V
,
Albagli-Curiel
O
,
Hastoy
B
, et al
RFX6 regulates insulin secretion by modulating Ca2+ homeostasis in human β cells
.
Cell Rep
2014
;
9
:
2206
2218
4.
Peiris
H
,
Park
S
,
Louis
S
, et al
Discovering human diabetes-risk gene function with genetics and physiological assays
.
Nat Commun
2018
;
9
:
3855
5.
Piccand
J
,
Strasser
P
,
Hodson
DJ
, et al
Rfx6 maintains the functional identity of adult pancreatic β cells
.
Cell Rep
2014
;
9
:
2219
2232
6.
Smith
SB
,
Qu
HQ
,
Taleb
N
, et al
Rfx6 directs islet formation and insulin production in mice and humans
.
Nature
2010
;
463
:
775
780
7.
Soyer
J
,
Flasse
L
,
Raffelsberger
W
, et al
Rfx6 is an Ngn3-dependent winged helix transcription factor required for pancreatic islet cell development
.
Development
2010
;
137
:
203
212
8.
Taleb
N
,
Polychronakos
C.
RFX6 is needed for the development and maintenance of the β-cell phenotype
.
Islets
2011
;
3
:
291
293
9.
Zhang
H
,
Colclough
K
,
Gloyn
AL
,
Pollin
TI.
Monogenic diabetes: a gateway to precision medicine in diabetes
.
J Clin Invest
2021
;
131
:
e142244
10.
Kambal
MA
,
Al-Harbi
DA
,
Al-Sunaid
AR
,
Al-Atawi
MS.
Mitchell-Riley syndrome due to a novel mutation in RFX6
.
Front Pediatr
2019
;
7
:
243
11.
Mitchell
J
,
Punthakee
Z
,
Lo
B
, et al
Neonatal diabetes, with hypoplastic pancreas, intestinal atresia and gall bladder hypoplasia: search for the aetiology of a new autosomal recessive syndrome
.
Diabetologia
2004
;
47
:
2160
2167
12.
Patel
KA
,
Kettunen
J
,
Laakso
M
, et al
Heterozygous RFX6 protein truncating variants are associated with MODY with reduced penetrance
.
Nat Commun
2017
;
8
:
888
13.
Brissova
M
,
Haliyur
R
,
Saunders
D
, et al
α cell function and gene expression are compromised in type 1 diabetes
.
Cell Rep
2018
;
22
:
2667
2676
14.
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
15.
Muraro
MJ
,
Dharmadhikari
G
,
Grün
D
, et al
A single-cell transcriptome atlas of the human pancreas
.
Cell Syst
2016
;
3
:
385
394.e3
16.
Benner
C
,
van der Meulen
T
,
Cacéres
E
,
Tigyi
K
,
Donaldson
CJ
,
Huising
MO.
The transcriptional landscape of mouse beta cells compared to human beta cells reveals notable species differences in long non-coding RNA and protein-coding gene expression
.
BMC Genomics
2014
;
15
:
620
17.
Cabrera
O
,
Berman
DM
,
Kenyon
NS
,
Ricordi
C
,
Berggren
PO
,
Caicedo
A.
The unique cytoarchitecture of human pancreatic islets has implications for islet cell function
.
Proc Natl Acad Sci U S A
2006
;
103
:
2334
2339
18.
Dai
C
,
Brissova
M
,
Hang
Y
, et al
Islet-enriched gene expression and glucose-induced insulin secretion in human and mouse islets
.
Diabetologia
2012
;
55
:
707
718
19.
Camunas-Soler
J
,
Dai
XQ
,
Hang
Y
, et al
Patch-seq links single-cell transcriptomes to human islet dysfunction in diabetes
.
Cell Metab
2020
;
31
:
1017
1031.e4
20.
Skene
PJ
,
Henikoff
S.
An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites
.
eLife
2017
;
6
:
e21856
21.
Tellez
K
,
Hang
Y
,
Gu
X
,
Chang
CA
,
Stein
RW
,
Kim
SK.
In vivo studies of glucagon secretion by human islets transplanted in mice
.
Nat Metab
2020
;
2
:
547
557
22.
Shapira
SN
,
Naji
A
,
Atkinson
MA
,
Powers
AC
,
Kaestner
KH.
Understanding islet dysfunction in type 2 diabetes through multidimensional pancreatic phenotyping: the Human Pancreas Analysis Program
.
Cell Metab
2022
;
34
:
1906
1913
23.
Dobin
A
,
Davis
CA
,
Schlesinger
F
, et al
STAR: ultrafast universal RNA-seq aligner
.
Bioinformatics
2013
;
29
:
15
21
24.
Anders
S
,
Pyl
PT
,
Huber
W.
HTSeq--a Python framework to work with high-throughput sequencing data
.
Bioinformatics
2015
;
31
:
166
169
25.
Satija
R
,
Farrell
JA
,
Gennert
D
,
Schier
AF
,
Regev
A.
Spatial reconstruction of single-cell gene expression data
.
Nat Biotechnol
2015
;
33
:
495
502
26.
Stuart
T
,
Butler
A
,
Hoffman
P
, et al
Comprehensive integration of single-cell data
.
Cell
2019
;
177
:
1888
1902.e21
27.
Dai
XQ
,
Camunas-Soler
J
,
Briant
LJB
, et al
Heterogenous impairment of α cell function in type 2 diabetes is linked to cell maturation state
.
Cell Metab
2022
;
34
:
256
268.e5
28.
Li
B
,
Dewey
CN.
RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome
.
BMC Bioinformatics
2011
;
12
:
323
29.
Love
MI
,
Huber
W
,
Anders
S.
Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2
.
Genome Biol
2014
;
15
:
550
30.
Huang
W
,
Sherman
BT
,
Lempicki
RA.
Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources
.
Nat Protoc
2009
;
4
:
44
57
31.
Raudvere
U
,
Kolberg
L
,
Kuzmin
I
, et al
g:Profiler: a web server for functional enrichment analysis and conversions of gene lists (2019 update)
.
Nucleic Acids Res
2019
;
47
:
W191
W198
32.
Zhu
Q
,
Liu
N
,
Orkin
SH
,
Yuan
GC.
CUT&RUNTools: a flexible pipeline for CUT&RUN processing and footprint analysis
.
Genome Biol
2019
;
20
:
192
33.
Bolger
AM
,
Lohse
M
,
Usadel
B.
Trimmomatic: a flexible trimmer for Illumina sequence data
.
Bioinformatics
2014
;
30
:
2114
2120
34.
Langmead
B
,
Salzberg
SL.
Fast gapped-read alignment with Bowtie 2
.
Nat Methods
2012
;
9
:
357
359
35.
Heinz
S
,
Benner
C
,
Spann
N
, et al
Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities
.
Mol Cell
2010
;
38
:
576
589
36.
McLean
CY
,
Bristor
D
,
Hiller
M
, et al
GREAT improves functional interpretation of cis-regulatory regions
.
Nat Biotechnol
2010
;
28
:
495
501
37.
Kent
WJ
,
Sugnet
CW
,
Furey
TS
, et al
The human genome browser at UCSC
.
Genome Res
2002
;
12
:
996
1006
38.
Edgar
R
,
Domrachev
M
,
Lash
AE.
Gene Expression Omnibus: NCBI gene expression and hybridization array data repository
.
Nucleic Acids Res
2002
;
30
:
207
210
39.
Arda
HE
,
Li
L
,
Tsai
J
, et al
Age-dependent pancreatic gene regulation reveals mechanisms governing human β cell function
.
Cell Metab
2016
;
23
:
909
920
40.
Augstein
P
,
Naselli
G
,
Loudovaris
T
, et al
Localization of dipeptidyl peptidase-4 (CD26) to human pancreatic ducts and islet alpha cells
.
Diabetes Res Clin Pract
2015
;
110
:
291
300
41.
Amisten
S
,
Atanes
P
,
Hawkes
R
, et al
A comparative analysis of human and mouse islet G-protein coupled receptor expression
.
Sci Rep
2017
;
7
:
46600
42.
Blodgett
DM
,
Nowosielska
A
,
Afik
S
, et al
Novel observations from next-generation RNA sequencing of highly purified human adult and fetal islet cell subsets
.
Diabetes
2015
;
64
:
3172
3181
43.
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
44.
Saunders
DC
,
Brissova
M
,
Phillips
N
, et al
Ectonucleoside triphosphate diphosphohydrolase-3 antibody targets adult human pancreatic β cells for in vitro and in vivo analysis
.
Cell Metab
2019
;
29
:
745
754.e4
45.
Nakamura
Y
,
Bryan
J.
Targeting SUR1/Abcc8-type neuroendocrine KATP channels in pancreatic islet cells
.
PLoS One
2014
;
9
:
e91525
46.
Basco
D
,
Zhang
Q
,
Salehi
A
, et al
α-cell glucokinase suppresses glucose-regulated glucagon secretion
.
Nat Commun
2018
;
9
:
546
47.
Matschinsky
FM
,
Wilson
DF.
The central role of glucokinase in glucose homeostasis: a perspective 50 years after demonstrating the presence of the enzyme in islets of Langerhans
.
Front Physiol
2019
;
10
:
148
48.
Miguel-Escalada
I
,
Bonàs-Guarch
S
,
Cebola
I
, et al
Human pancreatic islet three-dimensional chromatin architecture provides insights into the genetics of type 2 diabetes
.
Nat Genet
2019
;
51
:
1137
1148
49.
Mularoni
L
,
Ramos-Rodríguez
M
,
Pasquali
L.
The pancreatic islet regulome browser
.
Front Genet
2017
;
8
:
13
50.
Pasquali
L
,
Gaulton
KJ
,
Rodríguez-Seguí
SA
, et al
Pancreatic islet enhancer clusters enriched in type 2 diabetes risk-associated variants
.
Nat Genet
2014
;
46
:
136
143
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 https://www.diabetesjournals.org/journals/pages/license.