Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Nov 5.
Published in final edited form as: Cell Metab. 2024 Oct 8;36(11):2468–2488.e7. doi: 10.1016/j.cmet.2024.09.006

Multi-omic Mapping of Human Pancreatic Islet Endoplasmic Reticulum and Cytokine Stress Responses Provides Type 2 Diabetes Genetic Insights

Eishani K Sokolowski 1,2, Romy Kursawe 1, Vijay Selvam 1, Redwan M Bhuiyan 1,2, Asa Thibodeau 1, Chi Zhao 3, Cassandra N Spracklen 3, Duygu Ucar 1,2,4,5,**, Michael L Stitzel 1,2,4,5,6,*
PMCID: PMC11798411  NIHMSID: NIHMS2028079  PMID: 39383866

SUMMARY

Endoplasmic reticulum (ER) and inflammatory stress responses contribute to islet dysfunction in type 2 diabetes (T2D). Comprehensive genomic understanding of these human islet stress responses, and whether T2D-associated genetic variants modulate them, is lacking. Here, comparative transcriptome and epigenome analyses of human islets exposed ex vivo to these stressors revealed 30% of expressed genes and 14% of islet cis-regulatory elements (CREs) as stress-responsive, modulated largely in ER- or cytokine-specific fashion. T2D variants overlapped 86 stress-responsive CREs, including 21 induced by ER stress. We linked rs6917676-T T2D risk allele to increased islet ER stress-responsive CRE accessibility and allele-specific β-cell nuclear factor binding. MAP3K5, the ER stress-responsive putative rs6917676 T2D effector gene, promoted stress-induced β-cell apoptosis. Supporting its pro-diabetogenic role, MAP3K5 expression correlated inversely with human islet β-cell abundance and was elevated in T2D β-cells. This study provides genome-wide insights into human islet stress responses and context-specific T2D variant effects.

eTOC Blurb:

This multi-omic study dissects the genome-wide effects of ER stress and pro-inflammatory cytokines on human islet transcriptional regulatory networks and motivates variant-to-function studies linking rs6917676 T2D risk allele to enhanced ER stress-responsive MAP3K5-mediated β-cell apoptosis. Findings suggest MAP3K5 inhibitors (e.g., Selonsertib) could therapeutically reduce stress-induced β-cell apoptosis.

Graphical Abstract

graphic file with name nihms-2028079-f0006.jpg

INTRODUCTION

Type 2 diabetes (T2D) is a complex metabolic disorder characterized by gene-environment factors contributing to pancreatic islet beta cell dysfunction and/or death, and inadequate insulin secretion in response to insulin resistance15. Genome-wide association studies (GWAS) linked DNA sequence variants in >600 loci in the human genome with increased T2D risk610. These variants’ non-coding location, combined with studies demonstrating their significant enrichment in islet cis-regulatory elements (CREs), suggests they contribute to islet dysfunction and failure by altering CRE function and effector gene expression2,4,1114. A subset of T2D-associated variants altered in vivo human islet CRE chromatin accessibility or effector gene expression under steady-state conditions4,1113,1517. However, T2D pathogenesis involves dynamic interactions between genetic variants and environmental stressors1,2,4,5, yet functional effects of putative regulatory non-coding variants on islet transcriptional stress responses to pathophysiologic endoplasmic reticulum (ER) stress and pro-inflammatory cytokine stressors1, are largely unknown.

Proper ER trafficking and protein folding capacity is integral to β-cell protein quality control and insulin synthesis; both genetic and pathophysiologic ER functional perturbations (ER stress) are implicated in islet dysfunction across the diabetes spectrum18,19. Multiple INS mutations in patients with permanent neonatal diabetes lead to ER retention and induce ER stress. Rare variants in genes encoding proteins that prevent and sense protein misfolding (DNAJC31923) or activate and transduce unfolded protein response (UPR) signals (EIF2AK324, EIF2B125, ATF626, WFS127,28) are linked to syndromic, monogenic diabetes. Chronic hyperglycemia elicits sustained demand for insulin production, which can overwhelm the β-cell ER, increasing stress and activating UPR machinery18. Prolonged or excessive ER stress contributes to β-cell dysfunction and death18,19. β-cell dysfunction has been further linked to elevated pro-inflammatory cytokine levels in the blood19,29,30, which activates the NFKB pathway and impairs insulin secretion3032. Although ER and inflammatory stressors have been associated with T2D33,34, our understanding of pancreatic islet responses to each, and how T2D variants modulate these transcriptional programs, is incomplete.

To fill these knowledge gaps, we defined robust transcriptional regulatory programs controlling human islet responses to ER stress and pro-inflammatory cytokines by mapping genome-wide CRE accessibility (ATAC-seq) and gene expression (RNA-seq) in islets exposed to ER stress-inducing agent thapsigargin (250 nM) or pro-inflammatory cytokines IL-1β (25 U/mL) and IFN-γ (1000 U/mL). These stressors and their respective concentrations were selected based upon previous studies using comparable concentrations of thapsigargin15,3538 or IL-1β + IFN-γ3941 to model islet ER stress or cytokine-induced inflammatory responses, respectively. Comparison of stress-responsive genes and CREs revealed complementary stress- and cell type-specific changes in transcriptional regulatory programs and expression of the factors mediating these responses. We identified T2D-associated variants in 38 signals overlapping ER stress- or cytokine-induced CREs as candidate causal variants and linked them to stress-responsive target (putative T2D effector) genes. Targeted variant-to-function analyses linked rs6917676-T T2D risk allele to increased ER stress-responsive CRE accessibility and demonstrated that its putative T2D effector MAP3K5, the only ER stress-responsive gene in the locus, promoted stress-responsive apoptosis in human β-cells and primary human islets.

RESULTS

Comprehensive comparative mapping of human islet ER stress- and cytokine-responsive genes

To define characteristic human pancreatic islet responses to ER stress and pro-inflammatory cytokines, we exposed primary human islets from multiple non-diabetic donors (Table S1) to 24-hour treatment with thapsigargin (vs. DMSO solvent control)15,38,35 or IL-1β+IFN-γ cocktail (vs. untreated control)39, respectively. We determined and compared genome-wide gene expression changes elicited by these T2D-relevant stressors using whole islet RNA sequencing (RNA-seq)42.

In total, ~30% (5,131/17,096) of human islet-expressed, autosomal protein-coding genes (Methods) were modulated by ≥1 stressor compared to their respective controls (Table S2; FC≥1.5 or ≤0.667, FDR<5%). 2,967 genes were differentially expressed (DE) upon ER stress (1,517 induced; 1,450 reduced), whereas 3,443 genes were DE upon cytokine-induced inflammation (1,893 induced; 1,550 reduced) (Table S2). ER stress and cytokine DE genes were largely distinct. For example, ~85% of induced genes were stressor-specific (1,064 ER stress-specific; 1,440 cytokine-specific (Figure 1A; Table S2). Pathway analysis confirmed that thapsigargin treatment activated genes facilitating both homeostatic (e.g., ATF4, ERN1/IRE1α, EIF2AK3/PERK, HERPUD1, HSPA5/BiP) and terminal (e.g., DDIT3/CHOP, MAP3K5) arms of UPR and ER protein processing pathways (Figure 1B; Table S2) central to beta cell function and survival4349 (Figures 1B-C). Importantly, thapsigargin engaged each UPR sensing (e.g., calreticulin/calnexin; BiP) and signaling (e.g., PERK, ATF6, and IRE1) branch (Table S2) linked to islet dysfunction and diabetes genetics, thereby providing a comprehensive perspective on the transcriptional regulatory networks and output of these processes. In addition to anticipated UPR-related signals, ER stress significantly induced cholesterol synthesis/metabolism and amino acid biosynthesis pathways (Table S2), including pharmacologic targets PCSK9 and LDLR with underappreciated roles in beta-cell insulin secretion50,51 and ARG2, a gene with central roles in polyamine synthesis52 that is dysregulated in T2D islets53, suggesting broader and more complex impact of ER stress on cellular functions extending beyond protein folding.

Figure 1. Induced transcriptional responses of human pancreatic islets to ER stress (ERS) and pro-inflammatory cytokines (CYT).

Figure 1.

(A) Heatmap of genes induced by ERS and/or CYT treatment (FDR<5%; FC≥1.5). Induced gene categories are ERS-specific, CYT-specific, or shared between both conditions; gene numbers for each category are shown in parentheses. Note the majority of genes exhibit stress-specific induction. Expression values are z-score scaled. (B) Enriched pathways for induced genes; FDR values and example genes for each pathway are indicated. (C) Examples of enriched pathway genes induced by ERS, CYT, or both. Dot-and-box plots show normalized per-donor islet gene expression (CPM) in ERS (green), CYT (orange), or control samples (gray). ***=FDR<5%, FC≥1.5; ns=not significant. FDRs calculated using Benjamini-Hochberg p-value adjustment. FDR, false discovery rate; FC, fold change; CPM, counts per million. See also Tables S1-2, Data S1.

Consistent with previous reports5456, the most highly-enriched pathways for islet cytokine-induced genes were related to cytokine and chemokine signaling pathways (Figure 1A; Table S2), including NFKB complex members (e.g., NFKB1, NFKBIA) and signaling molecules (e.g., JAK2, STAT2) modulating inflammatory responses, promoting immune cell infiltration, and contributing to islet beta cell dysfunction (Figures 1B-C; Table S2)5763. In addition to canonical pathways, cytokines activated multiple innate immune pathways, including NOD- and RIG-I-like receptor signaling, CGAS/STING, NLRP3 inflammasome, and cytosolic DNA-sensing pathways (Table S2) that may contribute to sensing and signaling potentially immunogenic endogenous dsRNAs provocatively suggested as a non-viral source of autoimmune activation in early T1D64. Finally, 453 genes were induced by both ER and pro-inflammatory cytokine stressors (Figure 1A; Table S2) and enriched in pathways related to: (i) metal ion response, including free radical-scavenging metallothioneins (MT1 genes) associated with reduced insulin secretion upon stress; and (ii) DNA double-strand break (DSB) processing, including RAD9A, involved in repairing DNA damage and DSBs associated with T2D6567 (Figures 1B-C; Table S2).

Among 2,470 genes with reduced stress-responsive expression, ~79% were stressor-specific, including 920 ER stress-specific and 1,020 cytokine-specific genes (Figure S1A; Table S2). PDX1, ADCY5, GLP1R and IGFBP5, which encode factors integral to islet identity and function6871, were reduced upon ER stress (Figures S1B-C; Table S2). In contrast, SLC1A1, COL2A1, NPNT, and ITGA10, which participate in protein digestion/absorption and extracellular matrix (ECM) receptor signaling related pathways important for β-cell function7274, were reduced upon cytokine-induced inflammation (Figures S1B-C; Table S2). Further, 530 genes were reduced by both stressors (Figure S1A; Table S2), including CDC20, CDC45, UGT2B11 and UGT2B15, which are involved in cell cycle and retinol metabolism and crucial for islet function7375,7580 (Figures S1B-C; Table S1).

Together, the profiles indicate that islet thapsigargin and cytokine exposures engage multiple transcriptional responses as anticipated and provide comprehensive genome-wide views of genes and pathways modulated by ER stress and pro-inflammatory cytokines. Comparative analyses suggest these stressors elicit largely distinct, complementary transcriptional responses, inducing multiple stress-specific pathways and repressing islet cell type-specific critical functions.

ER stress induces robust, heterogeneous β-cell responses

To decipher cell type specificity of islet ER and cytokine stress responses, we completed single cell (sc) transcriptome profiling of islets treated with thapsigargin or pro-inflammatory cytokines (Table S1, n=3 donors/condition), yielding 18,945 total single cell transcriptomes (Figure S2A; Table S3). Unsupervised clustering identified each cell type (Figure 2A; Table S3) annotated by classic marker gene expression (Figures S2A-B; Table S3). α- (~38%) and β-cells (~37%) constituted the majority of islet cells (Figure S2C; Table S3). In striking contrast to α- and other islet cell types, β-cells distinctly exhibited increased sensitivity to ER and cytokine stressors, represented by presence of a distinct cluster (marked by *) comprised almost exclusively of stressed β-cells (Figure 2A; cluster composed of 6.53% thapsigargin; 92.91% cytokine; 0.31%; DMSO, 0.25% untreated β-cells).

Figure 2. Single-cell transcriptome analysis of human pancreatic islet ER stress (ERS) and pro-inflammatory cytokines (CYT) responses.

Figure 2.

(A) Uniform Manifold Approximation and Projections (UMAPs) of aggregated single cell transcriptomes from islets exposed to ERS (thapsigargin), CYT (IL-1β + IFNγ) or respective control conditions for 24 hours Table S1, 3 donors/condition). UMAPs are color-coded by cell type annotations (top) or condition (bottom). n=number of cells per cell type (top) or condition (bottom). Black asterisk denotes beta cluster comprised almost exclusively of stressed cells. (B) Scaled fold-change of alpha or beta cell expression of genes induced by ERS or CYT in whole islets. Genes are grouped into genes whose induction is ERS-specific, CYT-specific, or shared between conditions. (C) Response scores for the islet-induced genes in α- or β-cells. ***p<1.0E-10; ns=not significant, two-sided Wilcoxon test. (D) α- or β-cell expression of genes representing induced gene sets in panel B. ***FDR<5%, FC≥1.5; ns=not significant. (E) UMAP of islet scRNA-seq profiles (left) reveals two beta cell clusters (BC) in ER stressed islets (middle), ERS-Beta Cluster 1 (ERS-BC1) or ERS-Beta Cluster 2 (ERS-BC2), respectively (right). Cell numbers are indicated in parentheses. (F) DEGs in ERS-BC1 (top) or ERS-BC2 (bottom) versus DMSO control. DEG numbers are indicated in parentheses. Expression values are z-score scaled. (G) Left, Venn diagram of induced DEGs (FDR<5%, FC≥1.5) in ERS-BC1 or ERS-BC2; right, KEGG, Reactome, and WikiPathways pathways enriched for intersecting vs. unique induced DEGs. FDRs are reported beneath each pathway. Note specific enrichment of apoptosis-related pathways in ERS-BC2. (H) Examples of unfolded protein response (UPR) or apoptosis gene expression in ERS-BC1, ERS-BC2, or DMSO control conditions. ***FDR<5%, FC≥1.5. False discovery rates (FDR) calculated as in Figure 1. FC, fold-change; DEGs, differentially expressed genes. See also Tables S1, S3, Data S1.

Both stressors induced α- and β-cell expression changes (Table S3). To determine relative α- and β-cell contributions to whole islet transcriptional responses, we assessed their expression of 1,020 ER stress-specific, 1,395 cytokine-specific, and 437 shared islet stress response genes (Figure 2B; Table S3). We compared fold changes of the 1,020 ER stress-specific islet induced genes in α- vs. β-cells and found that β-cells display stronger gene induction to ER stress than α-cells (Table S3; p<1.0E-03, two-sided Wilcoxon test). In addition, ER stress induced expression of 98 genes in β-cells and 35 genes in α-cells, 30 of which were shared between the two cell types (Figure S2D; Table S3). Moreover, the magnitude of ER stress-induced induction for these shared genes was higher in β- vs. α-cells (Figure S2E; Table S3; p<1.0E-03, two-sided Wilcoxon test). Together, these data suggest β-cells respond more vigorously to ER stress than α-cells.

To quantify cumulative α- and β-cell responses to ER stress and pro-inflammatory cytokines, we built and assessed three ‘response scores’ representing aggregate expression of (i) 1,020 ER stress-specific; (ii) 1,395 cytokine-specific; or (iii) 437 stress-shared induced genes (Methods). These showed that, while both α- and β-cells contribute to ER stress and cytokine responses, β-cells were more likely to yield a response to both ER stress and cytokines than α-cells (Figure 2C; p<1.0E-10; two-sided Wilcoxon test). For example, DDIT3, S100A6, and MT1F were significantly induced in β-but not α-cells (Figure 2D; Table S3). We observed the same β- vs. α-cell trend for genes with reduced expression in ER-stressed islets (Figures S2F-G). For example, β-cell identity/function genes (MAFB, SCG2, and SHISAL2B8188) were robustly reduced in β-cells (Figure S2H; Table S3).

Further inspection revealed two ER-stressed β-cell subpopulations (Figure 2E), comprising ~94% (Beta Cluster 1 (BC1); 1,700 cells) and ~6% (Beta Cluster 2 (BC2); 105 cells) of the total (Table S3). Response heterogeneity was both ER stress- and β-cell specific (Figure 2E, Figure S2I). ER stress BC1/BC2 subpopulations were detected in all 3 donors (Figure S2J; Table S3), suggesting these are robust, reproducible transcriptional states. We compared BC1 and BC2 transcriptomes to solvent control to identify 113 (96 induced; 17 reduced) or 170 (147 induced; 23 reduced) DEGs, respectively (Figure 2F, Table S3). 89 induced DEGs (~58%) were shared between BC1 and BC2, including canonical ER stress/UPR genes such as DDIT3, ATF4, and HERPUD1 (Figures 2G-H). BC2-specific induced DEGs (n=58) were enriched in cellular death-related pathways comprised by proteasome superfamily genes that degrade misfolded proteins and regulate apoptosis87, such as PSMB8, PSMB9, and PSMB10 (Figures 2G-H). These ubiquitination, degradation, and apoptosis signatures were specific to the BC2 cluster (Figures S2K-L, Table S3).

In summary, scRNA-seq profiling revealed more extensive and vigorous ER stress responses in β- vs. α-cells. β-cell responses comprised of two distinct transcriptional states, including a smaller ER-stressed subset that highly expresses apoptosis-related genes. This β-cell subset may represent a distinct subpopulation more sensitive or vulnerable to ER stress-induced cell death or an inherent heterogeneity in the temporal dynamics of β-cell ER stress responses.

Identification of ER and inflammatory stress-responsive islet cis-regulatory architecture

To determine cis-regulatory elements (CREs) mediating islet ER and cytokine stress responses, we mapped and compared genome-wide CRE accessibility from six donors each in ER-stressed or cytokine-stressed islets vs. their respective DMSO or untreated controls (Table S1) using whole islet assay for transposase-accessible chromatin sequencing (ATAC-seq)88 (Methods). Approximately 14% of CREs (14,968/109,399) were significantly (FDR<5%) remodeled in response to stress; 7,171 CREs were ER stress-responsive (3,375 opening; 3,796 closing) and 8,819 CREs were cytokine-responsive (5,768 opening; 3,051 closing) (Table S4). Consistent with stress-responsive gene expression patterns, the majority of modulated CREs exhibited stress-specific accessibility changes (Figure 3A; Figure S3A; Table S4): among 8,750 opening CREs, 2,982 were ER stress-specific, 5,375 cytokine-specific, with only 393 shared between stressors.

Figure 3. Comprehensive mapping of islet ER stress- (ERS) and pro-inflammatory cytokine- (CYT) induced cis-regulatory elements (CREs).

Figure 3.

(A) Heatmap of human islet cis-regulatory elements (CREs) whose accessibility is increased by ERS and/or CYT treatment (FDR<5%). n=number of CREs per category. Accessibility values scaled using z-scores. (B) Percent of proximal vs. distal opening CREs (≤1kb vs. >1kb to nearest TSS, respectively). Bar chart (right) indicates percentage of opening CREs per condition. (C) Integrative Genomics Viewer (IGV) browser tracks of representative loci highlighting CREs with ERS-specific, CYT-specific, or shared accessibility increases. (D) Per control conditions for responsive genes in representative loci from panel C. ***FDR<5%, FC≥1.5; ns=not significant. (E) Heatmap of enriched transcription factor (TF) motifs in ERS-specific, CYT-specific, or shared opening distal CREs. Color gradient indicates the scaled fold-change of the motif (i.e., motif instances found in target vs. background sequences). *****FDR<1.0E-200; ****FDR<1.0E-100; ***FDR<1.0E-50; **FDR<1.0E-10; *FDR<1.0E-01; ns=not significant. (F) Chromatin footprint analyses indicating average islet chromatin accessibility in IRF8 (left), ATF4 (middle), or STAT1:STAT2 (right) TF binding sites of CYT-specific (left), ERS-specific (middle), or shared opening CREs (right). Number of footprints is indicated at the bottom of each plot. (G) Islet RNA-seq expression levels (CPM) in ERS, CYT, or control conditions for genes encoding TFs with enriched motifs or chromatin footprints in panels E or F. ***FDR<5%, FC≥1.5; ns=not significant. False discovery rates (FDR) calculated as in Figure 1. FC, fold-change; CPM, counts per million. See also Tables S1-2, Table S4 and Data S1.

Approximately 91% of stress-responsive CREs were distal, i.e., >1kb from transcription start site (TSS)89,90 of the nearest expressed gene (Figure 3B; Figure S3B; Table S4), revealing an extensive role for putative enhancers as mediators of both transcriptional stress responses. We associated opening and closing distal CREs with the nearest islet-expressed genes and completed enrichment analyses (Table S4). As anticipated, stress induced concordant islet chromatin accessibility and gene expression changes (ER stress-specific: p=4.5E-16; cytokine-specific: p=1E-62; shared: p=5.9E-12; Fisher’s exact test) (Table S4). For example, we identified intronic ER stress-specific opening CREs in ERN1 and AOPEP (Figure 3C; Table S4), genes significantly induced by ER stress (Figure 3D; Table S2). ERN1 encodes IRE1α, a central ER stress sensor that initiates UPR and catalyzes unconventional splicing of the ER stress factor XBP1, while AOPEP catalyzes N-terminal peptide and amino acid hydrolysis9193. Cytokine-responsive CREs included those within introns of NFKB1 and CALCOCO2 (Figure 3C; Table S4), two genes induced by cytokine-induced inflammation (Figure 3D; Table S2). NFKB1 is a central mediator of inflammatory responses including in β-cells9496, whereas CALCOCO2 encodes a selective autophagy receptor recently identified as a putative T2D effector gene that maintains proper β-cell mitochondrial morphology, insulin granule homeostasis, and insulin content97,98. Further, we identified stress-induced intronic CRE chromatin accessibility increases in NCKAP5 and ARID5B, genes induced by both stressors (Figure 3C-D; Tables S2-3).

To define regulatory drivers of islet ER and cytokine stress responses, we assessed transcription factor (TF) binding motif enrichment in opening distal peaks (Table S4). Motifs for ATF4 (FDR=2.20E-151), CHOP (FDR=4.40E-167), and NFIL3 (FDR=2.75E-38), transcriptional UPR mediators99102, were enriched in ER stress-specific opening distal peaks (Figure 3E; Table S4). In contrast, cytokine-specific opening distal peaks were enriched in motifs for interferon response factors IRF8 (FDR<2.23E-308) and IRF3 (FDR<2.23E-308), as well as the NFKB family member NFKB-p65 (alias RELA) (FDR=3.67E-68) (Figure 3E; Table S4). STAT1 (FDR=4.40E-33), BCL6 (FDR=1.52E-06), and CEBPB (FDR=1.76E-10) TF motifs were enriched in distal peaks opening in response to both islet stressors (Figure 3E). TF footprinting analyses, which integrate TF binding motifs with chromatin accessibility maps103, confirmed significant genome-wide chromatin accessibility increases for ATF4 binding sites upon ER stress (p=2.56E-02) and IRF8 upon cytokine-induced inflammation (p=2.95E-04) (Figure 3F; Table S4). Increased accessibility at these TF binding sites was concordant with expression changes for the genes encoding these TFs: ATF4, DDIT3, and NFIL3 were induced by ER stress; IRF8, IRF3 and RELA were induced by cytokine-induced inflammation; and STAT1, BCL6, and CEBPB were induced by both stressors (Figure 3G).

We also identified concordant ER stress- and cytokine-responsive reductions in CRE accessibility and nearest gene expression (ER stress-specific: p=9E-53; cytokine-specific: p=3.9E-09; shared: p=3.5E-08; Fisher’s exact test) (Table S4). RAB27B and SLC6A17 play key roles in insulin granule exocytosis and amino acid vesicular trafficking, respectively104108. These genes were reduced and linked with closing CREs upon ER stress (Figures S3C-D; Table S4). Similarly, IGF1R and PCSK1, which play pivotal roles in glucose homeostasis and proinsulin-to-insulin processing, respectively109111, were reduced and linked with chromatin closing upon cytokine-induced inflammation (Figures S3C-D; Table S4). SORL1, involved in insulin receptor sorting112 and PAX4, crucial for islet development113,114 were reduced and linked to chromatin closing upon both stressors (Figures S3C-D; Table S4).

TF motif enrichment analysis for closing distal CREs (Table S4) revealed enrichment of PDX1 (FDR=1.33E-05) and MAFA (FDR=2.09E-04) motifs in the ER stress-specific and shared closing distal CREs, respectively (Figure S3E). We observed concordant downregulation of PDX1 and MAFA under these stress conditions (Figure S3F). Given PDX1 and MAFA are TFs central to islet identity, development, and function68,115,116, their downregulation may signify a compensatory response to stress that impairs islet function, thereby altering glucose homeostasis and contributing to T2D.

Together, integrated comparative ATAC-seq profiling and analysis of these islet stress responses revealed that: (i) ER stress and cytokine responses in islets substantially remodel the islet epigenome, particularly modulating distal non-coding CREs (i.e., enhancers); and (ii) each stressor evokes a distinct epigenetic restructuring, mediated by different TFs (e.g., CHOP and ATF4 in ER stress; IRF8 and NFkB-p65 in cytokines) whose own expression (e.g., CHOP-encoding DDIT3, ATF4 upon ER stress) are themselves modulated by that stressor.

T2D-associated genetic variants overlap stress-responsive cis-regulatory elements

After identifying and comparing ER and cytokine stress-responsive cis-regulatory networks, we sought to identify genetic variants associated with diabetes (T2D/type 1 diabetes (T1D) GWAS) or related glycemic traits that might modulate the CREs and processes. Using a set of index and proxy variants collected from multiple GWAS studies and meta-analyses710,117122 (Table S5; Methods), we identified 161 T2D, T1D, or related glycemic trait-associated variants that overlap stress responsive (opening or closing) CREs (Figure 4A; Figure S4A; Table S5). We identified 21 and 24 T2D-associated variants overlapping ER stress- or cytokine-specific opening CREs, respectively (Figure 4A; Table S5). Among these, 11 variants overlapped ER stress-specific opening CREs located <500kb from ER stress-responsive genes (Figure 4B; Table S5), such as AOPEP - a key gene involved in peptide processing93 and robustly induced by ER stress in beta cells (Figure 4C; Tables S2-3). We detected an ER stress-specific induced intronic CRE in AOPEP that harbors T2D variant rs4744423 (Figure 4D; Tables S4-5). CRE chromatin accessibility increases correlated with rs4744423 T2D risk allele dosage (plus strand: T) (Figure 4E). The risk allele is predicted to significantly (p=3.28E-02) increase the binding affinity of TF BATF (Figure 4F; Table S5), but BATF is not islet-expressed. Further scrutiny revealed strong similarity between BATF and FOSB/JUNB heterodimer motifs (HOMER123 similarity score: 0.98), and FOSB is itself an ER stress-induced islet gene (Figure 4G; Table S2). Thus, these data suggest the rs4744423 T2D risk allele may enhance FOSB/JUNB binding in ER stressed islets and concordant AOPEP induction. This is supported by increased AOPEP expression (p<1.0E-02; two-sided Wilcoxon test) in β-cells from donors with T2D (T2D) vs. without (ND) (Figure 4H) from single cell transcriptome data generated in a parallel study (see Data S1).

Figure 4. Type 2 Diabetes (T2D)-associated variants overlapping stress-induced islet CREs.

Figure 4.

(A) Number of T2D- or glycemic trait-associated genome-wide association study (GWAS) variants overlapping opening cis-regulatory elements (CREs). (B) T2D-associated variants overlapping ER stress (ERS)-specific opening CREs <500 kb from TSS of ERS-specific induced genes. T2D association p-values were obtained from Mahajan et al. (2022)10 or Vujkovic et al. (2020)7 for each GWAS Source population listed. NR=Not Reported. (C) Normalized expression of putative T2D variant rs4744423 effector gene AOPEP in ERS, CYT, or control conditions in human islet RNA-seq (left) or scRNA-seq (right) profiles. Left, per-donor islet gene expression (CPM) in treated vs. control conditions. ***FDR<5%, FC≥1.5; ns=not significant. Right, AOPEP expression in α- vs. β-cell scRNA-seq profiles in ERS- or CYT-treated human islets. Dot size indicates percent of AOPEP-expressing cells per cell type; color denotes scaled average AOPEP expression in each. (D) Integrative Genomics Viewer (IGV) browser track of ERS-specific opening CRE containing T2D variant rs4744423. (E) Islet chromatin accessibility (CPM) in donors with rs4744423 TC or TT genotypes (plus strand). Homozygous T2D risk allele (TT) genotype is associated with the highest in vivo chromatin accessibility. (F) Composite logo plot illustrating atSNP174 prediction that rs4744423 T2D risk allele (plus strand T, minus strand A) strengthens a BATF TF binding motif (atSNP ‘SNP impact p-value=3.28E-02). (G) Normalized per-donor islet FOSB expression (CPM) in ERS, CYT or control conditions. ***FDR<5%, FC≥1.5; ns=not significant. (H) Normalized AOPEP expression (CPM) in human islet α- or -cell pseudobulk scRNA-seq profiles of donors with T2D (T2D) vs. without (ND). **p<1.0E-02; ns=not significant, two-sided wilcoxon test. (I) Left, normalized expression (CPM) of putative rs6444081 T2D effector gene in ERS, CYT, or control conditions ***FDR<5%, FC≥1.5; ns=not significant. expression in ERS or CYT-treated islets. Dot size indicates percent -expressing cells; color denotes scaled average ETV5 expression in them. (J) IGV browser track of ERS-specific opening CRE containing T2D variants rs6444081, rs146872661, rs937563893, and rs150111048. (K) Islet chromatin accessibility (CPM) in donors with rs6444081 TT, TC or CC genotypes (plus strand). Homozygous T2D risk allele (CC) genotype is associated with the lowest in vivo chromatin accessibility. (L) Composite logo plot illustrating atSNP174 prediction that rs6444081 T2D risk allele (plus strand C, minus strand G) disrupts an NFE2L2 (indicated by the position weight matrix) TF binding motif (atSNP ‘SNP impact p-value=3.88E-03). (M) Normalized per-donor islet NFE2L2 (protein alias NRF2) expression (CPM) in ERS, CYT or control conditions. **FDR<5%, FC>1. False discovery rates (FDR) calculated as in Figure 1. FC, fold-change; CPM, counts per million. See also Tables S1-5 and Data S1.

Similarly, we identified an ER stress-induced CRE harboring T2D variant rs6444081 whose putative effector gene is ETV5, an insulin secretion modulator124126 induced by ER stress in β-cells (Figures 4I-J; Tables S2-5). T2D rs6444081 risk allele (plus strand: C) was associated with reduced CRE accessibility (Figure 4K) and predicted to significantly disrupt an NFE2L2/NRF2 TF binding motif (p=3.88E-03, Figure 4L; Table S5), which we previously identified as a putative islet chromatin accessibility regulator127. NFE2L2 was ER stress-induced (Figure 4M), and together with KEAP1128130, may facilitate stress-responsive ETV5 activation. These data, therefore, suggest that ER stress increases rs6444081-harboring CRE accessibility and regulates ETV5 activation. However, our data indicate T2D rs6444081-C risk allele reduces chromatin accessibility, presumably by disrupting NRF2 binding, which would contribute to diminished ETV5 induction and islet dysfunction or death. Indeed, Etv5−/− mice exhibit impaired insulin secretion and glucose tolerance defects124. Etv5−/− islets are smaller and contain smaller β-cells than those from wildtype littermates124, and reduced ETV5 expression was previously reported in T2D vs. ND islets125.

We also found 14 T2D-associated variants overlapping 11 cytokine-specific opening CREs <500kb from cytokine-induced genes (Figure S4B; Table S5), including β-cell cytokine-induced GALNT15 (Figure S4C; Tables S2-3) - a member of the GALNT family involved in protein metabolism131,132. In addition, we identified a cytokine-induced intronic CRE in ANKRD28 containing T2D variant rs4685264 (Figure S4D). T2D rs4685264 risk allele (plus strand: G) was associated with increased CRE chromatin accessibility (Figure S4E) and significantly (p=2.84E-03) increased predicted binding affinity for the TF MAX (Figure S4F; Table S5), which was itself a cytokine-induced gene (Figure S4G; Table S2). These data suggest that cytokine exposure increases accessibility of the CRE harboring rs4685264, enabling MAX binding. We posit that the T2D risk allele MAX binding and induction of the putative T2D effector gene GALNT15 to alter protein metabolism in cytokine-exposed islets. Together, these analyses nominated putative T2D variants with context-specific effects on transcriptional processes modulating islet responses to ER stress and cytokine-induced inflammation.

Variant-to-function dissection of ER stress-responsive T2D variant in the SLC35D3 locus

Integrated islet multi-omic data analysis converged to provide variant-to-function insights for T2D-associated variant rs6917676, which overlapped an ER stress-induced CRE residing in an islet enhancer hub133 spanning ~479 kb on chromosome 6 (Figure 5A, magenta bracket). The rs6917676 T2D risk allele (plus strand: T) was associated with increased chromatin accessibility of this ER stress-responsive CRE (Figure 5B, compare GG, TG, and TT islet donors). We previously identified rs6917676 as the expression-modulating variant (emVar) in this CRE using massively parallel reporter assays (MPRA) in mouse MIN6 β-cells, with the T risk allele increasing MPRA activity15. The rs6917676-T risk allele is predicted to strengthen binding of NFIL3 (Figure 5C; Table S5), which was itself induced by ER stress in β-cells (Figure 5D; Tables S2-3). To test if rs6917676-T risk allele was differentially bound by different beta cell nuclear/transcription factor(s) as predicted, we completed electrophoretic mobility shift assays (EMSAs)134 using human EndoC-βH3 nuclear extracts (Figure 5E). EMSA revealed robust T allele-specific binding (red arrows) in untreated, ER stressed, and DMSO solvent control β-cell extracts, consistent with detectable NFIL3 protein in EndoC-βH3 cell nuclear extracts for each of these conditions (Figure S5B).

Figure 5. Variant-to-function analyses link rs6917676 to ER stress-responsive CRE accessibility, MAP3K5 expression, and increased β-cell apoptosis.

Figure 5.

(A) Integrated Genomics Viewer (IGV) browser track of 1 Mb window (blue gene annotations) centered on T2D-associated variant rs6917676 and enhancer hub (magenta) identified by Miguel-Escalada et al. (2019)133. Enhancer hub encompasses multiple enhancers, with one (inset) mapping to an ERS-specific, opening distal CRE containing T2D variants rs6937795 and rs6917676. All genes within the 1Mb interval are shown; ERS-induced (green check), non-responsive (red “X”), or non-expressed/non-protein coding (gray text) are indicated. (B) Normalized islet chromatin accessibility levels (CPM) in donors, stratified by rs6917676 plus strand genotype (GG, TG or TT). Note in vivo chromatin accessibility increases with T2D risk allele (T). (C) Composite logo plot illustrating atSNP174 prediction that rs6917676 T2D risk allele (plus strand T) creates an NFIL3 TF binding motif ( atSNP ‘SNP impact p-value=2.37E-03). (D) Left, normalized per-donor NFIL3 expression (CPM) in ERS- or CYT-treated islets versus their respective controls. ***FDR<5%, FC≥1.5; ns=not significant. Right, α- or β-cell scRNA-seq NFIL3 expression in ERS- or CYT-treated islets (right). Dot size indicates percent NFIL3-expressing cells per cell type; color indicates scaled average NFIL3 expression in each. (E) Electrophoretic mobility shift assay (EMSA) using untreated (Untx), DMSO solvent control, or thapsigargin (Tg)-treated human EndoC-βH3 cells nuclear extracts (NE). Red arrows highlight nuclear factors specifically binding T2D risk allele rs6917676-T. Representative image from n=3 EMSAs. (F) Normalized per-donor expression (CPM) of putative rs6917676 T2D effector gene MAP3K5 in human islets in ERS, pro-inflammatory cytokine (CYT), or control conditions. ***FDR<5%, FC≥1.5; ns=not significant. (G) α- vs. β-cell MAP3K5 scRNA-seq in ERS- or CYT-treated human islets. Dot size indicates percent MAP3K5-expressing cells in each cell type; color indicates scaled average MAP3K5 expression in each. (H) Normalized MAP3K5 expression (CPM) in human islet α- vs. β-cell pseudobulk scRNA-seq profiles from donors with T2D (T2D) vs. without (ND). **p<1.0E-02; ns=not significant, two-sided Wilcoxon test. (I) Correlation plot demonstrating significant (p=9.30E-04) inverse relationship between normalized MAP3K5 expression (CPM) and β-cell-to-endocrine cell proportions for each of 48 human islet donors from a separate study (see Data S1). (J) Percent apoptosis (Annexin V+ cells) detected in human EndoC-βH3 cells exposed to 500 nM thapsigargin vs. DMSO solvent control after MAP3K5 (shMAP3K5) or non-targeting shRNA control (nt-shCTRL) knockdown. n=5 biological replicates per condition. **p<1.0E-02; ns=not significant, unpaired two-tailed Student’s t-test. (K) Percent apoptotic (Annexin V+) cells detected in human islets from five donors incubated with MAP3K5 inhibitor Selonsertib (2 μM) or left untreated (0 μM) and exposed to glucolipotoxic (+Palmitate; 0.4 mM Palmitate +25 mM glucose) or control (-Palmitate; 25mM glucose) conditions for 72 hours. *p<0.05; ns=not significant, paired two-tailed Student’s t-test. False discovery rates (FDR) calculated as in Figure 1. FC, fold-change. See also Tables S1-5 and Data S1.

Islet promoter capture Hi-C (pcHi-C) data133 link this CRE to MAP3K5, PEX7, and IL20RA promoters (Figure 5A). MAP3K5 was the only of these and other genes in the locus (MAP7, SLC35D3, and IFNGR1) induced by ER stress – specifically in beta cells – thereby nominating it as the likely effector gene of this variant (Figures 5F-G; Figure S5A; Tables S2-3). These data suggest T2D risk allele rs6917676-T contributes to islet dysfunction or death by increasing ER stress-responsive MAP3K5 expression via increased NFIL3 binding at this ER stress-responsive opening CRE. Consistently, we detected increased MAP3K5 expression (p<1.0E-02; two-sided Wilcoxon test) in T2D vs. ND donor β-cells (Figure 5H) from a parallel islet single cell transcriptome study (see Data S1).

MAP3K5 encodes ASK1, a pro-apoptotic MAPK kinase reported as ER stress-responsive in MIN6 β cells135 that phosphorylates JNK and p38136. Ask1/Map3k5 knockdown or germline deletion increases MIN6 survival and reduces islet caspase activity, respectively135. Higher MAP3K5 expression in ER stress-BC1 vs. apoptosis-related ER stress-BC2 clusters suggest that elevated MAP3K5 expression may promote or enhance an apoptotic fate in ER-stressed β-cells, after which it is not required (Figure S5C; Table S3). Consistent with this pro-apoptotic role, MAP3K5 expression was significantly associated with reduced beta cell percentages in human islets (Figure 5I). Therefore, to test if MAP3K5 modulates ER stress-responsive apoptosis in human beta cells, we assessed if MAP3K5 shRNA knockdown altered apoptosis in EndoC-βH3 cells exposed to a (patho)physiologic range of thapsigargin concentrations (125–2000 nM). We achieved ~80% knockdown of MAP3K5 (Figures S5D-E), and western blot analyses confirmed that this knockdown nearly abrogated ER stress-induced phosphorylation of p38, one of its targets137,138, in EndoC-βH3 cells (Figures S5F-G). Importantly, MAP3K5-deficient cells exhibited significantly fewer apoptotic (Annexin V+) cells compared to the non-targeting shRNA control cells exposed to pathophysiologic thapsigargin concentrations (Figure 5J; Figure S5E). These data align with in vivo models of diabetes, where germline Map3k5/Ask1 deletion significantly reduced beta cell apoptosis in Akita mice carrying a permanent neonatal diabetes- (PNDM) linked and islet ER stress-inducing Ins2C96Y mutation139.

Next, we tested if pharmacologic MAP3K5 inhibition reduced stress-responsive apoptosis of primary human islets (Table S1) exposed to glucolipotoxicity, another T2D-associated pathophysiologic stress condition, using Selonsertib. Selonsertib is a MAP3K5 (alias ASK1) inhibitor140, and its structural analog GS-444217141,142 have been shown to improve diabetic nephropathy by targeting p38 in pre-clinical rodent models of diabetes143,144. Randomized placebo-controlled double-blind Phase 2 clinical trials (Clinical Trial Identifier: NCT04026165)145 for diabetic complications, such as diabetic kidney disease146,147, have been completed148, and Selonsertib has been approved for, and is currently undergoing, Phase 3 clinical trials to prevent/treat moderate to advanced diabetic nephropathy149151. As anticipated, glucolipotoxic exposure increased apoptosis in primary human islets from five donors (Figure 5K; Figures S5H-I; compare percent Annexin V+ cells in ‘+Palmitate, 0 μM Selonsertib’ vs. ‘-Palmitate 0 μM Selonsertib’, p=0.045). Importantly, Selonsertib treatment significantly reduced glucolipotoxicity-induced apoptosis in these donor islets (Figure 5K; Figures S5H-I; compare Annexin V+ cells in ‘+Palmitate, 0 μM Selonsertib’ vs. ‘+Palmitate, 2 μM Selonsertib’, p=0.047). Taken together, these variant-to-function analyses suggest that the T2D-associated rs6917676-T risk allele contributes to T2D risk or progression by enhancing ER stress-responsive islet CRE accessibility and MAP3K5 expression to promote, sensitize, or enhance stress-responsive islet beta cell apoptosis.

DISCUSSION

This study provides genome-wide insights into the transcriptional regulatory circuitry mediating pancreatic islet stress responses, particularly to ER stress and pro-inflammatory cytokines, two pathophysiologic stressors implicated in T2D pathogenesis. Through comprehensive RNA-seq and ATAC-seq analyses, we identified distinct sets of genes and CREs responsive to ER stress and cytokine-induced inflammation. The majority of stress-responsive genes and CREs were specific to either ER stress or cytokines. scRNA-seq analyses uncovered α- and β-cell specificity of these responses. Context-specific islet response to ER stress and cytokines are intriguing yet not entirely unexpected. The specificity likely reflects a finely tuned cellular mechanism allowing islets to adapt and tailor responses to diverse pathophysiologic stimuli. For example, ER stress predominantly triggered pathways related to protein folding and secretion, both crucial for β-cells’ insulin-producing function152. In contrast, cytokine treatment activated pro-inflammatory and signaling pathways that can interfere with islet identity and function58,153.

scRNA-seq profiling of these human islet stress responses revealed cell type-specificity of responses to ER stress and cytokine-induced inflammation. Although these data suggest β-cells respond to stress more extensively than α-cells, they also indicate α-cells are not devoid of response when faced with stress. As previous studies have primarily focused on β-cells due to their direct role in insulin production and extensive links to diabetes pathology15,153164, comparative scarcity of α-cell pathophysiologic studies underscores the significance of our findings and suggests that they, too, are not unaffected bystanders to islet stress. Recent studies have begun to uncover potential α-cell roles in islet pathological responses in diabetes. For example, α-cells exhibit autoimmune-induced ER stress signatures in T1D165, yet are protected from apoptosis under metabolic stress166,167. It was suggested this could be due to higher α- vs. β-cell production of pro-survival proteins such as BCL2L1166,168. Supporting these reports, basal BCL2L1 expression is higher in α- vs. β-cells (Table S3). These emerging data warrant further islet α-cell biologic studies and scrutiny of their putative role(s) in diabetes risk and progression. Our data also uncovered heterogeneity in β-cell responses to ER stress, marked by the presence of two transcriptionally distinct putative β-cell subpopulations. One subset (ER stress-BC1) reflected the activation of bona fide ER stress response genes and pathways (e.g., DDIT3 and ATF4), whereas the smaller subset (ER stress-BC2) featured induction of apoptosis-related genes (e.g., PSMB8 and PSMB9). Interestingly, this apoptotic β-cell subpopulation was detected in all donors. These findings suggest a fraction of β-cells are prone to ER stress-induced cell death, which could contribute to β-cell death associated with T2D169,170.

Epigenetic stress responses were concentrated in distal regulatory regions of the genome, highlighting the noncoding genome’s importance in modulating cellular stress responses. Stress-responsive opening CREs were enriched in the binding sites of critical TFs (e.g., ATF4 upon ER stress, IRF8 upon cytokine-induced inflammation). Genes encoding these TFs were also activated upon these stressors, suggesting that cellular responses are tightly regulated at the epigenetic level by the activation of critical TFs as well as by the increased chromatin accessibility at their binding sites. By overlapping the locations of T2D-associated genetic variants and stress-responsive islet CREs, we uncovered 52 variants residing in 38 ER or cytokine stress-induced CREs which we hypothesize contribute to T2D etiology by altering their use or activity. Although the identification of stress-responsive CREs, their overlap with T2D-associated variants, and targeted allelic analyses implicate this subset of T2D variants as genetic modulators of these responses, larger sample sizes are needed to formally demonstrate their allelic effects on stress-responsive chromatin accessibility and gene expression using allelic imbalance or quantitative trait locus approaches. Additionally, the exploration of additional T2D-associated pathophysiologic stressors (e.g., glucolipotoxicity or oxidative stress) or various stimuli, and their interaction with genetic variants, could further help elucidate the complex molecular landscape of T2D, stratify T2D association variants/signals into functional bins, and identify therapeutic gene targets and pathways.

For a subset of loci, this study nominated putative causal T2D variants and provided a range of variant-to-function insights into their context-specific effects on islet ER stress- and cytokine-responsive programs. This includes rs6917676, which overlaps an ER stress-responsive CRE. Human islet pcHi-C data from Ferrer and colleagues converge with RNA-seq from this study to nominate MAP3K5 as the target gene of this CRE and the T2D effector gene for this genetic association signal. MAP3K5 encodes MAP3K5 (alias ASK1), a kinase that promotes apoptosis via activation of JNK and p38 signaling pathways135,171173. Our variant-to-function dissection of this locus suggests a mechanistic model in which the rs6917676 T2D risk allele (T) increases ER stress-induced CRE accessibility and MAP3K5 expression to promote excessive apoptosis and exacerbate β-cell loss in T2D. The apoptosis-promoting association of elevated MAP3K5 in stressed β-cells seems to be reflected in the scRNA-seq data, wherein the apoptosis-related ER stress-BC2 beta cluster had reduced MAP3K5 expression compared to the ER stress-BC1 cluster, and it fits with previous in vivo studies demonstrating that germline Map3k5/Ask1 deletion significantly reduces islet β-cell apoptosis in Ins2Akita mice harboring a permanent neonatal diabetes (PNDM)-linked mutation that causes proinsulin misfolding and ER stress139. Moreover, MAP3K5 expression inversely correlated with β-cell abundance in a 48-donor islet scRNA-seq cohort, and T2D donors in this cohort exhibited higher β-cell MAP3K5 expression and significantly fewer β-cells comprising their islets. Finally, pharmacologic MAP3K5 inhibition reduced glucolipotoxicity-induced islet apoptosis, suggesting that: (i) elevated MAP3K5 expression or activity may contribute more broadly to T2D-associated pathophysiologic stress-induced apoptosis; and (ii) Selonsertib, the MAP3K5 inhibitor in Phase II/III clinical trials to prevent or treat diabetic nephropathy, might be an effective primary intervention to combat progression to T2D by preserving mass and function of stressed β-cells. More broadly, these findings highlight the significance of studying GWAS variants in the context of stress conditions, which more closely reflect cellular state(s) during disease, including for T2D.

LIMITATIONS OF STUDY

Despite uncovering islet responses to T2D-associated pathophysiologic factors, there are potential limitations. First, the focus on a single time point may not represent fully these stress response dynamics nor capture the compendium of stress-responsive CREs and genes engaged by chronic stressors. Future studies could employ longitudinal designs for more comprehensive understanding of these temporal cellular adaptation mechanisms. Second, although thapsigargin15,3538 and IL1β+IFNγ3941 exposure to model T2D-relevant islet ER or cytokine responses, respectively, is based on precedent in the literature, they likely miss features of the complex, progressive nature of in vivo islet dysfunction in T2D. This may limit direct translational relevance of our findings. Future studies employing more sophisticated organoid, pancreatic slice, or animal models with transplanted human (pseudo)islets should help to better understand the intricate in vivo milieu contributing to islet dysfunction in T2D.

Despite these potential limitations, the comprehensive, comparative multi-omic mapping we describe provides important mechanistic insights into human islet responses to two (patho)physiologic stressors: ER stress and cytokine-induced inflammation. Importantly, these maps enabled nomination of candidate causal T2D variants that likely contribute to T2D risk or progression by modulating these responses. These findings support growing literature revealing importance of cell- and context-specific responses in the pathophysiology of, and approaches to combat, islet dysfunction in T2D. This study both enhances understanding of T2D pathogenesis and provides potential genetics-based avenues or insights, such as repurposing MAP3K5/ASK1 inhibitors (e.g., Selonsertib) to combat ER stress-induced β-cell apoptosis, as targeted interventions to preserve β-cell function under pathophysiologic ER stress.

STAR METHODS

RESOURCE AVAILABILITY

Lead Contact

Further information and requests for resources and reagents should be directed to and will be fulfilled by Lead Contact Michael L. Stitzel (michael.stitzel@jax.org).

Materials Availability

This study did not generate new unique reagents.

Data and Code Availability

EXPERIMENTAL MODEL AND STUDY PARTICIPANT DETAILS

Study Subjects and Primary Islet Culture

Fresh human cadaveric pancreatic islets were procured from Prodo Labs or the Integrated Islet Distribution Program (IIDP) and were required to have a minimum of 80% purity as measured by dithizone and 90% viability. Because the islets are from de-identified cadaveric organ donors, the study was determined to be human subjects exempt. The characteristics of islet donors are listed in Table S1. The mean and standard deviation in donor age was 47.6 ± 12.4 years. Mean and standard deviation in donor BMI was 30.1 ± 7.1. Islets were obtained from 22 males and 13 females. Upon arrival, cells were transferred into PIM(S) media (Prodo Labs) supplemented with PIM(ABS) (Prodo Labs) and PIM(G) (Prodo Labs) and incubated in a T-150 non-tissue culture treated flask (VWR) for recovery at 37°C and 5% CO2 overnight. The following day, media was changed to CMRL (10% FBS, 1% Glutamax) supplemented with either 0.025%v DMSO, 250nM thapsigargin or 25 U/mL of IL1β + 1000 U/mL of IFNγ (R&D Systems). After 24-hr incubation at 37°C and 5% CO2, nuclei and total RNA were isolated for RNA-seq and ATAC-seq library preparation as previously described127.

EndoC-βH3 Human β-cell Line

For validation experiments, we used EndoC-βH3 (CVCL_IS72; age at sampling 9FW; sex unspecified) an engineered cell line resulting from the lentiviral transfection of SV40LT expressed under the control of the insulin promoter and hTERT that can be excised by the Cre recombinase upon addition of tamixofen175. This cell line has been shown to respond to glucose stimulus by secreting insulin and serves as an adequate model for studying human β-cells. EndoC- βH3 cells were obtained from Human Cell Design (1 place Pierre Potier, 31106 Toulouse, France) and cultured as described previously175. Briefly, EndoC- βH3 cells were maintained at 37C and 5% CO 2 atmosphere, in Advanced DMEM/F12 media (Gibco) consisting of 3g/L of D-glucose, supplemented with 2mM Glutamax (Gibco), 100 units/mL penicillin (Gibco), 100 ug/mL streptomycin (Gibco), 2% albumin from bovine fraction V (SIGMA), 50 uM 2-mercaptoethanol (SIGMA), 10 mM nicotinamide (SIGMA), 6.7 ng/mL sodium selenite (SIGMA) and 10ug/ml Puromycin (Calbiochem) on ECM (Sigma) and Fibronectin (Sigma) coated flasks175.

METHOD DETAILS

RNA-seq Library Preparation and Sequencing

Human islet RNA-seq libraries were prepared from total RNA using the stranded TruSeq kit (Illumina). ERCC Mix 1 or Mix 2 spike-ins were randomly added to each sample (Thermo Fisher, catalog #4456740) and sequenced on Illumina NovaSeq S4 as previously described127. The paired-end (2×150 bp) RNA-seq FASTQ files for each islet were aligned against the human genome (GRCh38/hg38) using STAR176 and counts were generated using QoRTs177 (Table S2; Data S1).

RNA-seq Analyses

Genes were annotated using Ensembl178, and only genes in autosomal chromosomes were considered for downstream analysis. Non-protein coding genes in autosomal chromosomes, including RNA and pseudogenes (annotated as ‘transcribed_unprocessed_pseudogene’, ‘processed_pseudogene’, ‘lncRNA’, ‘unprocessed_pseudogene’, ‘TR_V_pseudogene, snRNA’, ‘misc_RNA’, ‘rRNA_pseudogene’, ‘IG_V_pseudogene’, ‘IG_C_pseudogene’, ‘TEC, scRNA’, ‘translated_processed_pseudogene’, ‘vault_RNA’, ‘sRNA’, ‘pseudogene’, ‘transcribed_unitary_pseudogene’, ‘transcribed_processed_pseudogene’, ‘unitary_pseudogene’, ‘miRNA’, ‘snoRNA’, ‘rRNA’, ‘TR_J_pseudogene’, ‘ribozyme’, ‘IG_J_pseudogene’, ‘scaRNA’, ‘translated_unprocessed_pseudogene’, and ‘IG_pseudogene’) were filtered out. The remaining (protein coding) genes were then filtered for expression by requiring >0 CPM in ≥8 samples, and ERCCs were filtered for expression by requiring >5 reads in ≥2 samples. Normalization of protein coding genes with ERCC was performed using RUVSeq179, which also estimated unwanted variation (W_1) in the data. Surrogate variable analysis was then performed using svaseq180, and the surrogate variables that explained >10% of variance in the data (n=3) were considered in downstream analysis. Genes were then tested for differential expression (FDR<5%; |LFC|≥0.585) between their respective control (DMSO; untreated) and treatment (thapsigargin; IL-1β+IFN-γ) conditions (Table S2), with gene expression adjusted for age, sex, batch, BMI, surrogate variables and W_1, using edgeR’s181 ‘tagwise’ and robust dispersion estimation parameter on TMM normalized counts. FDR was calculated using Benjamini-Hochberg p-value adjustment. Differentially expressed (DE) genes were classified as specific or shared using a Venn diagram, and were input into DAVID182 to find the enriched pathways (FDR<10%) using KEGG183, Reactome184, and WikiPathways185 (Table S2).

scRNA-seq Library Preparation and Sequencing

After a 24-hr treatment, as described above, islets were treated with Accutase for 8–10 min at 37°C to generate a single cell suspension. Cells were then washed and suspended in Staining buffer (PBA, 2%BSA, 0.01%TweenS), and immediately processed as follows: Incubate with Fc Blocking reagent (FcX, BioLegend) 10 minutes at 4 °C before adding 0.5ug of a unique Cell Hashing antibody (TotalseqTM-A0251 to A0257 anti-human hashtag antibody, Biolegend) for 20 minutes at 4 °C, then wash cells 2 times with Staining buffer and once with PBS+0.04%BSA. Cell viability was assessed on a Countess II automated cell counter (ThermoFisher), and up to 30,000 cells (~5,000 cells from each hash-tagged (HTO) sample) were loaded onto one lane of a 10X Chromium Controller. One single cell suspension was loaded twice, i.e. added to two lanes of the 10x chip, and two gene expression and two HTO libraries were generated in total, which were processed into a single set of gene expression and HTO outputs. Single cell capture, barcoding, and library preparation were performed using the 10X Chromium platform V3 chemistry according to the manufacturer’s protocol (#GC000103). cDNA and libraries were checked for quality on Agilent 4200 Tapestation, quantified by KAPA qPCR, and pooled and sequenced on an Illumina NovaSeq 6000 S2/S4 flow cell lane, targeting an average sequencing depth of 50,000 reads per cell. Illumina base call files for all libraries were converted to FASTQ using Illumina’s bcl2fastq186. FASTQ files were associated with the gene expression libraries, aligned to the GRCh38.93 reference genome and merged, including all transcribed unitary pseudogenes, using the 10x Genomics Cell Ranger’s count pipeline187,188. FASTQ files representing the HTO libraries were then processed into hashtag-count matrices using CITE-seq-Count189 (Table S3; Data S1).

scRNA-seq Clustering and Annotation

Sample identities were determined using demuxlet190. Ambient RNA for each islet was removed using SoupX191 by setting contamination fraction to 20%. These SoupX ‘cleaned’ data were then demultiplexed based on enrichment of HTO using Seurat192. Only cells with >2000 genes <40% mitochondrial mapping were included in downstream analysis. Doublet cells were then identified using Scrublet193 and removed. To remove any remaining potential doublets or multiplets, cells in the >0.95 quantile with respect to the number of genes expressed were discarded (Table S3). These data were then merged into a single object using Seurat192 and corrected for batch effect using Harmony194. Seurat’s192 ‘FindClusters’ was implemented to identify cell clusters, which were then annotated for cell type identity using islet marker genes (Table S3). Seurat clusters expressing more than one marker gene were classified as doublets and removed from downstream analyses.

scRNA-seq Data Analyses

To generate response scores, differentially expressed (DE) genes from bulk data were curated into ER stress-specific, CYT-specific and shared response gene modules. UCell’s195 ‘AddModuleScore_UCell’ was then used to calculate each module’s enrichment (i.e. response) scores (Table S3). To identify expressed genes between the control (DMSO; untreated) and treatment (thapsigargin; IL-1β+IFN-γ) conditions in alpha and β-cells, Seurat’s192 ‘FindMarkers’ was implemented using the MAST196 test and adjusted with respect to batch and disease state (Table S3). This methodology was also implemented on only those genes that were detected in a minimum of 10% of cells in either of the populations being compared to identify DE genes between (i) ER stress-BC1 vs. DMSO (FDR<5% and |LFC|≥0.585), (ii) ER stress-BC2 vs. DMSO (FDR<5% and |LFC|≥0.585), (iii) ER stress-BC1 vs. ER stress-BC2 (FDR<5%), and (iv) thapsigargin vs. DMSO (FDR<5% and |LFC|≥0.585) in alpha and β-cells comparisons (Table S3). To find the alpha-specific, beta-specific, and common DE genes upon ER stress, a Venn diagram was created (Table S3). DE genes were input into DAVID182 to determine significantly enriched pathways (FDR<10%) using KEGG183, Reactome184, and WikiPathways185 (Table S3).

ATAC-seq Library Preparation and Sequencing

Human islet ATAC-seq libraries were prepared following the Active motif ATAC prep kit (Active motif catalog# 53150). Briefly, 50 islet equivalents (50,000 cells) per sample were transposed in triplicate, libraries were barcoded, pooled into three-islet batches, and sequenced using 2 × 150 bp Illumina NovaSeq S4 chemistry as previously described127. Paired-end (2×150 bp) ATAC-seq FASTQ files for each islet were trimmed using Trimmomatic197 and aligned against the human genome (GRCh38/hg38) using BWA-MEM198. Duplicate reads were removed, and the remaining reads were shifted as previously described199,200. Using SAMtools201, technical replicates were merged and peaks were called using the MACS2202 ‘BAMPE’ parameter. TDF files were generated using IGVTools203 to visualize peaks on IGV203. Separate consensus peak sets for ER stress and CYT samples were generated by considering peaks that were present in at least 2 samples, and those peaks that mapped to ENCODE DAC Exclusion List Regions204 were excluded using DiffBind205. The union of all peaks from ER stress and CYT samples was then determined using GenomicRanges206, and counts were normalized using CPM (Table S4; Data S1).

ATAC-seq Data Analyses

Only peaks in autosomal chromosomes were considered, which were then filtered for depth by requiring >0 CPM in ≥8 samples. Surrogate variable analysis was then performed using svaseq180, and the surrogate variables that explained >10% of variance in the data (ER stress: n=2; cytokines: n=3) were considered in downstream analysis. Peaks were then tested for differential accessibility (FDR<5%) between their respective control (DMSO; untreated) and treatment (thapsigargin; IL-1β+IFN-γ) conditions (Table S4), with accessibility adjusted for age, sex, batch, BMI, and surrogate variables using edgeR’s181 ‘tagwise’ and robust dispersion estimation parameter on TMM normalized counts. FDR was calculated using Benjamini-Hochberg p-value adjustment. Peaks were then annotated to the nearest expressed protein-coding gene extracted from GENCODE v35207 in islets using HOMER’s123 ‘annotatePeaks.pl’ command. Peaks with distance ≤1kb to the nearest expressed gene’s TSS were considered proximal, and the other peaks were considered distal89,90. IRange’s206‘subsetByOverlaps’ function was then used to classify proximal and distal peaks as specific or shared (Table S4).

Enrichment and Footprinting Analysis

Nearest genes to differentially accessible (DA) peaks were used as input into DAVID182 to find the enriched pathways (FDR<10%) using KEGG183, Reactome184, and WikiPathways185 (Table S4). TF motifs present in the DA peaks were found using HOMER’s123 ‘findMotifsGenome.pl’ command. FDR of TFs was calculated using Benjamini-Hochberg p-value adjustment, and the fold change was calculated by dividing `% of Targets Sequences with Motif’ by the `% of Background Sequences with Motif` (Table S4). Using SAMtools201, samples of the same control or treatment conditions were merged, and peaks were called using MACS2202 with ‘BAMPE’ parameter. HINT-ATAC103 was used to identify TF footprints and to calculate differences in TF activity between the respective control and treatment conditions (Table S4).

Overlapping Genetic Variants with Peaks

Index variants associated with T1D, T2D, and glycemic traits (fasting glucose, fasting insulin, HbA1c, 2-hour glucose, HOMA-B, HOMA-IR, proinsulin, modified Stumvoll insulin sensitivity index, and disposition index) were obtained from the largest and most recent genome-wide association meta-analyses for each trait710,117122 (Table S5). Proxy variants in strong linkage disequilibrium (LD) with the index variant were defined as any variant with r2≥0.75 with the index variant calculated using the 1000 Genomes Phase 3 reference panel208, accessed through ‘https://ldlink.nih.gov’, and the global ancestry group that most closely matched the original GWAS meta-analysis. All individuals in the reference panel were used for GWAS meta-analyses of multi-ancestry populations. When necessary, index and proxy variants were lifted over to hg38 genome. Variants that did not lift over were excluded from further analyses (Table S5). Index and proxy variants with a reference SNP ID (rsID) assigned by dbSNP209, accessed through ‘https://www.ncbi.nlm.nih.gov/snp/’, were then overlapped with DA peaks using IRange’s206 ‘findOverlapPairs’ function to identify T2D variants that overlap stress-responsive peaks (Table S5). T2D variants overlapping stress-responsive peaks and located <500kb from the nearest up- or downregulated gene were then entered into atSNP174, accessed through ‘http://atsnp.biostat.wisc.edu’, to identify all TF motifs being disrupted by the variant in the sense or antisense strand with a ‘SNP impact p-value’ <0.05. This list of motifs was compared to the list of enriched TF motifs identified by HOMER123 (as described above) to determine relevant TF motifs (Table S5). ATAC-seq read pileups were used to infer the genotypes of donors for the T2D variants using pysam201,210 (Table S5).

shRNA knockdown in EndoC-βH3

Plasmid pLKO-puro shRNA clones (Mission shRNA) were purchased from Sigma (SHC016 (shCTRL); TRCN0000000993 (shMAP3K5). Lentivirus was produced in HEK293T cells co-expressing the shRNA plasmid together with psPAX2 packaging plasmid and pVSVG envelope plasmid (Addgene). Virus was concentrated using Lenti-X Concentrator (Takara) and titer quantified using p24 ELISA antigen assay (Takara). MOI=5 was used to transduce 1×10211 EndoC-βH3 cells in culture media without pen/strep and puromycin. Cells were collected for RNA extraction 96 hrs post transduction using TRIZOL (Invitrogen), phase separation was achieved using Chloroform. Isopropanol was used for RNA precipitation using glycogen as a carrier, the pellets were washed using 75% ethanol, air-dried, and resuspended in DEPC water. RNA was measured using Qubit RNA HS Assay (Applied Biosystems). Total RNA was used to perform qPCR using RNA to CT kit (Applied Biosystems) and FAM-Taqman probes (Invitrogen) and analyzed on QuantStudio 7 (Applied Biosystems) normalized to TBP/HPRT1 Taqman probes (Figure S5D; Data S1).

Flow cytometry analysis of EndoC-βH3 cells

Eighteen hours post-transduction, media was changed to pen/strep and puromycin complete media with 0, 125, 250, 500, 1000, 1500, 2000 nM thapsigargin (Sigma Aldrich) dissolved in DMSO or 0.5% DMSO solvent control (VWR). Ninety hours after transduction, cells were collected using Trypsin (Gibco) and stained using PE-Annexin V Apoptosis Detection Kit (BioLegend) according to manufacturer’s instructions. The samples were assessed on Fortessa (BD Sciences) and Annexin V-positive cells were analyzed and quantified using FlowJo Software (BD Sciences) (Figure 5J; Figure S5E; Data S1).

Electrophoretic mobility shift assay (EMSA)

EMSAs were completed as previously described134. Nuclear extracts were prepared from EndoC-βH3 cells using NE-PER Extraction kit (PIERCE), quantified using Pierce BCA protein assay kit (PIERCE), and stored in −80°C until use. 21-bp biotin end-labeled, and unlabeled complementary oligonucleotides were designed to the variant rs6917676 (5’Biosg/TAATGACTGT[G/T]TTCTTAAGAT-3’, Integrated DNA Technologies), and double stranded probes were generated for both alleles. The Lightshift EMSA optimization and control kit (Thermo Scientific) was used according to the manufacturer’s instructions. Each reaction consisted of a 10x binding buffer, Poly Di-Dc, 4μg of nuclear extract, and 200nM of labeled probe. Reactions were incubated at 25°C for 25 minutes and separated on a 6% TBE gel (Invitrogen). DNA-protein complexes were detected using Lightshift Chemiluminescent Nucleic Acid Detection kit (PIERCE) according to manufacturer’s protocol. EMSAs were repeated at least three times and yielded comparable results. See Data S1.

Cell extracts and western blotting

Nuclear extracts were prepared from EndoC-βH3 cells after 24-hour incubation with 500 nM thapsigargin or DMSO solvent control using the NE-PER Extraction kit (PIERCE) and quantified using BCA protein assay kit (PIERCE). Ten micrograms of EndoC-BH3 cytosolic or nuclear extracts were loaded on a 4–12% Tris-Glycine gel (Biorad). The proteins were transferred onto a 0.2 uM PVDF membrane (Biorad) using Turboblot (Biorad). The membrane was blocked with 5% Milk in TBST solution (Tris buffer saline + 0.005% Tween 20) for 1 hour at room temperature and incubated with primary antibodies (STAR Methods) in 5% BSA in TBST solution at 4°C overnight. The membrane was washed three times with TBST and incubated with the secondary HRP-linked antibody in 5% BSA+TBST solution for an hour. After incubation, the blot was washed with TBST and developed using Supersignal West Pico PLUS Chemiluminescent Substrate (PIERCE) on a Biorad ChemiDoc Touch imager (Figure S5B). For phosphorylated and total p38 western blots and analyses (Figures S5F-G), cell extracts were prepared using RIPA buffer (supplemented with PMSF and ROCHE protein inhibitor) from EndoC-βH3 cells exposed for 30 minutes to 500 nM thapsigargin or DMSO solvent control for 30 minutes. Protein concentration was quantified using BCA protein assay kit (PIERCE), and 20 μg of each extract was separated, transferred, blocked, and probed with primary antibodies as described above using phosphorylated or total p38 primary antibodies (STAR Methods). Western blot bands were quantified using Fiji (ImageJ).

Flow cytometry analysis of human islets

Human islets from five donors were incubated for 72h in CMRL (10% FBS, 1% Glutamax, 25mM Glucose) supplemented with or without 0.4mM palmitate (stock couplings: 18.4% BSA + 8mM palmitate diluted 1:20 in culture media) with or without 2 μM Selonsertib (Gilead Sciences). A single cell solution was prepared by incubating islets in Accutase (Gibco) for 10 minutes at 37C as previously described212 and stained using DAPI (Roche) and PE-Annexin V Apoptosis Detection Kit (BioLegend) according to manufacturer’s instructions. DAPI and Annexin V staining was analyzed and quantified (Data S1) using FloJo Software (BD Sciences) for each islet donor sample sorted on a Fortessa flow cytometer (BD Sciences) using gating strategy shown in Figure S5H. These data can be found in Data S1.

QUANTIFICATION AND STATISTICAL ANALYSIS

Please refer to the figure legends for detailed information on the statistical tests used, what n represents, the exact value of n, and for how the significance was defined. Significant results have been indicated with asterisks, and the non-significant results have been indicated with ns.

Supplementary Material

1

Data S1. Data used to generate figures in this manuscript, related to Figures 15 and Figures S1-5

2
3

Table S1. Donor demographics, related to Figures 15

4

Table S2. RNA-seq analyses, related to Figures 1, 3, 4-5

5

Table S3. scRNA-seq analyses, related to Figures 2, 4-5

6

Table S4. ATAC-seq analyses, related to Figures 35

7

Table S5. Overlapping genetic variants with stress-responsive CREs, related to Figures 45

Key Resources Table

REAGENT or RESOURCE SOURCE IDENTIFIER
Antibodies
NFIL3 Proteintech Cat# 11773–1-AP
ATF4 Cell-signalling Cat# 11815S
Lamin A/C BioLegend Cat# 600002
bTubulin Cell-signalling Cat# 2128S
phospho-p38 MAPK (Thr180/Tyr182) Antibody Cell-signalling Cat# 9211S
p38 MAPK Antibody Cell-signalling Cat# 9212S
Anti-rabbit IgG, HRP-linked Antibody Cell-signalling Cat# 7074S
Anti-mouse IgG, HRP-linked Antibody Cell-signalling Cat# 7076S
Bacterial and virus strains
psPAX2 Addgene Cat# 12259
pCMV-VSV-G Addgene Cat# 8454
Biological samples
Cadaveric human pancreatic islets Integrated Islet Distribution Program (IIDP), Prodo Laboratories Table S1
Chemicals, peptides, and recombinant proteins
Prodo Media Prodo Laboratories Cat# PIM-S001GMP
Cat# PIM-ABS001GMP
Cat# PIM-G001GMP
CMRL1066 Media Gibco Cat# 11530037
Glutamax Gibco Cat# 35050061
Human recombinant Interleukin beta 1 (IL1β) R&D systems Cat# 201-LB-005/CF
Human recombinant Interferon gamma (IFNy) R&D systems Cat# 285_IF-100/CF
Thapsigargin Cayman Chemicals Cat# NC1412406
DMSO VWR Cat# 97063–136
StemPro Accutase Cell Dissociation Reagent Gibco Cat# A1110501
Trizol Life Technologies Cat# 15596018
Trypsin .05% EDTA Gibco Cat# 25300120
FBS, USDA approved origin- 500mL VWR Cat# 89510–186
BSA Sigma-Aldrich Cat# A7030
Advanced DMEM/F-12 Gibco Cat# 12634028
2-beta mercaptoethanol Gibco Cat# 21985023
nicotinamide Sigma-Aldrich Cat# 72340
Sodium selenite Sigma-Aldrich Cat# S1382
Puromycin Takara Cat# 631306
Penicillin/Streptomycin Gibco Cat# 15140163
Fibronectin Sigma-Aldrich Cat# F1141
ECM Sigma-Aldrich Cat# E1270
Sodium palmitate Sigma-Aldrich Cat# P9767
Selonsertib (GS-4997) Selleckchem/Gilead Sciences Cat# S8292
Critical commercial assays
10x snATACseq 10x Genomics Cat# PN-1000175
10x scRNAseq 10x Genomics Cat# PN-1000121
ATAC kit Active Motif Cat# 53150
ERCC Mix 1 & 2 Thermo Fisher Cat#4456740
LightShift EMSA optimization and control kit Thermo Scientific Cat# 20148X
Chemiluminescent Nucleic acid detection kit PIERCE Cat# PI89880
6% DNA Retardation gel Invitrogen Cat# EC6365BOX
NE-PER Nuclear and Cytoplasmic Extraction Kit PIERCE Cat# PI78833
BCA protein assay kit PIERCE Cat# PI-23225
PE Annexin V Apoptosis Detection Kit Biolegend Cat# 640934
MAP3K5 shRNA SIGMA TRCN0000000993
MISSION® pLKO.1-puro Non-Target shRNA Control SIGMA SHC016
RNA to CT kit Applied Biosystems Cat# 4392938
TaqMan HPRT1 Thermo Fisher Hs02800695_m1
TaqMan MAP3K5 Thermo Fisher Hs00178726_m1
LentiX concentrator Takara Cat# 631232
LentiX Takara p24 ELISA Takara Cat# 632200
Quibit RNA HS Assay Applied Biosystems Cat# Q32852
A0251 = GTCAACTCTTTAGCG
A0252 = TGATGGCCTATTGGG
A0253 = TTCCGCCTCTCTTTG
A0254 = AGTAAGTTCAGCGTA
A0255 = AAGTATCGTTTCGCA
A0256 = GGTTGCCAGATGTCA
A0257 = TGTCTTTCCTGCCAG
Biolegend Cat# 394601
Cat# 394603
Cat# 394605
Cat# 394607
Cat# 394609
Cat# 394611
Cat# 394613
4–20% Mini-PROTEAN® TGX Precast Protein Gels Biorad Cat#456–1095
Supersignal West Pico Plus
Chemiluminescence Substrate
PIERCE Cat# 34580
Deposited data
RNA-seq, scRNA-seq and ATAC-seq This paper GSE251913
Data used to generate figures This paper Data S1 and
https://doi.org/10.5281/zenodo.13730232
Scripts used for generating figures This paper https://github.com/UcarLab/ERS-vs-CYT-Islet-Stress-Responses
Experimental models: Cell lines
EndoCbH3 Univercell Biosolutions CVCL_IS72
HEK 293T ATCC Cat# CRL-3216
Experimental models: Organisms/strains
N/A N/A N/A
Oligonucleotides
rs6917676_G_FWD-/5Biosg/TAATGACTGTGTTCTTAAGAT Integrated DNA
Technologies (IDT)
N/A
rs6917676_G_REV-/5Biosg/ATCTTAAGAACACAGTCATTA Integrated DNA
Technologies (IDT)
N/A
rs6917676_T_FWD-/5Biosg/TAATGACTGTTTTCTTAAGAT Integrated DNA
Technologies (IDT)
N/A
rs6917676_T_REV-/5Biosg/ATCTTAAGAAAACAGTCATTA Integrated DNA
Technologies (IDT)
N/A
Recombinant DNA
N/A N/A N/A
Software and algorithms
SoupX Young and Behjati191 https://github.com/constantAmateur/SoupX
STAR Dobin et al.176 https://github.com/alexdobin/STAR
QoRTs Hartley and Mullikin177 https://hartleys.github.io/QoRTs/
Ensembl Howe et al.178 https://useast.ensembl.org/Homo_sapiens/Info/Index
RUVSeq Risso et al.179 https://bioconductor.org/packages/release/bioc/html/RUVSeq.html
svaseq Leek180 https://github.com/jtleek/svaseq
edgeR Robinson et al.181 https://bioconductor.org/packages/release/bioc/html/edgeR.html
DAVID Sherman et al.182 https://david.ncifcrf.gov/
KEGG Kanehisa and Goto183 https://www.genome.jp/kegg/pathway.html
Reactome Vastrik et al.184 https://reactome.org/
WikiPathways Pico et al.185 https://www.wikipathways.org/
bcl2fastq Illumina186 https://emea.support.illumina.com/sequencing/sequencing_software/bcl2fastq-conversion-software.html
Cell Ranger Count Pipeline 10X Genomics188 https://www.10xgenomics.com/support/software/cell-ranger/latest/analysis/running-pipelines/cr-gex-count
CITE-seq-Count Roelli et al.189 https://github.com/Hoohm/CITE-seq-Count
Demuxlet Kang et al.190 https://github.com/statgen/demuxlet
Seurat Hao et al.192 https://satijalab.org/seurat/
Scrublet Wolock et al.193 https://github.com/swolock/scrublet
Harmony Korsunsky et al.193 https://github.com/immunogenomics/harmony
UCell Andreatta et al.195 https://bioconductor.org/packages/release/bioc/html/UCell.html
MAST Finak et al.196 https://www.bioconductor.org/packages/release/bioc/html/MAST.html
Trimmomatic Bolger et al.197 http://www.usadellab.org/cms/?page=trimmomatic
BWA-MEM Li and Durbin198 https://github.com/lh3/bwa
SAMtools Li et al.201 https://github.com/samtools/samtools
MACS2 Zhang et al.202 https://pypi.org/project/MACS2/
IGV Thorvaldsdóttir et al.203 https://igv.org/doc/desktop/
ENCODE Blacklist Amemiya et al.204 https://github.com/Boyle-Lab/Blacklist/
DiffBind Ross-Innes et al.205 https://bioconductor.org/packages/release/bioc/html/DiffBind.html
GenomicRanges Lawrence et al.206 https://bioconductor.org/packages/release/bioc/html/GenomicRanges.html
GENCODE Harrow et al.207 https://www.gencodegenes.org/human/release_35.html
HOMER Heinz et al.123 http://homer.ucsd.edu/homer/
IRanges Lawrence et al.206 https://bioconductor.org/packages/release/bioc/html/GenomicRanges.html
HINT-ATAC Li et al.103 https://reg-gen.readthedocs.io/en/latest/hint/introduction.html
1000 Genomes Phase 3 Reference Panel The 1000 Genomes Project Consortium208 https://ldlink.nih.gov
dbSNP Sherry et al.209 https://www.ncbi.nlm.nih.gov/snp/
atSNP Zuo et al.174 http://atsnp.biostat.wisc.edu
pysam Li et al.201 https://github.com/pysam-developers/pysam
ImageJ Schneider et al.213 https://imagej.net/
Other
N/A N/A N/A

Highlights:

  1. Genome-wide comparative human islet ER/cytokine stress response maps

  2. T2D variants overlap 86 stress-responsive cis-regulatory elements (CREs)

  3. Variant-to-function links between T2D variant rs6917676, MAP3K5, islet apoptosis

  4. Risk allele increased ER stress-induced CRE accessibility, MAP3K5-mediated death

ACKNOWLEDGEMENTS

This study was made possible by generous financial support of the United States Department of Defense (DOD) under award number W81XWH-18-0401 (to M.L.S., D.U.), American Diabetes Association Pathway to Stop Diabetes Accelerator Award (1-18-ACE-015 to M.L.S.), and National Institutes of Health (NIH) under award numbers R01DK118011 (to M.L.S.), R01AG052608 (to D.U.), and U01AI165452 (to D.U.). C.N.S. was also supported by American Diabetes Association grant 11-22-JDFPM-06. Opinions, interpretations, conclusions, and recommendations are solely the responsibility of the authors and do not necessarily represent the official views of NIH, DOD, or ADA. We gratefully acknowledge contributions of JAX Single Cell Biology and Genome Technologies services and Research Cyberinfrastructure computational resources at The Jackson Laboratory for expert assistance with the work described in this publication. Human pancreatic islets were provided by NIDDK-funded Integrated Islet Distribution Program (IIDP) (RRID:SCR_014387) at City of Hope (2UC4DK098085); we are indebted to the anonymous islet organ donors and their families. Special thanks to Dr. Raphael Scharfmann at Institute Cochin for help optimizing EndoC-βH3 culture. We thank Ucar and Stitzel lab members for critical feedback throughout this study, Taneli Helenius for scientific writing assistance, and Natalia A. Mozzicato for help organizing citations. Graphical abstract was created with BioRender.com.

Footnotes

DECLARATION OF INTERESTS

The authors declare no competing interests.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

REFERENCES

  • 1.Halban PA,Polonsky KS, Bowden DW, Hawkins MA, Ling C, Mather KJ, Powers AC, Rhodes CJ, Sussel L, and Weir GC (2014). β-cell failure in type 2 diabetes: postulated mechanisms and prospects for prevention and treatment. Diabetes Care 37, 1751–1758. 10.2337/dc14-0396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Lawlor N, Khetan S, Ucar D, and Stitzel ML (2017). Genomics of Islet (Dys)function and Type 2 Diabetes. Trends Genet. 33, 244–255. 10.1016/j.tig.2017.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Ashcroft FM, and Rorsman P (2012). Diabetes mellitus and the β cell: the last ten years. Cell 148, 1160–1171. 10.1016/j.cell.2012.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Khetan S, Kursawe R, Youn A, Lawlor N, Jillette A, Marquez EJ, Ucar D, and Stitzel ML (2018). Type 2 Diabetes-Associated Genetic Variants Regulate Chromatin Accessibility in Human Islets. Diabetes 67, 2466–2477. 10.2337/db18-0393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Cornelis MC, and Hu FB (2012). Gene-environment interactions in the development of type 2 diabetes: recent progress and continuing challenges. Annu Rev Nutr 32, 245–259. 10.1146/annurev-nutr-071811-150648. [DOI] [PubMed] [Google Scholar]
  • 6.Suzuki K, Hatzikotoulas K, Southam L, Taylor HJ, Yin X, Lorenz KM, Mandla R, Huerta-Chagoya A, Rayner NW, Bocher O, et al. (2023). Multi-ancestry genome-wide study in >2.5 million individuals reveals heterogeneity in mechanistic pathways of type 2 diabetes and complications. medRxiv, 2023.03.31.23287839. 10.1101/2023.03.31.23287839. [DOI] [Google Scholar]
  • 7.Vujkovic M, Keaton JM, Lynch JA, Miller DR, Zhou J, Tcheandjieu C, Huffman JE, Assimes TL, Lorenz K, Zhu X, et al. (2020). Discovery of 318 new risk loci for type 2 diabetes and related vascular outcomes among 1.4 million participants in a multi-ancestry meta-analysis. Nat Genet 52, 680–691. 10.1038/s41588-020-0637-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Mahajan A, Taliun D, Thurner M, Robertson NR, Torres JM, Rayner NW, Payne AJ, Steinthorsdottir V, Scott RA, Grarup N, et al. (2018). Fine-mapping type 2 diabetes loci to single-variant resolution using high-density imputation and islet-specific epigenome maps. Nat. Genet 50, 1505–1513. 10.1038/s41588-018-0241-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Spracklen CN, Horikoshi M, Kim YJ, Lin K, Bragg F, Moon S, Suzuki K, Tam CHT, Tabara Y, Kwak S-H, et al. (2020). Identification of type 2 diabetes loci in 433,540 East Asian individuals. Nature 582, 240–245. 10.1038/s41586-020-2263-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Mahajan A, Spracklen CN, Zhang W, Ng MCY, Petty LE, Kitajima H, Yu GZ, Rüeger S, Speidel L, Kim YJ, et al. (2022). Multi-ancestry genetic study of type 2 diabetes highlights the power of diverse populations for discovery and translation. Nat Genet 54, 560–572. 10.1038/s41588-022-01058-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Parker SCJ, Stitzel ML, Taylor DL, Orozco JM, Erdos MR, Akiyama JA, van Bueren KL, Chines PS, Narisu N, NISC Comparative Sequencing Program, et al. (2013). Chromatin stretch enhancer states drive cell-specific gene regulation and harbor human disease risk variants. Proc. Natl. Acad. Sci. U.S.A 110, 17921–17926. 10.1073/pnas.1317023110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Pasquali L, Gaulton KJ, Rodríguez-Seguí SA, Mularoni L, Miguel-Escalada I, Akerman I, Tena JJ, Morán I, Gómez-Marín C, van de Bunt M, et al. (2014). Pancreatic islet enhancer clusters enriched in type 2 diabetes risk-associated variants. Nat. Genet 46, 136–143. 10.1038/ng.2870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Varshney A, Scott LJ, Welch RP, Erdos MR, Chines PS, Narisu N, Albanus RD, Orchard P, Wolford BN, Kursawe R, et al. (2017). Genetic regulatory signatures underlying islet gene expression and type 2 diabetes. Proc. Natl. Acad. Sci. U.S.A 10.1073/pnas.1621192114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Gaulton KJ, Ferreira T, Lee Y, Raimondo A, Mägi R, Reschen ME, Mahajan A, Locke A, Rayner NW, Robertson N, et al. (2015). Genetic fine mapping and genomic annotation defines causal mechanisms at type 2 diabetes susceptibility loci. Nat. Genet 47, 1415–1425. 10.1038/ng.3437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Khetan S, Kales S, Kursawe R, Jillette A, Ulirsch JC, Reilly SK, Ucar D, Tewhey R, and Stitzel ML (2021). Functional characterization of T2D-associated SNP effects on baseline and ER stress-responsive β cell transcriptional activation. Nat Commun 12, 5242. 10.1038/s41467-021-25514-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Roman TS, Cannon ME, Vadlamudi S, Buchkovich ML, Wolford BN, Welch RP, Morken MA, Kwon GJ, Varshney A, Kursawe R, et al. (2017). A Type 2 Diabetes-Associated Functional Regulatory Variant in a Pancreatic Islet Enhancer at the Adcy5 Locus. Diabetes 10.2337/db17-0464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kycia I, Wolford BN, Huyghe JR, Fuchsberger C, Vadlamudi S, Kursawe R, Welch RP, Albanus RD, Uyar A, Khetan S, et al. (2018). A common type 2 diabetes risk variant potentiates activity of an evolutionarily conserved islet stretch enhancer and increases C2CD4A/B expression. American Journal of Human Genetics in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Lee J-H, and Lee J (2022). Endoplasmic Reticulum (ER) Stress and Its Role in Pancreatic β-Cell Dysfunction and Senescence in Type 2 Diabetes. Int J Mol Sci 23, 4843. 10.3390/ijms23094843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Yong J, Johnson JD, Arvan P, Han J, and Kaufman RJ (2021). Therapeutic opportunities for pancreatic β-cell ER stress in diabetes mellitus. Nat Rev Endocrinol 17, 455–467. 10.1038/s41574-021-00510-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Synofzik M, Haack TB, Kopajtich R, Gorza M, Rapaport D, Greiner M, Schönfeld C, Freiberg C, Schorr S, Holl RW, et al. (2014). Absence of BiP co-chaperone DNAJC3 causes diabetes mellitus and multisystemic neurodegeneration. Am J Hum Genet 95, 689–697. 10.1016/j.ajhg.2014.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Lytrivi M, Senée V, Salpea P, Fantuzzi F, Philippi A, Abdulkarim B, Sawatani T, Marín-Cañas S, Pachera N, Degavre A, et al. (2021). DNAJC3 deficiency induces β-cell mitochondrial apoptosis and causes syndromic young-onset diabetes. Eur J Endocrinol 184, 455–468. 10.1530/EJE-20-0636. [DOI] [PubMed] [Google Scholar]
  • 22.Fugassa E, Palmero S, and Gallo G (1987). Triiodothyronine decreases the production of androgen binding protein by rat Sertoli cells. Biochem Biophys Res Commun 143, 241–247. 10.1016/0006-291x(87)90656-5. [DOI] [PubMed] [Google Scholar]
  • 23.Kulanuwat S, Tangjittipokin W, Jungtrakoon P, Chanprasert C, Sujjitjoon J, Binnima N, Yenchitsomanus P-T, and Plengvidhya N (2018). DNAJC3 mutation in Thai familial type 2 diabetes mellitus. Int J Mol Med 42, 1064–1073. 10.3892/ijmm.2018.3678. [DOI] [PubMed] [Google Scholar]
  • 24.Delépine M, Nicolino M, Barrett T, Golamaully M, Lathrop GM, and Julier C (2000). EIF2AK3, encoding translation initiation factor 2-alpha kinase 3, is mutated in patients with Wolcott-Rallison syndrome. Nat. Genet 25, 406–409. 10.1038/78085. [DOI] [PubMed] [Google Scholar]
  • 25.De Franco E, Caswell R, Johnson MB, Wakeling MN, Zung A, Dũng VC, Bích Ngọc CT, Goonetilleke R, Vivanco Jury M, El-Khateeb M, et al. (2020). De Novo Mutations in EIF2B1 Affecting eIF2 Signaling Cause Neonatal/Early-Onset Diabetes and Transient Hepatic Dysfunction. Diabetes 69, 477–483. 10.2337/db19-1029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Thameem F, Farook VS, Bogardus C, and Prochazka M (2006). Association of amino acid variants in the activating transcription factor 6 gene (ATF6) on 1q21-q23 with type 2 diabetes in Pima Indians. Diabetes 55, 839–842. 10.2337/diabetes.55.03.06.db05-1002. [DOI] [PubMed] [Google Scholar]
  • 27.Inoue H, Tanizawa Y, Wasson J, Behn P, Kalidas K, Bernal-Mizrachi E, Mueckler M, Marshall H, Donis-Keller H, Crock P, et al. (1998). A gene encoding a transmembrane protein is mutated in patients with diabetes mellitus and optic atrophy (Wolfram syndrome). Nat Genet 20, 143–148. 10.1038/2441. [DOI] [PubMed] [Google Scholar]
  • 28.Bonnycastle LL, Chines PS, Hara T, Huyghe JR, Swift AJ, Heikinheimo P, Mahadevan J, Peltonen S, Huopio H, Nuutila P, et al. (2013). Autosomal dominant diabetes arising from a Wolfram syndrome 1 mutation. Diabetes 62, 3943–3950. 10.2337/db13-0571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ehses JA, Böni-Schnetzler M, Faulenbach M, and Donath MY (2008). Macrophages, cytokines and beta-cell death in Type 2 diabetes. Biochem Soc Trans 36, 340–342. 10.1042/BST0360340. [DOI] [PubMed] [Google Scholar]
  • 30.Novotny GW, Lundh M, Backe MB, Christensen DP, Hansen JB, Dahllöf MS, Pallesen EMH, and Mandrup-Poulsen T (2012). Transcriptional and translational regulation of cytokine signaling in inflammatory β-cell dysfunction and apoptosis. Arch Biochem Biophys 528, 171–184. 10.1016/j.abb.2012.09.014. [DOI] [PubMed] [Google Scholar]
  • 31.Nordmann TM, Dror E, Schulze F, Traub S, Berishvili E, Barbieux C, Böni-Schnetzler M, and Donath MY (2017). The Role of Inflammation in β-cell Dedifferentiation. Sci Rep 7, 6285. 10.1038/s41598-017-06731-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Cieślak M, Wojtczak A, and Cieślak M (2015). Role of pro-inflammatory cytokines of pancreatic islets and prospects of elaboration of new methods for the diabetes treatment. Acta Biochim Pol 62, 15–21. 10.18388/abp.2014_853. [DOI] [PubMed] [Google Scholar]
  • 33.Back SH, and Kaufman RJ (2012). Endoplasmic reticulum stress and type 2 diabetes. Annu. Rev. Biochem 81, 767–793. 10.1146/annurev-biochem-072909-095555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Calle MC, and Fernandez ML (2012). Inflammation and type 2 diabetes. Diabetes Metab 38, 183–191. 10.1016/j.diabet.2011.11.006. [DOI] [PubMed] [Google Scholar]
  • 35.Sharma RB, O’Donnell AC, Stamateris RE, Ha B, McCloskey KM, Reynolds PR, Arvan P, and Alonso LC (2015). Insulin demand regulates β cell number via the unfolded protein response. J. Clin. Invest 125, 3831–3846. 10.1172/JCI79264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Snyder JT, Darko C, Sharma RB, and Alonso LC (2021). Endoplasmic Reticulum Stress Induced Proliferation Remains Intact in Aging Mouse β-Cells. Front Endocrinol (Lausanne) 12, 734079. 10.3389/fendo.2021.734079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Brownrigg GP, Xia YH, Chu CMJ, Wang S, Chao C, Zhang JA, Skovsø S, Panzhinskiy E, Hu X, Johnson JD, et al. (2023). Sex differences in islet stress responses support female β cell resilience. Mol Metab 69, 101678. 10.1016/j.molmet.2023.101678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hohmeier HE, Zhang L, Taylor B, Stephens S, Lu D, McNamara P, Laffitte B, and Newgard CB (2020). Identification of a small molecule that stimulates human β-cell proliferation and insulin secretion, and protects against cytotoxic stress in rat insulinoma cells. PLoS One 15, e0224344. 10.1371/journal.pone.0224344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lopes M, Kutlu B, Miani M, Bang-Berthelsen CH, Størling J, Pociot F, Goodman N, Hood L, Welsh N, Bontempi G, et al. (2014). Temporal profiling of cytokine-induced genes in pancreatic β-cells by meta-analysis and network inference. Genomics 103, 264–275. 10.1016/j.ygeno.2013.12.007. [DOI] [PubMed] [Google Scholar]
  • 40.Dos Santos RS, Guzman-Llorens D, Perez-Serna AA, Nadal A, and Marroqui L (2023). Deucravacitinib, a tyrosine kinase 2 pseudokinase inhibitor, protects human EndoC-βH1 β-cells against proinflammatory insults. Front Immunol 14, 1263926. 10.3389/fimmu.2023.1263926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Giusti L, Tesi M, Ciregia F, Marselli L, Zallocco L, Suleiman M, De Luca C, Del Guerra S, Zuccarini M, Trerotola M, et al. (2022). The Protective Action of Metformin against Pro-Inflammatory Cytokine-Induced Human Islet Cell Damage and the Mechanisms Involved. Cells 11, 2465. 10.3390/cells11152465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Nagalakshmi U, Wang Z, Waern K, Shou C, Raha D, Gerstein M, and Snyder M (2008). The transcriptional landscape of the yeast genome defined by RNA sequencing. Science 320, 1344–1349. 10.1126/science.1158441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Fonseca SG, Gromada J, and Urano F (2011). Endoplasmic reticulum stress and pancreatic β-cell death. Trends Endocrinol Metab 22, 266–274. 10.1016/j.tem.2011.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Papa FR (2012). Endoplasmic reticulum stress, pancreatic β-cell degeneration, and diabetes. Cold Spring Harb Perspect Med 2, a007666. 10.1101/cshperspect.a007666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Kim I, Xu W, and Reed JC (2008). Cell death and endoplasmic reticulum stress: disease relevance and therapeutic opportunities. Nat Rev Drug Discov 7, 1013–1030. 10.1038/nrd2755. [DOI] [PubMed] [Google Scholar]
  • 46.Meyerovich K, Ortis F, Allagnat F, and Cardozo AK (2016). Endoplasmic reticulum stress and the unfolded protein response in pancreatic islet inflammation. J Mol Endocrinol 57, R1–R17. 10.1530/JME-15-0306. [DOI] [PubMed] [Google Scholar]
  • 47.Ho DV, and Chan JY (2015). Induction of Herpud1 expression by ER stress is regulated by Nrf1. FEBS Lett 589, 615–620. 10.1016/j.febslet.2015.01.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Harding HP, Zeng H, Zhang Y, Jungries R, Chung P, Plesken H, Sabatini DD, and Ron D (2001). Diabetes mellitus and exocrine pancreatic dysfunction in perk−/− mice reveals a role for translational control in secretory cell survival. Mol Cell 7, 1153–1163. 10.1016/s1097-2765(01)00264-7. [DOI] [PubMed] [Google Scholar]
  • 49.Wong N, Morahan G, Stathopoulos M, Proietto J, and Andrikopoulos S (2013). A novel mechanism regulating insulin secretion involving Herpud1 in mice. Diabetologia 56, 1569–1576. 10.1007/s00125-013-2908-y. [DOI] [PubMed] [Google Scholar]
  • 50.Mbikay M, Sirois F, Mayne J, Wang G-S, Chen A, Dewpura T, Prat A, Seidah NG, Chretien M, and Scott FW (2010). PCSK9-deficient mice exhibit impaired glucose tolerance and pancreatic islet abnormalities. FEBS Lett 584, 701–706. 10.1016/j.febslet.2009.12.018. [DOI] [PubMed] [Google Scholar]
  • 51.Marku A, Da Dalt L, Galli A, Dule N, Corsetto P, Rizzo AM, Moregola A, Uboldi P, Bonacina F, Marciani P, et al. (2022). Pancreatic PCSK9 controls the organization of the β-cell secretory pathway via LDLR-cholesterol axis. Metabolism 136, 155291. 10.1016/j.metabol.2022.155291. [DOI] [PubMed] [Google Scholar]
  • 52.Marselli L, Bosi E, De Luca C, Del Guerra S, Tesi M, Suleiman M, and Marchetti P (2021). Arginase 2 and Polyamines in Human Pancreatic Beta Cells: Possible Role in the Pathogenesis of Type 2 Diabetes. Int J Mol Sci 22, 12099. 10.3390/ijms222212099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Solimena M, Schulte AM, Marselli L, Ehehalt F, Richter D, Kleeberg M, Mziaut H, Knoch K-P, Parnis J, Bugliani M, et al. (2018). Systems biology of the IMIDIA biobank from organ donors and pancreatectomised patients defines a novel transcriptomic signature of islets from individuals with type 2 diabetes. Diabetologia 61, 641–657. 10.1007/s00125-017-4500-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Ramos-Rodríguez M, Raurell-Vila H, Colli ML, Alvelos MI, Subirana-Granés M, Juan-Mateu J, Norris R, Turatsinze J-V, Nakayasu ES, Webb-Robertson B-JM, et al. (2019). The impact of proinflammatory cytokines on the β-cell regulatory landscape provides insights into the genetics of type 1 diabetes. Nat. Genet 51, 1588–1595. 10.1038/s41588-019-0524-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Eizirik DL, Sammeth M, Bouckenooghe T, Bottu G, Sisino G, Igoillo-Esteve M, Ortis F, Santin I, Colli ML, Barthson J, et al. (2012). The human pancreatic islet transcriptome: expression of candidate genes for type 1 diabetes and the impact of pro-inflammatory cytokines. PLoS Genet. 8, e1002552. 10.1371/journal.pgen.1002552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Benaglio P, Zhu H, Okino M-L, Yan J, Elgamal R, Nariai N, Beebe E, Korgaonkar K, Qiu Y, Donovan MKR, et al. (2022). Type 1 diabetes risk genes mediate pancreatic beta cell survival in response to proinflammatory cytokines. Cell Genom 2, 100214. 10.1016/j.xgen.2022.100214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Collier JJ, Sparer TE, Karlstad MD, and Burke SJ (2017). Pancreatic islet inflammation: an emerging role for chemokines. J Mol Endocrinol 59, R33–R46. 10.1530/JME-17-0042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Imai Y, Dobrian AD, Morris MA, and Nadler JL (2013). Islet inflammation: a unifying target for diabetes treatment? Trends Endocrinol Metab 24, 351–360. 10.1016/j.tem.2013.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Esser N, Legrand-Poels S, Piette J, Scheen AJ, and Paquot N (2014). Inflammation as a link between obesity, metabolic syndrome and type 2 diabetes. Diabetes Res Clin Pract 105, 141–150. 10.1016/j.diabres.2014.04.006. [DOI] [PubMed] [Google Scholar]
  • 60.Donath MY, Størling J, Berchtold LA, Billestrup N, and Mandrup-Poulsen T (2008). Cytokines and beta-cell biology: from concept to clinical translation. Endocr Rev 29, 334–350. 10.1210/er.2007-0033. [DOI] [PubMed] [Google Scholar]
  • 61.Cosentino C, and Regazzi R (2021). Crosstalk between Macrophages and Pancreatic β-Cells in Islet Development, Homeostasis and Disease. Int J Mol Sci 22, 1765. 10.3390/ijms22041765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Cnop M, Welsh N, Jonas J-C, Jörns A, Lenzen S, and Eizirik DL (2005). Mechanisms of pancreatic beta-cell death in type 1 and type 2 diabetes: many differences, few similarities. Diabetes 54 Suppl 2, S97–107. 10.2337/diabetes.54.suppl_2.s97. [DOI] [PubMed] [Google Scholar]
  • 63.Eizirik DL, Pasquali L, and Cnop M (2020). Pancreatic β-cells in type 1 and type 2 diabetes mellitus: different pathways to failure. Nat Rev Endocrinol 16, 349–362. 10.1038/s41574-020-0355-7. [DOI] [PubMed] [Google Scholar]
  • 64.Knebel UE, Peleg S, Dai C, Cohen-Fultheim R, Jonsson S, Poznyak K, Israeli M, Zamashanski L, Glaser B, Levanon EY, et al. (2024). Disrupted RNA editing in beta cells mimics early-stage type 1 diabetes. Cell Metab 36, 48–61.e6. 10.1016/j.cmet.2023.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Bensellam M, Laybutt DR, and Jonas J-C (2021). Emerging Roles of Metallothioneins in Beta Cell Pathophysiology: Beyond and Above Metal Homeostasis and Antioxidant Response. Biology (Basel) 10, 176. 10.3390/biology10030176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Vasileva A, Hopkins KM, Wang X, Weisbach MM, Friedman RA, Wolgemuth DJ, and Lieberman HB (2013). The DNA damage checkpoint protein RAD9A is essential for male meiosis in the mouse. J Cell Sci 126, 3927–3938. 10.1242/jcs.126763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Huerta Guevara AP, McGowan SJ, Kazantzis M, Stallons TR, Sano T, Mulder NL, Jurdzinski A, van Dijk TH, Eggen BJL, Jonker JW, et al. (2021). Increased insulin sensitivity and diminished pancreatic beta-cell function in DNA repair deficient Ercc1d/-mice. Metabolism 117, 154711. 10.1016/j.metabol.2021.154711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Zhu Y, Liu Q, Zhou Z, and Ikeda Y (2017). PDX1, Neurogenin-3, and MAFA: critical transcription regulators for beta cell development and regeneration. Stem Cell Res Ther 8, 240. 10.1186/s13287-017-0694-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Hodson DJ, Mitchell RK, Marselli L, Pullen TJ, Gimeno Brias S, Semplici F, Everett KL, Cooper DMF, Bugliani M, Marchetti P, et al. (2014). ADCY5 couples glucose to insulin secretion in human islets. Diabetes 63, 3009–3021. 10.2337/db13-1607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Hall E, Dayeh T, Kirkpatrick CL, Wollheim CB, Dekker Nitert M, and Ling C (2013). DNA methylation of the glucagon-like peptide 1 receptor (GLP1R) in human pancreatic islets. BMC Med Genet 14, 76. 10.1186/1471-2350-14-76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Clemmons DR (2018). Role of IGF-binding proteins in regulating IGF responses to changes in metabolism. J Mol Endocrinol 61, T139–T169. 10.1530/JME-18-0016. [DOI] [PubMed] [Google Scholar]
  • 72.Lu M, and Li C (2018). Nutrient sensing in pancreatic islets: lessons from congenital hyperinsulinism and monogenic diabetes. Ann N Y Acad Sci 1411, 65–82. 10.1111/nyas.13448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Llacua LA, Faas MM, and de Vos P (2018). Extracellular matrix molecules and their potential contribution to the function of transplanted pancreatic islets. Diabetologia 61, 1261–1272. 10.1007/s00125-017-4524-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Wieland FC, Van Blitterswijk CA, Van Apeldoorn A, and LaPointe VLS (2021). The functional importance of the cellular and extracellular composition of the islets of Langerhans. Journal of Immunology and Regenerative Medicine 13, 100048. 10.1016/j.regen.2021.100048. [DOI] [Google Scholar]
  • 75.Zhu X, Oguh A, Gingerich MA, Soleimanpour SA, Stoffers DA, and Gannon M (2021). Cell Cycle Regulation of the Pdx1 Transcription Factor in Developing Pancreas and Insulin-Producing β-Cells. Diabetes 70, 903–916. 10.2337/db20-0599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Keller MP, Choi Y, Wang P, Davis DB, Rabaglia ME, Oler AT, Stapleton DS, Argmann C, Schueler KL, Edwards S, et al. (2008). A gene expression network model of type 2 diabetes links cell cycle regulation in islets with diabetes susceptibility. Genome Res 18, 706–716. 10.1101/gr.074914.107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.El-Badawy A, and El-Badri N (2016). The cell cycle as a brake for β-cell regeneration from embryonic stem cells. Stem Cell Res Ther 7, 9. 10.1186/s13287-015-0274-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Cozar-Castellano I, Fiaschi-Taesch N, Bigatel TA, Takane KK, Garcia-Ocaña A, Vasavada R, and Stewart AF (2006). Molecular control of cell cycle progression in the pancreatic beta-cell. Endocr Rev 27, 356–370. 10.1210/er.2006-0004. [DOI] [PubMed] [Google Scholar]
  • 79.Zhou Y, Zhou J, Sun B, Xu W, Zhong M, Li Y, He C, Chen Y, Wang X, Jones PM, et al. (2020). Vitamin A deficiency causes islet dysfunction by inducing islet stellate cell activation via cellular retinol binding protein 1. Int J Biol Sci 16, 947–956. 10.7150/ijbs.37861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Zhou Y, Wang H, Zhou J, Qiu S, Cai T, Li H, Shen Z, Hu Y, Ding B, Luo M, et al. (2021). Vitamin A and Its Multi-Effects on Pancreas: Recent Advances and Prospects. Front Endocrinol (Lausanne) 12, 620941. 10.3389/fendo.2021.620941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Xiafukaiti G, Maimaiti S, Ogata K, Kuno A, Kudo T, Shawki HH, Oishi H, and Takahashi S (2019). MafB Is Important for Pancreatic β-Cell Maintenance under a MafA-Deficient Condition. Mol Cell Biol 39, e00080-19. 10.1128/MCB.00080-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Conrad E, Dai C, Spaeth J, Guo M, Cyphert HA, Scoville D, Carroll J, Yu W-M, Goodrich LV, Harlan DM, et al. (2016). The MAFB transcription factor impacts islet α-cell function in rodents and represents a unique signature of primate islet β-cells. Am J Physiol Endocrinol Metab 310, E91–E102. 10.1152/ajpendo.00285.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Artner I, Blanchi B, Raum JC, Guo M, Kaneko T, Cordes S, Sieweke M, and Stein R (2007). MafB is required for islet beta cell maturation. Proc. Natl. Acad. Sci. U.S.A 104, 3853–3858. 10.1073/pnas.0700013104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Santo-Domingo J, Galindo AN, Cominetti O, De Marchi U, Cutillas P, Dayon L, and Wiederkehr A (2019). Glucose-dependent phosphorylation signaling pathways and crosstalk to mitochondrial respiration in insulin secreting cells. Cell Commun Signal 17, 14. 10.1186/s12964-019-0326-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Chabosseau P, Rutter GA, and Millership SJ (2021). Importance of Both Imprinted Genes and Functional Heterogeneity in Pancreatic Beta Cells: Is There a Link? Int J Mol Sci 22, 1000. 10.3390/ijms22031000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Dos Santos T, Galipeau M, Schukarucha Gomes A, Greenberg M, Larsen M, Lee D, Maghera J, Mulchandani CM, Patton M, Perera I, et al. (2022). Islet Biology During COVID-19: Progress and Perspectives. Can J Diabetes 46, 419–427. 10.1016/j.jcjd.2021.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Lundh M, Bugliani M, Dahlby T, Chou DH-C, Wagner B, Ghiasi SM, De Tata V, Chen Z, Lund MN, Davies MJ, et al. (2017). The immunoproteasome is induced by cytokines and regulates apoptosis in human islets. J Endocrinol 233, 369–379. 10.1530/JOE-17-0110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Buenrostro JD, Giresi PG, Zaba LC, Chang HY, and Greenleaf WJ (2013). Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat Meth 10, 1213–1218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Dao LTM, Galindo-Albarrán AO, Castro-Mondragon JA, Andrieu-Soler C, Medina-Rivera A, Souaid C, Charbonnier G, Griffon A, Vanhille L, Stephen T, et al. (2017). Genome-wide characterization of mammalian promoters with distal enhancer functions. Nat Genet 49, 1073–1081. 10.1038/ng.3884. [DOI] [PubMed] [Google Scholar]
  • 90.Grosveld F, van Staalduinen J, and Stadhouders R (2021). Transcriptional Regulation by (Super)Enhancers: From Discovery to Mechanisms. Annu Rev Genomics Hum Genet 22, 127–146. 10.1146/annurev-genom-122220-093818. [DOI] [PubMed] [Google Scholar]
  • 91.Ron D, and Walter P (2007). Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol 8, 519–529. 10.1038/nrm2199. [DOI] [PubMed] [Google Scholar]
  • 92.Ghosh R, Colon-Negron K, and Papa FR (2019). Endoplasmic reticulum stress, degeneration of pancreatic islet β-cells, and therapeutic modulation of the unfolded protein response in diabetes. Mol Metab 27S, S60–S68. 10.1016/j.molmet.2019.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Ichikawa M, Konoshita T, Makino Y, Suzuki J, Ishizuka T, and Nakamura H (2020). An association study of C9orf3, a novel component of the renin-angiotensin system, and hypertension in diabetes. Sci Rep 10, 16111. 10.1038/s41598-020-73094-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Sever D, Hershko-Moshe A, Srivastava R, Eldor R, Hibsher D, Keren-Shaul H, Amit I, Bertuzzi F, Krogvold L, Dahl-Jørgensen K, et al. (2021). NF-κB activity during pancreas development regulates adult β-cell mass by modulating neonatal β-cell proliferation and apoptosis. Cell Death Discov 7, 2. 10.1038/s41420-020-00386-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Meyerovich K, Ortis F, and Cardozo AK (2018). The non-canonical NF-κB pathway and its contribution to β-cell failure in diabetes. J Mol Endocrinol 61, F1–F6. 10.1530/JME-16-0183. [DOI] [PubMed] [Google Scholar]
  • 96.Liu T, Zhang L, Joo D, and Sun S-C (2017). NF-κB signaling in inflammation. Signal Transduct Target Ther 2, 17023-. 10.1038/sigtrans.2017.23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Rottner AK, Ye Y, Navarro-Guerrero E, Rajesh V, Pollner A, Bevacqua RJ, Yang J, Spigelman AF, Baronio R, Bautista A, et al. (2023). A genome-wide CRISPR screen identifies CALCOCO2 as a regulator of beta cell function influencing type 2 diabetes risk. Nat Genet 55, 54–65. 10.1038/s41588-022-01261-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Pearson GL, Gingerich MA, Walker EM, Biden TJ, and Soleimanpour SA (2021). A Selective Look at Autophagy in Pancreatic β-Cells. Diabetes. 10.2337/dbi20-0014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Lenghel A, Gheorghita AM, Vacaru AM, and Vacaru A-M (2020). What Is the Sweetest UPR Flavor for the β-cell? That Is the Question. Front Endocrinol (Lausanne) 11, 614123. 10.3389/fendo.2020.614123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Rabhi N, Salas E, Froguel P, and Annicotte J-S (2014). Role of the unfolded protein response in β cell compensation and failure during diabetes. J Diabetes Res 2014, 795171. 10.1155/2014/795171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Keniry M, Dearth RK, Persans M, and Parsons R (2014). New Frontiers for the NFIL3 bZIP Transcription Factor in Cancer, Metabolism and Beyond. Discoveries (Craiova) 2, e15. 10.15190/d.2014.7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Byles V, Cormerais Y, Kalafut K, Barrera V, Hughes Hallett JE, Sui SH, Asara JM, Adams CM, Hoxhaj G, Ben-Sahra I, et al. (2021). Hepatic mTORC1 signaling activates ATF4 as part of its metabolic response to feeding and insulin. Mol Metab 53, 101309. 10.1016/j.molmet.2021.101309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Li Z, Schulz MH, Look T, Begemann M, Zenke M, and Costa IG (2019). Identification of transcription factor binding sites using ATAC-seq. Genome Biol 20, 45. 10.1186/s13059-019-1642-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Gomi H, Mori K, Itohara S, and Izumi T (2007). Rab27b is expressed in a wide range of exocytic cells and involved in the delivery of secretory granules near the plasma membrane. Mol Biol Cell 18, 4377–4386. 10.1091/mbc.e07-05-0409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Izumi T (2021). In vivo Roles of Rab27 and Its Effectors in Exocytosis. Cell Struct Funct 46, 79–94. 10.1247/csf.21043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Izumi T (2007). Physiological roles of Rab27 effectors in regulated exocytosis. Endocr J 54, 649–657. 10.1507/endocrj.kr-78. [DOI] [PubMed] [Google Scholar]
  • 107.Zhou Y, Waanders LF, Holmseth S, Guo C, Berger UV, Li Y, Lehre A-C, Lehre KP, and Danbolt NC (2014). Proteome analysis and conditional deletion of the EAAT2 glutamate transporter provide evidence against a role of EAAT2 in pancreatic insulin secretion in mice. J Biol Chem 289, 1329–1344. 10.1074/jbc.M113.529065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Rorsman P, and Ashcroft FM (2018). Pancreatic β-Cell Electrical Activity and Insulin Secretion: Of Mice and Men. Physiol Rev 98, 117–214. 10.1152/physrev.00008.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Kulkarni RN, Holzenberger M, Shih DQ, Ozcan U, Stoffel M, Magnuson MA, and Kahn CR (2002). beta-cell-specific deletion of the Igf1 receptor leads to hyperinsulinemia and glucose intolerance but does not alter beta-cell mass. Nat Genet 31, 111–115. 10.1038/ng872. [DOI] [PubMed] [Google Scholar]
  • 110.Xuan S, Kitamura T, Nakae J, Politi K, Kido Y, Fisher PE, Morroni M, Cinti S, White MF, Herrera PL, et al. (2002). Defective insulin secretion in pancreatic beta cells lacking type 1 IGF receptor. J Clin Invest 110, 1011–1019. 10.1172/JCI15276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Wei X, Ma X, Lu R, Bai G, Zhang J, Deng R, Gu N, Feng N, and Guo X (2014). Genetic variants in PCSK1 gene are associated with the risk of coronary artery disease in type 2 diabetes in a Chinese Han population: a case control study. PLoS One 9, e87168. 10.1371/journal.pone.0087168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Barthelson K, Newman M, and Lardelli M (2020). Sorting Out the Role of the Sortilin-Related Receptor 1 in Alzheimer’s Disease. J Alzheimers Dis Rep 4, 123–140. 10.3233/ADR-200177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Lorenzo PI, Juárez-Vicente F, Cobo-Vuilleumier N, García-Domínguez M, and Gauthier BR (2017). The Diabetes-Linked Transcription Factor PAX4: From Gene to Functional Consequences. Genes (Basel) 8, 101. 10.3390/genes8030101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Ko J, Fonseca VA, and Wu H (2023). Pax4 in Health and Diabetes. Int J Mol Sci 24, 8283. 10.3390/ijms24098283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Ebrahim N, Shakirova K, and Dashinimaev E (2022). PDX1 is the cornerstone of pancreatic β-cell functions and identity. Front Mol Biosci 9, 1091757. 10.3389/fmolb.2022.1091757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Nishimura W, Iwasa H, and Tumurkhuu M (2022). Role of the Transcription Factor MAFA in the Maintenance of Pancreatic β-Cells. Int J Mol Sci 23, 4478. 10.3390/ijms23094478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Chiou J, Geusz RJ, Okino M-L, Han JY, Miller M, Melton R, Beebe E, Benaglio P, Huang S, Korgaonkar K, et al. (2021). Interpreting type 1 diabetes risk with genetics and single-cell epigenomics. Nature 594, 398–402. 10.1038/s41586-021-03552-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Chen J, Spracklen CN, Marenne G, Varshney A, Corbin LJ, Luan J, Willems SM, Wu Y, Zhang X, Horikoshi M, et al. (2021). The trans-ancestral genomic architecture of glycemic traits. Nat Genet 53, 840–860. 10.1038/s41588-021-00852-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Huyghe JR, Jackson AU, Fogarty MP, Buchkovich ML, Stančáková A, Stringham HM, Sim X, Yang L, Fuchsberger C, Cederberg H, et al. (2013). Exome array analysis identifies new loci and low-frequency variants influencing insulin processing and secretion. Nat. Genet 45, 197–201. 10.1038/ng.2507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Dupuis J, Langenberg C, Prokopenko I, Saxena R, Soranzo N, Jackson AU, Wheeler E, Glazer NL, Bouatia-Naji N, Gloyn AL, et al. (2010). New genetic loci implicated in fasting glucose homeostasis and their impact on type 2 diabetes risk. Nat. Genet 42, 105–116. 10.1038/ng.520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Strawbridge RJ, Dupuis J, Prokopenko I, Barker A, Ahlqvist E, Rybin D, Petrie JR, Travers ME, Bouatia-Naji N, Dimas AS, et al. (2011). Genome-wide association identifies nine common variants associated with fasting proinsulin levels and provides new insights into the pathophysiology of type 2 diabetes. Diabetes 60, 2624–2634. 10.2337/db11-0415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Robertson CC, Inshaw JRJ, Onengut-Gumuscu S, Chen W-M, Santa Cruz DF, Yang H, Cutler AJ, Crouch DJM, Farber E, Bridges SL, et al. (2021). Fine-mapping, trans-ancestral and genomic analyses identify causal variants, cells, genes and drug targets for type 1 diabetes. Nat Genet 53, 962–971. 10.1038/s41588-021-00880-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Heinz S, Benner C, Spann N, Bertolino E, Lin YC, Laslo P, Cheng JX, Murre C, Singh H, and Glass CK (2010). Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589. 10.1016/j.molcel.2010.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Gutierrez-Aguilar R, Kim D-H, Casimir M, Dai X-Q, Pfluger PT, Park J, Haller A, Donelan E, Park J, D’Alessio D, et al. (2014). The role of the transcription factor ETV5 in insulin exocytosis. Diabetologia 57, 383–391. 10.1007/s00125-013-3096-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Ofori JK, Karagiannopoulos A, Nagao M, Westholm E, Ramadan S, Wendt A, Esguerra JLS, and Eliasson L (2022). Human Islet MicroRNA-200c Is Elevated in Type 2 Diabetes and Targets the Transcription Factor ETV5 to Reduce Insulin Secretion. Diabetes 71, 275–284. 10.2337/db21-0077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Suriben R, Kaihara KA, Paolino M, Reichelt M, Kummerfeld SK, Modrusan Z, Dugger DL, Newton K, Sagolla M, Webster JD, et al. (2015). β-Cell Insulin Secretion Requires the Ubiquitin Ligase COP1. Cell 163, 1457–1467. 10.1016/j.cell.2015.10.076. [DOI] [PubMed] [Google Scholar]
  • 127.Vujkovic M, Keaton JM, Lynch JA, Miller DR, Zhou J, Tcheandjieu C, Huffman JE, Assimes TL, Lorenz K, Zhu X, et al. (2020). Discovery of 318 new risk loci for type 2 diabetes and related vascular outcomes among 1.4 million participants in a multi-ancestry meta-analysis. Nat Genet 52, 680–691. 10.1038/s41588-020-0637-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Rupérez AI, Gil A, and Aguilera CM (2014). Genetics of Oxidative Stress in Obesity. Int J Mol Sci 15, 3118–3144. 10.3390/ijms15023118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Swentek L, Chung D, and Ichii H (2021). Antioxidant Therapy in Pancreatitis. Antioxidants (Basel) 10, 657. 10.3390/antiox10050657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Jarrin Lopez A, Lau H, Li S, and Ichii H (2020). Potential Benefits of Nrf2/Keap1 Targeting in Pancreatic Islet Cell Transplantation. Antioxidants (Basel) 9, 321. 10.3390/antiox9040321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Cheng L, Tachibana K, Iwasaki H, Kameyama A, Zhang Y, Kubota T, Hiruma T, Tachibana K, Kudo T, Guo J-M, et al. (2004). Characterization of a novel human UDP-GalNAc transferase, pp-GalNAc-T15. FEBS Lett 566, 17–24. 10.1016/j.febslet.2004.03.108. [DOI] [PubMed] [Google Scholar]
  • 132.Song K-H, Park MS, Nandu TS, Gadad S, Kim S-C, and Kim M-Y (2016). GALNT14 promotes lung-specific breast cancer metastasis by modulating self-renewal and interaction with the lung microenvironment. Nat Commun 7, 13796. 10.1038/ncomms13796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Miguel-Escalada I, Bonàs-Guarch S, Cebola I, Ponsa-Cobas J, Mendieta-Esteban J, Atla G, Javierre BM, Rolando DMY, Farabella I, Morgan CC, et al. (2019). Human pancreatic islet three-dimensional chromatin architecture provides insights into the genetics of type 2 diabetes. Nat Genet 51, 1137–1148. 10.1038/s41588-019-0457-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Hellman LM, and Fried MG (2007). Electrophoretic mobility shift assay (EMSA) for detecting protein-nucleic acid interactions. Nat Protoc 2, 1849–1861. 10.1038/nprot.2007.249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Pepin E, Higa A, Schuster-Klein C, Bernard C, Sulpice T, Guardiola B, Chevet E, and Alquier T (2014). Deletion of apoptosis signal-regulating kinase 1 (ASK1) protects pancreatic beta-cells from stress-induced death but not from glucose homeostasis alterations under pro-inflammatory conditions. PLoS One 9, e112714. 10.1371/journal.pone.0112714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Katagiri K, Matsuzawa A, and Ichijo H (2010). Chapter 16 - Regulation of Apoptosis Signal-Regulating Kinase 1 in Redox Signaling. In Methods in Enzymology Thiol Redox Transitions in Cell Signaling, Part B: Cellular Localization and Signaling., Cadenas E and Packer L, eds. (Academic Press; ), pp. 277–288. 10.1016/S0076-6879(10)74016-7. [DOI] [PubMed] [Google Scholar]
  • 137.Das D, Paul A, Lahiri A, Adak M, Maity SK, Sarkar A, Paul S, and Chakrabarti P (2021). Proteasome dysfunction under compromised redox metabolism dictates liver injury in NASH through ASK1/PPARγ binodal complementary modules. Redox Biol 45, 102043. 10.1016/j.redox.2021.102043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Tobiume K, Matsuzawa A, Takahashi T, Nishitoh H, Morita K, Takeda K, Minowa O, Miyazono K, Noda T, and Ichijo H (2001). ASK1 is required for sustained activations of JNK/p38 MAP kinases and apoptosis. EMBO Rep 2, 222–228. 10.1093/embo-reports/kve046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Yamaguchi K, Takeda K, Kadowaki H, Ueda I, Namba Y, Ouchi Y, Nishitoh H, and Ichijo H (2013). Involvement of ASK1-p38 pathway in the pathogenesis of diabetes triggered by pancreatic ß cell exhaustion. Biochim Biophys Acta 1830, 3656–3663. 10.1016/j.bbagen.2013.01.029. [DOI] [PubMed] [Google Scholar]
  • 140.Obsilova V, Honzejkova K, and Obsil T (2021). Structural Insights Support Targeting ASK1 Kinase for Therapeutic Interventions. International Journal of Molecular Sciences 22, 13395. 10.3390/ijms222413395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Boucherat O, Provencher S, and Bonnet S (2018). Therapeutic Value of ASK1 Inhibition in Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 197, 284–286. 10.1164/rccm.201708-1767ED. [DOI] [PubMed] [Google Scholar]
  • 142.Weiss A, Boehm M, Egemnazarov B, Grimminger F, Savai Pullamsetti S, Kwapiszewska G, and Schermuly RT (2021). Kinases as potential targets for treatment of pulmonary hypertension and right ventricular dysfunction. British Journal of Pharmacology 178, 31–53. 10.1111/bph.14919. [DOI] [PubMed] [Google Scholar]
  • 143.Liles JT, Corkey BK, Notte GT, Budas GR, Lansdon EB, Hinojosa-Kirschenbaum F, Badal SS, Lee M, Schultz BE, Wise S, et al. (2018). ASK1 contributes to fibrosis and dysfunction in models of kidney disease. J Clin Invest 128, 4485–4500. 10.1172/JCI99768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Tesch GH, Ma FY, Han Y, Liles JT, Breckenridge DG, and Nikolic-Paterson DJ (2015). ASK1 Inhibitor Halts Progression of Diabetic Nephropathy in Nos3- Deficient Mice. Diabetes 64, 3903–3913. 10.2337/db15-0384. [DOI] [PubMed] [Google Scholar]
  • 145.Gilead Sciences (2022). MOSAIC - A Phase 2b, Randomized, Double-Blind, Placebo-Controlled, Parallel Group, Multicenter Study Evaluating the Efficacy and Safety of Selonsertib in Subjects With Moderate to Advanced Diabetic Kidney Disease (clinicaltrials.gov). [Google Scholar]
  • 146.Wang J, Xiang H, Lu Y, Wu T, and Ji G (2021). New progress in drugs treatment of diabetic kidney disease. Biomedicine & Pharmacotherapy 141, 111918. 10.1016/j.biopha.2021.111918. [DOI] [PubMed] [Google Scholar]
  • 147.Chertow GM, Pergola PE, Chen F, Kirby BJ, Sundy JS, Patel UD, and GS-US-223–1015 Investigators (2019). Effects of Selonsertib in Patients with Diabetic Kidney Disease. J Am Soc Nephrol 30, 1980–1990. 10.1681/ASN.2018121231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Oda Y, Nishi H, and Nangaku M (2023). Role of Inflammation in Progression of Chronic Kidney Disease in Type 2 Diabetes Mellitus: Clinical Implications. Seminars in Nephrology 43. 10.1016/j.semnephrol.2023.151431. [DOI] [PubMed] [Google Scholar]
  • 149.Leehey D, and Zhang X (2023). Inflammation in Diabetic Kidney Disease: Focus on New Therapeutic Considerations. Medical Research Archives 11. 10.18103/mra.v11i2.3614. [DOI] [Google Scholar]
  • 150.Rayego-Mateos S, Morgado-Pascual JL, Opazo-Ríos L, Guerrero-Hue M, García-Caballero C, Vázquez-Carballo C, Mas S, Sanz AB, Herencia C, Mezzano S, et al. (2020). Pathogenic Pathways and Therapeutic Approaches Targeting Inflammation in Diabetic Nephropathy. Int J Mol Sci 21, 3798. 10.3390/ijms21113798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Ogier JM, Nayagam BA, and Lockhart PJ (2020). ASK1 inhibition: a therapeutic strategy with multi-system benefits. J Mol Med (Berl) 98, 335–348. 10.1007/s00109-020-01878-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Liu M, Weiss MA, Arunagiri A, Yong J, Rege N, Sun J, Haataja L, Kaufman RJ, and Arvan P (2018). Biosynthesis, structure, and folding of the insulin precursor protein. Diabetes Obes Metab 20 Suppl 2, 28–50. 10.1111/dom.13378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Donath MY, Böni-Schnetzler M, Ellingsgaard H, and Ehses JA (2009). Islet inflammation impairs the pancreatic beta-cell in type 2 diabetes. Physiology (Bethesda) 24, 325–331. 10.1152/physiol.00032.2009. [DOI] [PubMed] [Google Scholar]
  • 154.Sharma RB, Landa-Galván HV, and Alonso LC (2021). Living Dangerously: Protective and Harmful ER Stress Responses in Pancreatic β-Cells. Diabetes 70, 2431–2443. 10.2337/dbi20-0033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Satoh T, Abiru N, Kobayashi M, Zhou H, Nakamura K, Kuriya G, Nakamura H, Nagayama Y, Kawasaki E, Yamasaki H, et al. (2011). CHOP deletion does not impact the development of diabetes but suppresses the early production of insulin autoantibody in the NOD mouse. Apoptosis 16, 438–448. 10.1007/s10495-011-0576-2. [DOI] [PubMed] [Google Scholar]
  • 156.Kaneto H, Kimura T, Shimoda M, Obata A, Sanada J, Fushimi Y, Matsuoka T-A, and Kaku K (2022). Molecular Mechanism of Pancreatic β-Cell Failure in Type 2 Diabetes Mellitus. Biomedicines 10, 818. 10.3390/biomedicines10040818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Ma J, Liu Y, Valladolid-Acebes I, Recio-López P, Peng G, Li J, Berggren P-O, Juntti-Berggren L, and Tong N (2023). ATF5 is a regulator of ER stress and β-cell apoptosis in different mouse models of genetic- and diet-induced obesity and diabetes mellitus. Cell Signal 102, 110535. 10.1016/j.cellsig.2022.110535. [DOI] [PubMed] [Google Scholar]
  • 158.Song B, Scheuner D, Ron D, Pennathur S, and Kaufman RJ (2008). Chop deletion reduces oxidative stress, improves beta cell function, and promotes cell survival in multiple mouse models of diabetes. J Clin Invest 118, 3378–3389. 10.1172/JCI34587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Lee J-H, Ryu H, Lee H, Yu HR, Gao Y, Lee K-M, Kim Y-J, and Lee J (2023). Endoplasmic reticulum stress in pancreatic β cells induces incretin desensitization and β-cell dysfunction via ATF4-mediated PDE4D expression. Am J Physiol Endocrinol Metab 325, E448–E465. 10.1152/ajpendo.00156.2023. [DOI] [PubMed] [Google Scholar]
  • 160.Cuenco J, and Dalmas E (2022). Islet Inflammation and β Cell Dysfunction in Type 2 Diabetes. Handb Exp Pharmacol 274, 227–251. 10.1007/164_2021_571. [DOI] [PubMed] [Google Scholar]
  • 161.Weaver JR, Odanga JJ, Breathwaite EK, Treadwell ML, Murchinson AC, Walters G, Fuentes DP, and Lee JB (2021). An increase in inflammation and islet dysfunction is a feature of prediabetes. Diabetes Metab Res Rev 37, e3405. 10.1002/dmrr.3405. [DOI] [PubMed] [Google Scholar]
  • 162.Pitasi CL, Liu J, Gausserès B, Pommier G, Delangre E, Armanet M, Cattan P, Mégarbane B, Hanak A-S, Maouche K, et al. (2020). Implication of glycogen synthase kinase 3 in diabetes-associated islet inflammation. J Endocrinol 244, 133–148. 10.1530/JOE-19-0239. [DOI] [PubMed] [Google Scholar]
  • 163.Liu N, Li R, Cao J, Song X, Ma W, Liu T, Wang L, Zou J, Zhang B, Liu Z, et al. (2023). The inhibition of FKBP5 protects β-cell survival under inflammation stress via AKT/FOXO1 signaling. Cell Death Discov 9, 247. 10.1038/s41420-023-01506-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Prause M, Pedersen SS, Tsonkova V, Qiao M, and Billestrup N (2021). Butyrate Protects Pancreatic Beta Cells from Cytokine-Induced Dysfunction. Int J Mol Sci 22, 10427. 10.3390/ijms221910427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Bosi E, Marchetti P, Rutter GA, and Eizirik DL (2022). Human alpha cell transcriptomic signatures of types 1 and 2 diabetes highlight disease-specific dysfunction pathways. iScience 25, 105056. 10.1016/j.isci.2022.105056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Marroqui L, Masini M, Merino B, Grieco FA, Millard I, Dubois C, Quesada I, Marchetti P, Cnop M, and Eizirik DL (2015). Pancreatic α Cells are Resistant to Metabolic Stress-induced Apoptosis in Type 2 Diabetes. EBioMedicine 2, 378–385. 10.1016/j.ebiom.2015.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Wali JA, and Thomas HE (2015). Pancreatic Alpha Cells Hold the Key to Survival. EBioMedicine 2, 368–369. 10.1016/j.ebiom.2015.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Eizirik DL, Szymczak F, and Mallone R (2023). Why does the immune system destroy pancreatic β-cells but not α-cells in type 1 diabetes? Nat Rev Endocrinol 19, 425–434. 10.1038/s41574-023-00826-3. [DOI] [PubMed] [Google Scholar]
  • 169.Butler AE, Janson J, Bonner-Weir S, Ritzel R, Rizza RA, and Butler PC (2003). Beta-cell deficit and increased beta-cell apoptosis in humans with type 2 diabetes. Diabetes 52, 102–110. [DOI] [PubMed] [Google Scholar]
  • 170.Shrestha N, De Franco E, Arvan P, and Cnop M (2021). Pathological β-Cell Endoplasmic Reticulum Stress in Type 2 Diabetes: Current Evidence. Front Endocrinol (Lausanne) 12, 650158. 10.3389/fendo.2021.650158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Jiang W-L, Zhang Y-F, Xia Q-Q, Zhu J, Yu X, Fan T, and Wang F (2015). MicroRNA-19a regulates lipopolysaccharide-induced endothelial cell apoptosis through modulation of apoptosis signal-regulating kinase 1 expression. BMC Mol Biol 16, 11. 10.1186/s12867-015-0034-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Luyendyk JP, Piper JD, Tencati M, Reddy KV, Holscher T, Zhang R, Luchoomun J, Chen X, Min W, Kunsch C, et al. (2007). A novel class of antioxidants inhibit LPS induction of tissue factor by selective inhibition of the activation of ASK1 and MAP kinases. Arterioscler Thromb Vasc Biol 27, 1857–1863. 10.1161/ATVBAHA.107.143552. [DOI] [PubMed] [Google Scholar]
  • 173.Ichijo H, Nishida E, Irie K, ten Dijke P, Saitoh M, Moriguchi T, Takagi M, Matsumoto K, Miyazono K, and Gotoh Y (1997). Induction of apoptosis by ASK1, a mammalian MAPKKK that activates SAPK/JNK and p38 signaling pathways. Science 275, 90–94. 10.1126/science.275.5296.90. [DOI] [PubMed] [Google Scholar]
  • 174.Zuo C, Shin S, and Keleş S (2015). atSNP: transcription factor binding affinity testing for regulatory SNP detection. Bioinformatics 31, 3353–3355. 10.1093/bioinformatics/btv328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Benazra M, Lecomte M-J, Colace C, Müller A, Machado C, Pechberty S, Bricout-Neveu E, Grenier-Godard M, Solimena M, Scharfmann R, et al. (2015). A human beta cell line with drug inducible excision of immortalizing transgenes. Mol Metab 4, 916–925. 10.1016/j.molmet.2015.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, and Gingeras TR (2013). STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21. 10.1093/bioinformatics/bts635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Hartley SW, and Mullikin JC (2015). QoRTs: a comprehensive toolset for quality control and data processing of RNA-Seq experiments. BMC Bioinformatics 16, 224. 10.1186/s12859-015-0670-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Howe KL, Achuthan P, Allen J, Allen J, Alvarez-Jarreta J, Amode MR, Armean IM, Azov AG, Bennett R, Bhai J, et al. (2021). Ensembl 2021. Nucleic Acids Res 49, D884–D891. 10.1093/nar/gkaa942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Risso D, Ngai J, Speed TP, and Dudoit S (2014). Normalization of RNA-seq data using factor analysis of control genes or samples. Nat Biotechnol 32, 896–902. 10.1038/nbt.2931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Leek JT (2014). svaseq: removing batch effects and other unwanted noise from sequencing data. Nucleic Acids Res 42, e161. 10.1093/nar/gku864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Robinson MD, McCarthy DJ, and Smyth GK (2010). edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140. 10.1093/bioinformatics/btp616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Sherman BT, Hao M, Qiu J, Jiao X, Baseler MW, Lane HC, Imamichi T, and Chang W (2022). DAVID: a web server for functional enrichment analysis and functional annotation of gene lists (2021 update). Nucleic Acids Res 50, W216–W221. 10.1093/nar/gkac194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Kanehisa M, and Goto S (2000). KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res 28, 27–30. 10.1093/nar/28.1.27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Vastrik I, D’Eustachio P, Schmidt E, Gopinath G, Croft D, de Bono B, Gillespie M, Jassal B, Lewis S, Matthews L, et al. (2007). Reactome: a knowledge base of biologic pathways and processes. Genome Biol 8, R39. 10.1186/gb-2007-8-3-r39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Pico AR, Kelder T, van Iersel MP, Hanspers K, Conklin BR, and Evelo C (2008). WikiPathways: pathway editing for the people. PLoS Biol 6, e184. 10.1371/journal.pbio.0060184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.bcl2fastq Conversion Software (2023). https://support.illumina.com/sequencing/sequencing_software/bcl2fastq-conversion-software.html.
  • 187.Zheng GXY, Terry JM, Belgrader P, Ryvkin P, Bent ZW, Wilson R, Ziraldo SB, Wheeler TD, McDermott GP, Zhu J, et al. (2017). Massively parallel digital transcriptional profiling of single cells. Nat Commun 8, 14049. 10.1038/ncomms14049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Single-Library Analysis with Cell Ranger count -Software -Single Cell Gene Expression - Official 10x Genomics Support (2023). https://support.10xgenomics.com/single-cell-gene-expression/software/pipelines/latest/using/count.
  • 189.Hoohm/CITE-seq-Count: 1.4.2 (2023). 10.5281/zenodo.2590196. [DOI]
  • 190.Kang HM, Subramaniam M, Targ S, Nguyen M, Maliskova L, McCarthy E, Wan E, Wong S, Byrnes L, Lanata CM, et al. (2018). Multiplexed droplet single-cell RNA-sequencing using natural genetic variation. Nat. Biotechnol 36, 89–94. 10.1038/nbt.4042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Young MD, and Behjati S (2020). SoupX removes ambient RNA contamination from droplet-based single-cell RNA sequencing data. Gigascience 9, giaa151. 10.1093/gigascience/giaa151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Hao Y, Hao S, Andersen-Nissen E, Mauck WM, Zheng S, Butler A, Lee MJ, Wilk AJ, Darby C, Zager M, et al. (2021). Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587.e29. 10.1016/j.cell.2021.04.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Wolock SL, Lopez R, and Klein AM (2019). Scrublet: Computational Identification of Cell Doublets in Single-Cell Transcriptomic Data. Cell Syst 8, 281–291.e9. 10.1016/j.cels.2018.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Korsunsky I, Millard N, Fan J, Slowikowski K, Zhang F, Wei K, Baglaenko Y, Brenner M, Loh P-R, and Raychaudhuri S (2019). Fast, sensitive and accurate integration of single-cell data with Harmony. Nat Methods 16, 1289–1296. 10.1038/s41592-019-0619-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Andreatta M, and Carmona SJ (2021). UCell: Robust and scalable single-cell gene signature scoring. Comput Struct Biotechnol J 19, 3796–3798. 10.1016/j.csbj.2021.06.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Finak G, McDavid A, Yajima M, Deng J, Gersuk V, Shalek AK, Slichter CK, Miller HW, McElrath MJ, Prlic M, et al. (2015). MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data. Genome Biol 16, 278. 10.1186/s13059-015-0844-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Bolger AM, Lohse M, and Usadel B (2014). Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120. 10.1093/bioinformatics/btu170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Li H, and Durbin R (2009). Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760. 10.1093/bioinformatics/btp324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Lawlor N, Youn A, Kursawe R, Ucar D, and Stitzel ML (2017). Alpha TC1 and Beta-TC-6 genomic profiling uncovers both shared and distinct transcriptional regulatory features with their primary islet counterparts. Sci Rep 7, 11959. 10.1038/s41598-017-12335-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Ucar D, Márquez EJ, Chung C-H, Marches R, Rossi RJ, Uyar A, Wu T-C, George J, Stitzel ML, Palucka AK, et al. (2017). The chromatin accessibility signature of human immune aging stems from CD8+ T cells. J Exp Med 214, 3123–3144. 10.1084/jem.20170416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, Marth G, Abecasis G, Durbin R, and 1000 Genome Project Data Processing Subgroup (2009). The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079. 10.1093/bioinformatics/btp352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Zhang Y, Liu T, Meyer CA, Eeckhoute J, Johnson DS, Bernstein BE, Nusbaum C, Myers RM, Brown M, Li W, et al. (2008). Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137. 10.1186/gb-2008-9-9-r137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Thorvaldsdóttir H, Robinson JT, and Mesirov JP (2013). Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief Bioinform 14, 178–192. 10.1093/bib/bbs017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Amemiya HM, Kundaje A, and Boyle AP (2019). The ENCODE Blacklist: Identification of Problematic Regions of the Genome. Sci Rep 9, 9354. 10.1038/s41598-019-45839-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Ross-Innes CS, Stark R, Teschendorff AE, Holmes KA, Ali HR, Dunning MJ, Brown GD, Gojis O, Ellis IO, Green AR, et al. (2012). Differential oestrogen receptor binding is associated with clinical outcome in breast cancer. Nature 481, 389–393. 10.1038/nature10730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Lawrence M, Huber W, Pagès H, Aboyoun P, Carlson M, Gentleman R, Morgan MT, and Carey VJ (2013). Software for computing and annotating genomic ranges. PLoS Comput Biol 9, e1003118. 10.1371/journal.pcbi.1003118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Harrow J, Denoeud F, Frankish A, Reymond A, Chen C-K, Chrast J, Lagarde J, Gilbert JGR, Storey R, Swarbreck D, et al. (2006). GENCODE: producing a reference annotation for ENCODE. Genome Biol 7 Suppl 1, S4.1–9. 10.1186/gb-2006-7-s1-s4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.1000 Genomes Project Consortium, Abecasis GR, Auton A, Brooks LD, DePristo MA, Durbin RM, Handsaker RE, Kang HM, Marth GT, and McVean GA (2012). An integrated map of genetic variation from 1,092 human genomes. Nature 491, 56–65. 10.1038/nature11632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Sherry ST, Ward MH, Kholodov M, Baker J, Phan L, Smigielski EM, and Sirotkin K (2001). dbSNP: the NCBI database of genetic variation. Nucleic Acids Res 29, 308–311. 10.1093/nar/29.1.308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Bonfield JK, Marshall J, Danecek P, Li H, Ohan V, Whitwham A, Keane T, and Davies RM (2021). HTSlib: C library for reading/writing high-throughput sequencing data. Gigascience 10, giab007. 10.1093/gigascience/giab007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Chen J, Spracklen CN, Marenne G, Varshney A, Corbin LJ, Luan J, Willems SM, Wu Y, Zhang X, Horikoshi M, et al. (2021). The trans-ancestral genomic architecture of glycemic traits. Nat Genet 53, 840–860. 10.1038/s41588-021-00852-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Lawlor N, George J, Bolisetty M, Kursawe R, Sun L, Sivakamasundari V, Kycia I, Robson P, and Stitzel ML (2017). Single-cell transcriptomes identify human islet cell signatures and reveal cell-type–specific expression changes in type 2 diabetes. Genome Res. 27, 208–222. 10.1101/gr.212720.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Schneider CA, Rasband WS, and Eliceiri KW (2012). NIH Image to ImageJ: 25 years of image analysis. Nat Methods 9, 671–675. 10.1038/nmeth.2089. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

Data S1. Data used to generate figures in this manuscript, related to Figures 15 and Figures S1-5

2
3

Table S1. Donor demographics, related to Figures 15

4

Table S2. RNA-seq analyses, related to Figures 1, 3, 4-5

5

Table S3. scRNA-seq analyses, related to Figures 2, 4-5

6

Table S4. ATAC-seq analyses, related to Figures 35

7

Table S5. Overlapping genetic variants with stress-responsive CREs, related to Figures 45

Data Availability Statement

RESOURCES