Type 1 diabetes is a multifactorial inflammatory disease in genetically susceptible individuals characterized by progressive autoimmune destruction of pancreatic β-cells initiated by yet unknown factors. Although animal models of type 1 diabetes have substantially increased our understanding of disease pathogenesis, heterogeneity seen in human patients cannot be reflected by a single model and calls for additional models covering different aspects of human pathophysiology. Inhibitor of κB kinase (IKK)/nuclear factor-κB (NF-κB) signaling is a master regulator of inflammation; however, its role in diabetes pathogenesis is controversially discussed by studies using different inhibition approaches. To investigate the potential diabetogenic effects of NF-κB in β-cells, we generated a gain-of-function model allowing conditional IKK2/NF-κB activation in β-cells. A transgenic mouse model that expresses a constitutively active mutant of human IKK2 dependent on Pdx-1 promoter activity (IKK2-CAPdx-1) spontaneously develops full-blown immune-mediated diabetes with insulitis, hyperglycemia, and hypoinsulinemia. Disease development involves a gene expression program mimicking virus-induced diabetes and allergic inflammatory responses as well as increased major histocompatibility complex class I/II expression by β-cells that could collectively promote diabetes development. Potential novel diabetes candidate genes were also identified. Interestingly, animals successfully recovered from diabetes upon transgene inactivation. Our data give the first direct evidence that β-cell–specific IKK2/NF-κB activation is a potential trigger of immune-mediated diabetes. Moreover, IKK2-CAPdx-1 mice provide a novel tool for studying critical checkpoints in diabetes pathogenesis and mechanisms governing β-cell degeneration/regeneration.

Type 1 diabetes is an autoimmune disease in genetically predisposed individuals and presumably triggered and/or accelerated by environmental factors (1). Analyses of animal models of type 1 diabetes have greatly improved our knowledge about disease pathophysiology and genetic contribution. However, there is still an unmet need to understand islet cell pathology and ongoing inflammatory processes within the islets of Langerhans.

In type 1 diabetes, inflammation contributes to the induction and amplification of the immune insult against β-cells and, at later stages, to the stabilization and maintenance of the insulitic process, thus promoting disease development and progression (2). Furthermore, β-cell response to stress and inflammation is thought to be a critical determinant in disease outcome (2). Canonical inhibitor of κB (IκB) kinase 2 (IKK2)/nuclear factor-κB (NF-κB) signaling is the master regulator of inflammatory responses and innate immunity (3), and it is activated by viral and bacterial pathogens, various cytokines, and general stress factors. Apart from its antiapoptotic function in most cell types, the predominant role of NF-κB activation in β-cells appears to be of a death-promoting nature (4,5).

The transcription factor NF-κB exists as homo- and heterodimers of five different subunits. In resting cells, NF-κB dimers are sequestered in the cytoplasm by IκB proteins. A key step in connecting extracellular stimuli to NF-κB induction is the activation of the IKK complex composed of the catalytic subunits IKK1 and IKK2 and the regulatory component NF-κB essential modulator. This complex phosphorylates IκB proteins, thereby initiating their ubiquitination and subsequent proteasomal degradation, thus allowing nuclear translocation of NF-κB (4,5).

To mimic an inflammatory insult selectively in pancreatic β-cells, we created a mouse model that allows the conditional expression of a constitutively active IKK2 allele in β-cells. Interestingly, prolonged IKK2/NF-κB activation in β-cells is sufficient to induce insulitis with marked hyperglycemia, hypoinsulinemia, and reduced β-cell mass in transgenic animals. Intriguingly, upon switching off transgene expression, diabetes could be efficiently reversed.

Mice

Male mice were housed under specific pathogen-free conditions at the animal facility of the University of Ulm. Pdx-1.tTA mice (C57BL/6) (6) and (tetO)7.IKK2-CA (constitutively active mutant of human IKK2) mice (NMRI) (7) were described previously. Pdx-1.tTA mice are knockin animals in which the coding sequence of tetracycline-dependent transactivator (tTA) has replaced the endogenous Pdx-1 gene, thereby rendering this mouse line heterozygous for Pdx-1 (Pdx-1+/−). Control mice group include wild-type and single-transgenic (tetO)7.IKK2-CA mice unless otherwise stated. Experiments were performed in accordance with institutional guidelines and German animal protection law.

Metabolic Studies

Blood glucose was measured using the One-Touch Ultra glucometer (LifeScan Inc., Mipitas, CA). Pancreatic insulin was extracted by overnight agitation with cold acid ethanol (0.18 mol/L HCl in 70% ethanol) at 4°C. The supernatant was collected, and the pellet was re-extracted. The pooled supernatant was used for insulin measurement. Insulin was determined in plasma samples containing protease inhibitor and pancreatic extracts using the Ultra-Sensitive Mouse Insulin ELISA Kit (Chrystal Chem Inc.) following the manufacturer’s instructions.

Protein Extraction, Western Immunoblotting, Luciferase Assay, and Electrophoretic Mobility Shift Assay

Pancreata were snap-frozen in liquid nitrogen and pulverized, and proteins were extracted for Western immunoblotting and luciferase activity measurement (7). For immunoblotting, antibodies against IKK2 (Abcam) and extracellular signal–related kinase-2 (Santa Cruz Biotechnology) were used. Electrophoretic mobility shift assay was performed as described (7) using whole-cell extract from isolated islets.

Histology and Immunostaining

For paraffin sections, pancreata were excised, fixed overnight in 3.8% buffered formalin, dehydrated, paraffin embedded, cut in 3-μm sections, and further processed as previously described (8). For cryosections, 10-μm slices from natively frozen pancreata were fixed with 4% paraformaldehyde. Sections were incubated with the primary antibodies: rabbit (Cell Signaling Technology) or guinea pig (Abcam) anti-insulin, goat anti-human IKK2 (Santa Cruz Biotechnology), rabbit anti-RelA/p65 (Laboratory Vision), anti–Pdx-1 and antichromogranin A (Abcam), anti-Ki67 (Thermo Scientific), rat anti-CD4 and anti-CD8 (Abcam), anti–CD11c-phycoerythrin and anti-B220 (BD Biosciences), and anti–CD25-phycoerythrin and anti–major histocompatibility complex (MHC) class II (MHC II)–FITC (eBioscience). Secondary antibodies were coupled with Alexa Fluor (Invitrogen) for immunofluorescence and with horseradish peroxidase in immunohistochemistry that was developed by 3-amino-9-ethylcarbazole (DakoCytomation). Immunofluorescent stainings were visualized as before (8), and other stainings were analyzed on a Leica DM IRB microscope (Leica Microsystems) equipped with ProgRes C14 digital camera (Jenoptik) and Openlab software (Improvision).

Detection of Apoptosis

In situ detection of DNA strand breaks was performed using the TUNEL labeling method with the FragEL DNA Fragmentation Detection Kit and colorimetric-TdT enzyme (EMD Millipore) according to the manufacturer’s instructions.

Islet Isolation

Pancreata were inflated in situ with 0.5 mg/mL ice-cold collagenase XI solution (Sigma-Aldrich, St. Louis, MO) dissolved in PBS (with Ca/Mg). Pancreatic tissue was dissected out and digested for 19–24 min at 37°C with subsequent washing and centrifugation. Islets in the sediment were purified with Histopaque 10771 (Sigma-Aldrich), washed, and frozen in liquid N2.

RNA Extraction, Quantitative PCR, and Microarray

RNA was extracted with RNeasy kit (Quiagen) and cDNA synthesis was done using the Transcriptor High Fidelity cDNA Synthesis Kit (Roche). Quantitative real-time PCR was performed with the Roche LightCycler 480 (Roche) using gene-specific primers and hydrolysis probes designed by the Roche Universal Probe Library system (Roche). Microarray analysis was performed with the Mouse Gene 1.0 ST array (Affymetrix) and evaluated with the “Genesifter” sofware (Geospiza). The expression data are available at Gene Expression Omnibus super series (accession number GSE47504).

Flow Cytometry

Pancreatic cells were isolated and purified as previously described (9) and subsequently stained with monoclonal antibodies using standard procedures. The following antibodies were purchased from eBioscience: anti–MHC II (M5/114.15.2), anti–MHC class I (MHC I; AF6–88.5.5.3), anti-CD11b (M1/70), anti-CD3e (17A2), anti-CD25 (PC61.5), and anti-CD69 (H1.2F3); from BD Biosciences: anti-CD11b (M1/70), anti-CD8a (53–6.7), anti-CD4 (RM4–5), CD44 (IM7), anti-NK1.1 (PK136), and anti-CD11c (HL3). Anti-CD45 (30-F11), anti-CD19 (6D5), anti-F4/80 (BM8), and anti-Ly6G (1A8) were from BioLegend, while Ly6C (1G7.G10) was from Miltenyi Biotec. Fixable viability dye (eBioscience) was used to exclude dead cells. FACS was performed on an FACSCanto II (BD Biosciences), and data were analyzed with FACSDiva 6.2 (BD Biosciences) and FlowJo softwares (Tree Star).

Statistical Analysis

Values are given as mean ± SEM. Statistical analysis was performed with the Prism software (GraphPad) using two-tailed Student t test. Data with P values of ≤0.05 were considered statistically significant.

Generation of Conditional, Gain-of-Function Mouse Model for Canonical NF-κB Signaling in Pancreatic β-Cells

To directly explore the biological consequences of canonical NF-κB activation in pancreatic β-cells, we generated IKK2-CAPdx-1. This was achieved by crossing transgenic mice expressing the tTA under the control of Pdx-1 promoter (Pdx-1+/−) (6) with mice carrying the luciferase-(tetO)7-IKK2-CA minigene (7) (Fig. 1A). The transgenic expression system was not activated until weaning in order to avoid any effects of IKK2-CA activity on pancreas development. Measurement of luciferase activity, the coexpressed reporter gene, revealed strong transgene activity in the pancreas of IKK2-CAPdx-1 mice after doxycycline withdrawal and only minor activity in the intestine (Fig. 1B). Pancreatic IKK2-CA expression was confirmed by immunoblot and could be switched off by doxycycline (Fig. 1C), thus allowing conditional regulation of IKK2 activity. Immunofluorescence staining demonstrated a mosaic expression of IKK2-CA exclusively in the islets of IKK2-CAPdx-1 mice (Fig. 1D), correlating with obvious reduction in insulin immunoreactivity (Fig. 1D).

Figure 1

Gain-of-function mouse model for conditional activation of IKK2 in pancreatic β-cells. A: Transgenic approach for doxycycline (Dox)-regulated expression of IKK2-CA in pancreatic β-cells. tTA protein is expressed under the control of Pdx-1 promoter and can bind to a bidirectional promoter [(tetO)7; 7xtetO] driving the transcription of luciferase and IKK2-CA transgene. This binding is inhibited by Dox, thereby shutting off transgene expression. The Pdx-1.tTA mice are knockin animals in which the coding sequence of tTA has replaced the endogenous Pdx-1 gene, thereby rendering this mouse line heterozygous for Pdx-1 (Pdx-1+/−). Animals were bred under Dox (0.1 g/L Dox in drinking water) to avoid developmental defects, which then had been withdrawn after weaning (3 to 4 weeks) to activate transgene expression. B: Luciferase activity was measured in pancreas (Pa), spleen (Sp), liver (Li), kidney (Ki), stomach (St), intestine (Int), lung (Lu), thymus (Thy), thalamus/hypothalamus (Tha), and brain (Br) of 26- to 32-week-old control (white bars) and IKK2-CAPdx-1 mice (black bars) (n = 2). High luciferase activity was detected in pancreas of IKK2-CAPdx-1 mice and only minor activity in their intestine. C: Strong IKK2 expression was detected by Western blot in pancreatic extracts of IKK2-CAPdx-1 mice after doxycycline (Dox) withdrawal and disappeared after Dox readministration; extracellular signal–related kinase-2 (ERK2) was used as loading control. D: Immunofluorescence staining of paraffin-embedded pancreatic sections showing mosaic expression of transgenic human IKK2 in islets of 12-week-old IKK2-CAPdx-1 mice. The staining shows that IKK2-positive cells have reduced or even no insulin immunoreactivity. Sections were costained with DAPI (blue) for nuclei. Scale bar, 50 μm. E: An electrophoretic mobility shift assay of whole-cell islet extracts with NF-κB and SP1-specific probes showing strong activation of NF-κB in 12-week-old IKK2-CAPdx-1 mice. SP1 serves as a quality control. F: Immunohistological staining of paraffin-embedded pancreatic sections showing IKK2-CA–induced nuclear localization of RelA in β-cells of 12-week-old IKK2-CAPdx-1 mice. Scale bar, 50 μm. Controls denoted in this figure were single transgenic for IKK2-CA transgene. RLU, relative light unit.

Figure 1

Gain-of-function mouse model for conditional activation of IKK2 in pancreatic β-cells. A: Transgenic approach for doxycycline (Dox)-regulated expression of IKK2-CA in pancreatic β-cells. tTA protein is expressed under the control of Pdx-1 promoter and can bind to a bidirectional promoter [(tetO)7; 7xtetO] driving the transcription of luciferase and IKK2-CA transgene. This binding is inhibited by Dox, thereby shutting off transgene expression. The Pdx-1.tTA mice are knockin animals in which the coding sequence of tTA has replaced the endogenous Pdx-1 gene, thereby rendering this mouse line heterozygous for Pdx-1 (Pdx-1+/−). Animals were bred under Dox (0.1 g/L Dox in drinking water) to avoid developmental defects, which then had been withdrawn after weaning (3 to 4 weeks) to activate transgene expression. B: Luciferase activity was measured in pancreas (Pa), spleen (Sp), liver (Li), kidney (Ki), stomach (St), intestine (Int), lung (Lu), thymus (Thy), thalamus/hypothalamus (Tha), and brain (Br) of 26- to 32-week-old control (white bars) and IKK2-CAPdx-1 mice (black bars) (n = 2). High luciferase activity was detected in pancreas of IKK2-CAPdx-1 mice and only minor activity in their intestine. C: Strong IKK2 expression was detected by Western blot in pancreatic extracts of IKK2-CAPdx-1 mice after doxycycline (Dox) withdrawal and disappeared after Dox readministration; extracellular signal–related kinase-2 (ERK2) was used as loading control. D: Immunofluorescence staining of paraffin-embedded pancreatic sections showing mosaic expression of transgenic human IKK2 in islets of 12-week-old IKK2-CAPdx-1 mice. The staining shows that IKK2-positive cells have reduced or even no insulin immunoreactivity. Sections were costained with DAPI (blue) for nuclei. Scale bar, 50 μm. E: An electrophoretic mobility shift assay of whole-cell islet extracts with NF-κB and SP1-specific probes showing strong activation of NF-κB in 12-week-old IKK2-CAPdx-1 mice. SP1 serves as a quality control. F: Immunohistological staining of paraffin-embedded pancreatic sections showing IKK2-CA–induced nuclear localization of RelA in β-cells of 12-week-old IKK2-CAPdx-1 mice. Scale bar, 50 μm. Controls denoted in this figure were single transgenic for IKK2-CA transgene. RLU, relative light unit.

Close modal

Electrophoretic mobility shift assays showed strong basal NF-κB activity in IKK2-CAPdx-1 mice relative to their Pdx-1+/− littermates, demonstrating transgene functionality (Fig. 1E). NF-κB activity was also elevated in Pdx-1+/− mice compared with controls. Furthermore, IKK2 activation led to nuclear translocation of the NF-κB subunit RelA in β-cells of IKK2-CAPdx-1 mice, while almost no nuclear RelA was detected in islets of either Pdx-1+/− or control mice (Fig. 1F).

IKK2/NF-κB Activation in β-Cells Induces Full-Blown Diabetes

We then assessed the physiological and histological consequences of IKK2/NF-κB signaling in pancreatic β-cells. Interestingly, at the age of 11 weeks, some animals started to develop hyperglycemia, and at ∼24 weeks of age, all IKK2-CAPdx-1 mice showed substantial elevation in fed (538.5 ± 16 vs. 154.9 ± 7 mg/dL in Pdx-1+/− mice) and fasting (423.3 ± 34 vs. 96.9 ± 4 mg/dL in Pdx-1+/− mice) blood glucose levels, with several animals >600 mg/dL (Fig. 2A and B). Hyperglycemia was accompanied by a 57% reduction of plasma insulin levels compared with Pdx-1+/− mice (Fig. 2C), indicating that these mice were overtly diabetic. Animals displayed clinical signs of diabetes, including polyuria, polydipsia, weight loss, and general sickness, and in severe cases, mortality was recorded. Furthermore, immunohistological analyses indicated an extensive loss of insulin-positive β-cells in IKK2-CAPdx-1 mice, with the remaining β-cells appearing degranulated with only faint residual insulin immunoreactivity (Fig. 2D) and reduced Pdx-1 expression (Supplementary Fig. 1A). Costaining with IKK2 revealed that most of the IKK2-CA–expressing cells were no longer positive for insulin. Interestingly, few insulin-negative cells still expressed nuclear Pdx-1, while more such cells retained the expression of the endocrine marker chromogranin A (Supplementary Fig. 1A and B). This implicates that IKK2/NF-κB activation in β-cells leads to suppression of insulin expression and thus loss of their functionality in addition (or prior) to their demise. In accordance, pancreatic insulin content and insulin transcription had dropped to ∼3 and 26% of the values of the Pdx-1+/− littermates, respectively (Fig. 2E and F).

Figure 2

Expression of IKK2-CA in β-cells results in diabetes development. Animals were kept under doxycycline during breeding, doxycycline was removed after weaning, and mice were then analyzed at the indicated ages (A) or at 24–28 weeks (BF). A: Fed blood glucose levels of 8, 11–13, 14–17, and 24- to 28-week-old control (n = 9, 13, 7, and 28), Pdx-1+/− (n = 6, 12, 3, and 17), and IKK2-CAPdx-1 mice (n = 5, 12, 7, and 28). The upper detection limit of the glucometer is 600 mg/dL. B: Overnight-fasted blood glucose levels of control (n = 21), Pdx-1+/− (n = 16), and IKK2-CAPdx-1 mice (n = 22). C: Fed plasma insulin level of control (n = 12), Pdx-1+/− (n = 6), and IKK2-CAPdx-1 mice (n = 12). D: Representative immunofluorescent images of paraffin sections of IKK2-CAPdx-1 and Pdx-1+/− pancreata stained for insulin (red) and IKK2 (green) and costained with DAPI for nuclei in blue. Scale bar, 50 μm. E: Pancreatic insulin content of control, Pdx-1+/−, and IKK2-CAPdx-1 mice (n = 6–9/group). F: Quantitative RT-PCR of Ins2 mRNA of control, Pdx-1+/−, and IKK2-CAPdx-1 mice (fold upregulation vs. control; n = 8–12/group). Hprt was used as a reference gene. Results were analyzed by Student t test and presented as the mean ± SEM. **P < 0.01; ***P < 0.001.

Figure 2

Expression of IKK2-CA in β-cells results in diabetes development. Animals were kept under doxycycline during breeding, doxycycline was removed after weaning, and mice were then analyzed at the indicated ages (A) or at 24–28 weeks (BF). A: Fed blood glucose levels of 8, 11–13, 14–17, and 24- to 28-week-old control (n = 9, 13, 7, and 28), Pdx-1+/− (n = 6, 12, 3, and 17), and IKK2-CAPdx-1 mice (n = 5, 12, 7, and 28). The upper detection limit of the glucometer is 600 mg/dL. B: Overnight-fasted blood glucose levels of control (n = 21), Pdx-1+/− (n = 16), and IKK2-CAPdx-1 mice (n = 22). C: Fed plasma insulin level of control (n = 12), Pdx-1+/− (n = 6), and IKK2-CAPdx-1 mice (n = 12). D: Representative immunofluorescent images of paraffin sections of IKK2-CAPdx-1 and Pdx-1+/− pancreata stained for insulin (red) and IKK2 (green) and costained with DAPI for nuclei in blue. Scale bar, 50 μm. E: Pancreatic insulin content of control, Pdx-1+/−, and IKK2-CAPdx-1 mice (n = 6–9/group). F: Quantitative RT-PCR of Ins2 mRNA of control, Pdx-1+/−, and IKK2-CAPdx-1 mice (fold upregulation vs. control; n = 8–12/group). Hprt was used as a reference gene. Results were analyzed by Student t test and presented as the mean ± SEM. **P < 0.01; ***P < 0.001.

Close modal

Pdx-1 is an important transcription factor for β-cell function, and its heterozygosity, present in the Pdx-1+/− knockin mouse, was shown to be associated with some β-cell defects (6). Indeed, Pdx-1+/− mice showed slight elevation in fed blood glucose level (Fig. 2A) and reduction in insulin transcription and pancreatic content compared with control littermates (Fig. 2E and F); however, in contrast to IKK2-CAPdx-1 mice, they were not diabetic.

IKK2-CA–Induced Diabetes Is Associated With Inflammation and Leukocytic Infiltration

Invasion of pancreatic islets by leukocytes is the hallmark of immune-mediated diabetes. Unlike Pdx-1+/− mice, islets of IKK2-CAPdx-1 showed marked peri-insulitis (perivascular, periductal, and peri-islet infiltrates) and insulitis (Fig. 3A), reminiscent of the insulitic process in humans. Of note, similar to human histopathology, not all islets showed mononuclear infiltration (Supplementary Fig. 2).

Figure 3

IKK2/NF-κB activation in β-cells induces inflammation, insulitis, and increased islet antigen presentation. A: Hematoxylin-eosin staining of paraffin sections from pancreas of Pdx-1+/− and IKK2-CAPdx-1 mice showing peri-insulitis (I) and invasive insulitis (II) in IKK2-CAPdx-1 mice. B: Flow cytometric analysis of pancreatic cells from control (white bars) and IKK2-CAPdx-1 (black bars). The data are represented as relative to the number of CD45+ cells in the control samples, which is set to one (n = 5/group from two independent experiments). Immune cells were analyzed for the expression of surface markers CD3+ (T cells), CD19+ (B cells), and CD11b+/CD11c+ (dendritic cells [DCs]), CD11b+/F4/80+ (macrophages [Mϕ]), and NK1.1+ (natural killer cells [NK]). MHC II expression on pancreatic cells of the myeloid (C) and lymphoid lineage (D), analyzed by flow cytometry and gated as indicated. Elevated MHC II levels show that infiltrated cells in IKK2-CAPdx-1 were evidently activated. Data are representative of two experiments with five mice per group (C) or one experiment with two mice per group (D). Pdx-1+/− mice were included in the control group in BD, as they show the same pattern. E: Immunohistochemical staining of paraffin sections from Pdx-1+/− and IKK2-CAPdx-1 mice (n = 3/group) for the B-cell marker B220. F: Immunofluorescence staining of cryosections from pancreas of Pdx-1+/− and IKK2-CAPdx-1 mice (n = 3/group) for CD4, CD8, or CD11c (DC marker) together with insulin and DAPI. G: Immunofluorescence staining of cryosections from pancreas of Pdx-1+/− and IKK2-CAPdx-1 mice for insulin (red) and MHC II (green) (n = 3/group) showing the expression of MHC II by some β-cells (arrows, inset). Quantitative RT-PCR for mRNA transcripts involved in antigen presentation (H) and inflammation (I) from control (white bars), Pdx-1+/− (gray bars), and IKK2-CAPdx-1 (black bars) mice (fold upregulation vs. control; n = 8–12). Hprt was used as a reference gene. Results were analyzed by Student t test and presented as the mean ± SEM. Animals were 24–28 weeks old. Scale bars, 50 μm. *P < 0.05; **P < 0.01; ***P < 0.001. Icam1, intracellular adhesion molecule-1.

Figure 3

IKK2/NF-κB activation in β-cells induces inflammation, insulitis, and increased islet antigen presentation. A: Hematoxylin-eosin staining of paraffin sections from pancreas of Pdx-1+/− and IKK2-CAPdx-1 mice showing peri-insulitis (I) and invasive insulitis (II) in IKK2-CAPdx-1 mice. B: Flow cytometric analysis of pancreatic cells from control (white bars) and IKK2-CAPdx-1 (black bars). The data are represented as relative to the number of CD45+ cells in the control samples, which is set to one (n = 5/group from two independent experiments). Immune cells were analyzed for the expression of surface markers CD3+ (T cells), CD19+ (B cells), and CD11b+/CD11c+ (dendritic cells [DCs]), CD11b+/F4/80+ (macrophages [Mϕ]), and NK1.1+ (natural killer cells [NK]). MHC II expression on pancreatic cells of the myeloid (C) and lymphoid lineage (D), analyzed by flow cytometry and gated as indicated. Elevated MHC II levels show that infiltrated cells in IKK2-CAPdx-1 were evidently activated. Data are representative of two experiments with five mice per group (C) or one experiment with two mice per group (D). Pdx-1+/− mice were included in the control group in BD, as they show the same pattern. E: Immunohistochemical staining of paraffin sections from Pdx-1+/− and IKK2-CAPdx-1 mice (n = 3/group) for the B-cell marker B220. F: Immunofluorescence staining of cryosections from pancreas of Pdx-1+/− and IKK2-CAPdx-1 mice (n = 3/group) for CD4, CD8, or CD11c (DC marker) together with insulin and DAPI. G: Immunofluorescence staining of cryosections from pancreas of Pdx-1+/− and IKK2-CAPdx-1 mice for insulin (red) and MHC II (green) (n = 3/group) showing the expression of MHC II by some β-cells (arrows, inset). Quantitative RT-PCR for mRNA transcripts involved in antigen presentation (H) and inflammation (I) from control (white bars), Pdx-1+/− (gray bars), and IKK2-CAPdx-1 (black bars) mice (fold upregulation vs. control; n = 8–12). Hprt was used as a reference gene. Results were analyzed by Student t test and presented as the mean ± SEM. Animals were 24–28 weeks old. Scale bars, 50 μm. *P < 0.05; **P < 0.01; ***P < 0.001. Icam1, intracellular adhesion molecule-1.

Close modal

Flow cytometry revealed that hematopoietic (CD45+) cells in IKK2-CAPdx-1 pancreata increased fourfold (Fig. 3B) and that infiltrating leukocytes were activated CD4+ and CD8+ T, B, and dendritic cells as well as macrophages and few natural killer cells (Fig. 3B–D and Supplementary Fig. 3A). This was further confirmed on the histological level (Fig. 3E and F and Supplementary Figs. 3B and C and 4). The majority of infiltrating T cells was of the CD4+ type (Fig. 3B and F). Furthermore, we detected hyperexpression of MHC I and II by β-cells of IKK2-CAPdx-1 mice (Fig. 3G,Supplementary Fig. 5A–C), as previously reported in diabetic patients (10). This suggests increased antigen presentation capacity of β-cells in these mice, which might be critical for phenotype development. In accordance, there was transcriptional upregulation of the MHC I and II components, H2-Q4 and H2-Aa (Fig. 3H), respectively.

The insulitic process in IKK2-CAPdx-1 mice was associated with elevated expression of inflammatory cytokines and chemokines like tumor necrosis factor (Tnf), Ccl5, Ccl2, and Cxcl10 and the adhesion molecule intracellular adhesion molecule-1 (Fig. 3I). This inflammatory profile is reminiscent of those found in islets from patients, in animals with type 1 diabetes (11,12), and in normal human islets subjected to cytokines or enteroviruses (1315).

IKK2-CAPdx-1 Mice Show Signs of Apoptosis and Endoplasmic Reticulum Stress

As apoptosis is thought to be the major cause of β-cell death in diabetes (16), we performed TUNEL assays that showed the presence of apoptotic cells only in IKK2-CAPdx-1 islets (Fig. 4A and B). This indicates that apoptosis may account at least in part for the reduced β-cell mass in IKK2-CAPdx-1 mice. β-Cells are susceptible to endoplasmic reticulum (ER) stress, and ER stress-mediated apoptosis in β-cells has been implicated in the pathogenesis of diabetes (17). Moreover, islets from prediabetic NOD mice (18) and patients with type 1 diabetes (19) exhibit signs of ER stress. Consistent with that, the ER stress-related factors Ddit3/Chop and Atf3 were upregulated in IKK2-CAPdx-1 mice (Fig. 4C). In addition, other stress-associated apoptosis-inducing factors like Nos2 (iNOS) and Myc were also elevated in IKK2-CAPdx-1 pancreata (Fig. 4D and E).

Figure 4

Islets of IKK2-CAPdx-1 mice show signs of apoptosis and ER stress. A: TUNEL assay on paraffin sections of pancreas from 24- to 26-week-old control, Pdx-1+/−, and IKK2-CAPdx-1 mice (n = 3/group). Sections were counterstained with methyl green. Arrowheads indicate the presence of apoptotic nuclei (TUNEL-positive nuclei) in the islets of IKK2-CAPdx-1 mice. Dashed line marks the islet area. Scale bar, 50 μm. B: Quantification of TUNEL staining in A. A total of >400 islets from three animals per group were analyzed. CE: Quantitative RT-PCR for mRNA transcripts encoding stress and apoptosis-inducing genes in pancreata of 24- to 26-week-old control, Pdx-1+/−, and IKK2-CAPdx-1 (fold upregulation vs. control; n = 6–10/group). Hprt was used as a reference gene. *P < 0.05; **P < 0.01; ***P < 0.001. Results were analyzed by Student t test and presented as the mean ± SEM. ND, not detectable.

Figure 4

Islets of IKK2-CAPdx-1 mice show signs of apoptosis and ER stress. A: TUNEL assay on paraffin sections of pancreas from 24- to 26-week-old control, Pdx-1+/−, and IKK2-CAPdx-1 mice (n = 3/group). Sections were counterstained with methyl green. Arrowheads indicate the presence of apoptotic nuclei (TUNEL-positive nuclei) in the islets of IKK2-CAPdx-1 mice. Dashed line marks the islet area. Scale bar, 50 μm. B: Quantification of TUNEL staining in A. A total of >400 islets from three animals per group were analyzed. CE: Quantitative RT-PCR for mRNA transcripts encoding stress and apoptosis-inducing genes in pancreata of 24- to 26-week-old control, Pdx-1+/−, and IKK2-CAPdx-1 (fold upregulation vs. control; n = 6–10/group). Hprt was used as a reference gene. *P < 0.05; **P < 0.01; ***P < 0.001. Results were analyzed by Student t test and presented as the mean ± SEM. ND, not detectable.

Close modal

IKK2-CA–Induced Diabetes Is Reversible

To examine the possibility of reverse remodeling the diabetic phenotype, doxycycline was readministered for 30 days to diabetic IKK2-CAPdx-1 mice (Fig. 5A). Diabetes in IKK2-CAPdx-1 mice was confirmed by elevated fed and fasted blood glucose values and reduced plasma insulin levels (Fig. 5B and C). We observed a clear reduction in fed blood glucose values already within the first 10 days, which virtually completely normalized in all animals by 30 days (Fig. 5B). Consistently, fasting blood glucose and fed plasma insulin levels were also restored (Fig. 5C). Doxycycline-dependent transgene inactivation was confirmed by immunofluorescence staining (Fig. 5D) and Western blot (Fig. 1C). Diabetic IKK2-CAPdx-1 mice also regained normal structured islets showing virtual absence of infiltrating cells upon doxycycline treatment (Fig. 5D and E). Insulin immunostaining demonstrated the reappearance of β-cell–rich islets and prominent β-cell regranulation, which strongly stained for insulin (Fig. 5D). Importantly, the reversion of the diabetes status was accompanied by increased levels of Ki67 immunoreactivity in islets of IKK2-CAPdx-1 mice (Fig. 5F and Supplementary Fig. 6), indicating that proliferation is involved in β-cell regeneration in this model. Furthermore, transcription of insulin, MHC I/II molecules, and various inflammatory markers was effectively normalized (Fig. 5G–I).

Figure 5

Recovery from diabetes, alleviation of the inflammatory status, and regeneration of pancreatic islets upon transgene inactivation. A: Experimental design. Mice were kept under doxycycline (Dox; 0.1 g/L Dox in drinking water) until weaning and discontinued until 24–28 weeks to activate transgene expression. The diabetic phenotype was confirmed by fed and fasted blood glucose as well as plasma insulin measurements (before Dox). Dox (1 g/L) was then readministered in drinking water for 30 days (except for F) to inactivate transgene expression. Fed blood glucose was monitored during that time after which animals were analyzed (after Dox). B: Fed blood glucose monitoring before and during Dox readministration of control (white circles), Pdx-1+/− (white squares), and IKK2-CAPdx-1 (white triangles) mice (n = 7–9/group from two independent experiments). Significance is relative to the Pdx-1+/− group at the specified time point. At 20 and 30 days after Dox administration, Pdx-1+/− mice had significantly higher blood glucose values relative to the control littermates. C: Overnight-fasting blood glucose (top) and fed plasma insulin levels (bottom) before and after doxycycline readministration. Immunofluorescence (D) and hematoxylin-eosin stainings (E) of paraffin sections from Pdx-1+/− and IKK2-CAPdx-1 mice after Dox readministration showing regenerated β-cells with normal insulin content and near absence of infiltrating cells. F: Immunofluorescence staining of insulin (red), Ki67 (green), and DAPI (blue) of paraffin sections from Pdx-1+/− and IKK2-CAPdx-1 mice 10 days after Dox readministration. The staining shows the presence of increased Ki67-positive β-cells (arrows) in the islets of IKK2-CAPdx-1 mice as compared with Pdx-1+/− ones. Scale bars in DF, 50 μm. Quantitative RT-PCR for insulin mRNA (G), MHC I (H2-Q4) and MHC II (H2-Aa) molecules (H), and inflammatory genes (I) in pancreata of control (white bars), Pdx-1+/− (gray bars), and IKK2-CAPdx-1 (black bars) mice after Dox readministration (fold upregulation vs. control; n = 6–10/group from 2 independent experiments). Hprt was used as a reference gene. *P < 0.05; **P < 0.01; ***P < 0.001. Results were analyzed by Student t test and presented as the mean ± SEM. Icam1, intracellular adhesion molecule-1.

Figure 5

Recovery from diabetes, alleviation of the inflammatory status, and regeneration of pancreatic islets upon transgene inactivation. A: Experimental design. Mice were kept under doxycycline (Dox; 0.1 g/L Dox in drinking water) until weaning and discontinued until 24–28 weeks to activate transgene expression. The diabetic phenotype was confirmed by fed and fasted blood glucose as well as plasma insulin measurements (before Dox). Dox (1 g/L) was then readministered in drinking water for 30 days (except for F) to inactivate transgene expression. Fed blood glucose was monitored during that time after which animals were analyzed (after Dox). B: Fed blood glucose monitoring before and during Dox readministration of control (white circles), Pdx-1+/− (white squares), and IKK2-CAPdx-1 (white triangles) mice (n = 7–9/group from two independent experiments). Significance is relative to the Pdx-1+/− group at the specified time point. At 20 and 30 days after Dox administration, Pdx-1+/− mice had significantly higher blood glucose values relative to the control littermates. C: Overnight-fasting blood glucose (top) and fed plasma insulin levels (bottom) before and after doxycycline readministration. Immunofluorescence (D) and hematoxylin-eosin stainings (E) of paraffin sections from Pdx-1+/− and IKK2-CAPdx-1 mice after Dox readministration showing regenerated β-cells with normal insulin content and near absence of infiltrating cells. F: Immunofluorescence staining of insulin (red), Ki67 (green), and DAPI (blue) of paraffin sections from Pdx-1+/− and IKK2-CAPdx-1 mice 10 days after Dox readministration. The staining shows the presence of increased Ki67-positive β-cells (arrows) in the islets of IKK2-CAPdx-1 mice as compared with Pdx-1+/− ones. Scale bars in DF, 50 μm. Quantitative RT-PCR for insulin mRNA (G), MHC I (H2-Q4) and MHC II (H2-Aa) molecules (H), and inflammatory genes (I) in pancreata of control (white bars), Pdx-1+/− (gray bars), and IKK2-CAPdx-1 (black bars) mice after Dox readministration (fold upregulation vs. control; n = 6–10/group from 2 independent experiments). Hprt was used as a reference gene. *P < 0.05; **P < 0.01; ***P < 0.001. Results were analyzed by Student t test and presented as the mean ± SEM. Icam1, intracellular adhesion molecule-1.

Close modal

IKK2-CAPdx-1 Mice Exhibit Gene Expression Profile Mimicking Antiviral and Allergic Inflammatory Responses

To gain insight into molecular events induced by IKK2/NF-κB activation and possibly involved in diabetes development, we performed gene expression profiling of pancreatic islets isolated from 11-week-old IKK2-CAPdx-1 animals. This age represents a critical checkpoint in phenotype development at which some animals already showed hyperglycemia, while others were still normoglycemic (Fig. 6A). In this analysis, including samples from both types, 288 transcripts were found to be upregulated, 46 of which are known NF-κB targets (Supplementary Table 2) and 28 to be downregulated (threshold, 1.5-fold; P < 0.05 in t test with Benjamini-Hochberg correction) (Supplementary Table 1, selected genes, and Table 1). Several of those genes were verified by quantitative PCR (qPCR) as depicted in Fig. 6B. The increased expression of various cytokines, chemokines, adhesion molecules, and antigen presentation molecules clearly demonstrated an activation of innate and adaptive immune responses in islets of IKK2-CAPdx-1 mice (Table 1, Supplementary Table 1, and Fig. 6B). However, aside from these factors typically detected in other type 1 diabetes models, other less-characterized genes were highly upregulated (Table 1, Supplementary Table 1, and Fig. 6B). The T-cell–directed chemokine Ccl17 (also known as thymus and activation-regulated chemokine) was the most prominent one. Ccl17 together with Ccl22 (also known as macrophage-derived chemokine), another elevated chemokine, are known ligands for the Ccr4 receptor, which has been linked to the development of autoimmune diabetes (20). Also, a cluster of inflammation-related serine proteases called serine protease inhibitor clade A member 1 (Serpina1) and different members of the cationic protein products of eosinophils, the eosinophil-associated ribonucleases (Ears), were upregulated. Kyoto Encyclopedia of Genes and Genomes analysis clearly revealed a signature reflecting antiviral responses and reactions to infections. This was represented by the upregulation of different interferon (IFN)-regulated genes, especially involved in the type 1 IFN response including signal transducer and activator of transcription 1 (Stat1), IFN regulatory factor (Irf)7, Irf8, and Irf5 as well as IFN-inducible genes like Ifih1 and Ifit1. Furthermore, genes involved in oxidative stress (Nox1) and tissue remodeling (Mmp12) were also elevated (Table 1 and Supplementary Table 1). Taken together, the observed gene expression profile in the IKK2-CAPdx-1 model mimics to a great extent that detected in virus-induced immune-mediated diabetes and, importantly, points to other potential novel candidate genes for diabetes.

Figure 6

Validation of selected microarray data by real-time PCR. A: Fed and fasted blood glucose values from 11- to 13-week-old control (white circles), Pdx-1+/− (white squares), and IKK2-CAPdx-1 (white and black triangles) mice (n = 12–13/group). Black triangles represent samples used in the microarray analysis. B: Quantitative RT-PCR analysis of isolated islets from 11- to 13-week-old control, Pdx-1+/−, and IKK2-CAPdx-1. Samples used for the qPCR assays include those used in the array analysis and additional samples. Shown is the fold upregulation vs. control animals (n = 5–8/group). Actb was used as a reference gene. Results were analyzed by Student t test and presented as the mean ± SEM. *P < 0.05; **P < 0.01; ***P < 0.001. Il12b, interleukin-12b; Madcam1, mucosal addressin cell adhesion molecule-1.

Figure 6

Validation of selected microarray data by real-time PCR. A: Fed and fasted blood glucose values from 11- to 13-week-old control (white circles), Pdx-1+/− (white squares), and IKK2-CAPdx-1 (white and black triangles) mice (n = 12–13/group). Black triangles represent samples used in the microarray analysis. B: Quantitative RT-PCR analysis of isolated islets from 11- to 13-week-old control, Pdx-1+/−, and IKK2-CAPdx-1. Samples used for the qPCR assays include those used in the array analysis and additional samples. Shown is the fold upregulation vs. control animals (n = 5–8/group). Actb was used as a reference gene. Results were analyzed by Student t test and presented as the mean ± SEM. *P < 0.05; **P < 0.01; ***P < 0.001. Il12b, interleukin-12b; Madcam1, mucosal addressin cell adhesion molecule-1.

Close modal
Table 1

List of selected genes differentially regulated in islets of 11-week-old IKK2-CAPdx-1 mice as compared with Pdx-1+/− littermates

List of selected genes differentially regulated in islets of 11-week-old IKK2-CAPdx-1 mice as compared with Pdx-1+/− littermates
List of selected genes differentially regulated in islets of 11-week-old IKK2-CAPdx-1 mice as compared with Pdx-1+/− littermates

Ccl17 Expression Precedes Phenotype Development

In an attempt to identify candidate genes involved in phenotype initiation, we followed up the temporal expression pattern of distinct genes. Interestingly, Ccl17 was massively upregulated in islets of 8-week-old IKK2-CAPdx-1, when animals were asymptomatic, while there was only mild upregulation of Ear2, Serpina1a, Irf7, and Madcam1 and no elevation of MHC I/II molecules (Fig. 7A). These data support the hypothesis that Ccl17 might be a critical initial effector of IKK2/NF-κB–mediated diabetes development. In addition, qPCR analysis of highly diseased IKK2-CAPdx-1 mice (∼24 weeks old) revealed sustained upregulation of Ccl17, Ear2, and Serpina1a that was completely normalized in the reverse-remodeling experiment (Fig. 7B).

Figure 7

Gene-expression kinetics during diabetes development in IKK2-CAPdx-1 mice. A: Quantitative RT-PCR from islets isolated from 8-week-old control, Pdx-1+/−, and IKK2-CAPdx-1 mice. Shown is the fold upregulation vs. control animals (n = 4–6/group). Actb was used as a reference gene. B: Quantitative RT-PCR from pancreata of 24- to 26-week-old animals before (diseased) and after doxycycline readministration (reversed). Shown is the fold upregulation vs. control animals (in diseased state, n = 6–10/group; in reversed state, n = 4–5/group). Hprt was used as a reference gene. Results were analyzed by Student t test and presented as the mean ± SEM. C: Proposed model for IKK2/NF-κB–induced diabetes. Activation of IKK2/NF-κB in β-cells is sufficient to induce gene expression programs involved in innate immunity, type I IFN response, and antigen presentation. This may subsequently result in oxidative stress, ER stress, and nitric oxide (NO) production that finally funnels in β-cell dysfunction and death that is probably mediated by apoptosis. In addition, there was a massive increase in the chemokine Ccl17, formerly shown to be involved in autoimmunity and allergic inflammatory processes. Moreover, a direct effect of IKK2/NF-κB activation on β-cell function via unknown mechanisms may also contribute to diabetes development in this model. *P < 0.05; **P < 0.01. CHOP, C/EBP homologous protein; Madcam1, mucosal addressin cell adhesion molecule-1.

Figure 7

Gene-expression kinetics during diabetes development in IKK2-CAPdx-1 mice. A: Quantitative RT-PCR from islets isolated from 8-week-old control, Pdx-1+/−, and IKK2-CAPdx-1 mice. Shown is the fold upregulation vs. control animals (n = 4–6/group). Actb was used as a reference gene. B: Quantitative RT-PCR from pancreata of 24- to 26-week-old animals before (diseased) and after doxycycline readministration (reversed). Shown is the fold upregulation vs. control animals (in diseased state, n = 6–10/group; in reversed state, n = 4–5/group). Hprt was used as a reference gene. Results were analyzed by Student t test and presented as the mean ± SEM. C: Proposed model for IKK2/NF-κB–induced diabetes. Activation of IKK2/NF-κB in β-cells is sufficient to induce gene expression programs involved in innate immunity, type I IFN response, and antigen presentation. This may subsequently result in oxidative stress, ER stress, and nitric oxide (NO) production that finally funnels in β-cell dysfunction and death that is probably mediated by apoptosis. In addition, there was a massive increase in the chemokine Ccl17, formerly shown to be involved in autoimmunity and allergic inflammatory processes. Moreover, a direct effect of IKK2/NF-κB activation on β-cell function via unknown mechanisms may also contribute to diabetes development in this model. *P < 0.05; **P < 0.01. CHOP, C/EBP homologous protein; Madcam1, mucosal addressin cell adhesion molecule-1.

Close modal

In this study, we describe a novel mouse model of immune-mediated diabetes, the IKK2-CAPdx-1 model that is based on genetic activation of proinflammatory IKK2/NF-κB signaling, specifically in β-cells. IKK2-CAPdx-1 animals develop severe hyperglycemia and hypoinsulinemia mirroring substantial β-cell loss that is associated with marked islet inflammation. Similar to the human immunopathology, infiltrates are not seen in all pancreatic islets and include different kinds of immune cells (21). This model is the first animal model to activate IKK2/NF-κB in β-cells and to prove directly its capability to trigger diabetes development on its own. IKK2-CAPdx-1 islets show a gene expression signature reflecting the activation of innate immunity and type I IFN response, which could generate a proinflammatory microenvironment sufficient to recruit different immune cells. These infiltrating cells are able to contribute to and/or amplify the inflammatory insult, thereby promoting β-cell destruction as observed in diabetic subjects and NOD mice (1,2). Furthermore, increased expression of MHC I and II in transgenic β-cells indicates that IKK2/NF-κB signaling is capable of increasing islet antigen presentation to infiltrating T cells, which could participate in the autoimmune insult. In addition, MHC II expression by β-cells can promote their death, as seen by MHC II ligation in antigen-presenting cells (22). This may allow for an intensive dialogue between β-cells and immune cells that finally promotes β-cell destruction in a T-cell–dependent manner. The main T-cell subtype in our model is the CD4+ type, in contrast to the concept that CD8+ T cells predominate in humans (21), However, there is a considerable heterogeneity within diabetic patients (e.g., even no autoreactive T cells were found in a subset of recent-onset patients) (23). In addition, CD4+ T cells can mediate β-cell death in transgenic NOD mice (24), and diabetes development was shown to require the presence of both CD4+ and CD8+ T cells (25).

The IKK2-CAPdx-1 phenotype is in line with the reported role of NF-κB as a mediator of cytokine-induced β-cell destruction (4,5) and its activation in islets of prediabetic NOD mice (18). Consistent with this notion, resistance to streptozotocin (STZ)-induced diabetes was achieved by β-cell–specific inhibition of NF-κB (26,27), and various natural products were found to inhibit STZ-induced diabetes and protect against β-cell damage through NF-κB inhibition (4). However, β-cell–specific repression of NF-κB in normal mice elicits hyperglycemia and defective glucose-stimulated insulin secretion and accelerates diabetes development in NOD mice (4,5), which may implicate that the extent/level, context, and timing of NF-κB activation dictate the overall outcome of diabetes pathogenesis.

Our gene expression data suggest that activation of IKK2/NF-κB in β-cells is sufficient to initiate different cellular mechanisms formerly shown to affect β-cell function/survival and induce diabetes in humans and animals (16,17,2832). These diverse factors (TNF-α, Atf3, C/EBP homologous protein, Nos2, c-myc, Stat1, and Nox-1) are known to induce inflammation, oxidative stress, ER stress, and nitric oxide production (Fig. 7C) that together may finally funnel in β-cell dysfunction and/or apoptosis in our model. However, we cannot exclude other unknown effects of IKK2/NF-κB signaling that might interfere with β-cell function and promote disease development (Fig. 7C).

The expression profile in our model is further pointing to the involvement of innate immunity and type I IFN response (Fig. 7C), characteristic for viral infection, and is similarly detected in pancreata and islets from patients at clinical onset and long-standing diabetes (12). Furthermore, the gene-expression program induced by enteroviral infection of human islets (14,15) is also prominently mimicked by the IKK2-CAPdx-1 model, suggesting that IKK/NF-κB is a critical downstream effector in this context. Viral infections have been proposed as a triggering factor of type 1 diabetes in humans and animals (33), and importantly, they activate Toll-like and nucleotide-binding oligomerization domain–like receptors, which are well-known inducers of NF-κB. Indeed, NF-κB signaling is activated in β-cells by enterovirus infection (15) and double-stranded RNA treatment (34) and mediates at least partially the deleterious effects of these insults. Recently, Irf7, the master regulator of type I IFN–dependent immune responses and upregulated in our model, has been implicated in type 1 diabetes pathogenesis (35). Similarly, Ifih, another candidate gene for type 1 diabetes that is expressed in human islets and can be induced by inflammatory cytokines (13) and by double-stranded RNA in rat β-cells (36), is elevated in IKK2-CAPdx-1 mice. Therefore, it is well conceivable that NF-κB activation in β-cells on its own could create an innate immune response similar to an antiviral response, which finally culminates in diabetes development. However, we cannot exclude a possible role of Pdx-1 haploinsufficiency on promoting β-cell susceptibility to IKK2/NF-κB–induced inflammation and phenotype development.

We also identified other novel diabetes-associated candidate genes. Ccl17, the most upregulated one, is a chemokine involved in immune and allergic inflammatory responses (20,3742). The early marked upregulation of Ccl17 at normoglycemia supports the idea of being a foremost effector of IKK2/NF-κB signaling that contributes to diabetes development. Consistently, viral infection of human B cells induces NF-κB–dependent expression of Ccl17 and Ccl22 (also elevated in our model) (37), and IKK/NF-κB inhibition prevents cytokine-induced Ccl17 production in keratinocytes (38). Ccl17 is mainly produced by dendritic cells (39) and plays an important role in T cell development, trafficking, and activation. Furthermore, it was highlighted as a novel biomarker for allergic inflammatory diseases like asthma (40) and atopic dermatitis (41), in which it is strongly coexpressed with Ccl22 (42).

Both Ccl17 and Ccl22 preferentially attract CD4+ T cells via the Ccr4 receptor. Cells expressing Ccl17 were detected within infiltrated islets from prediabetic NOD mice, and Ccr4-positive T cells were shown to be critically involved in autoimmune diabetes development (20). Additionally, neutralizing Ccl22 antibodies inhibited insulitis and diabetes, whereas transgenic Ccl22 expression accelerated disease development (20). However, autoimmune diabetes was prevented by Ccl22-mediated regulatory T-cell recruitment in another study (43). Furthermore, no significant difference in plasma Ccl17 was detected in type 1 diabetes patients (44). Yet, this study was conducted in a very small cohort of diabetic Japanese subjects, which certainly does not exclude a different pattern in other patient subsets.

Ears, also prominently induced in our model, represent a subgroup of the RNase A family secreted by rodent eosinophils (45) and also expressed by macrophages. Ears participate in host defense by their antibacterial and antiviral activity together with chemotaxis to dendritic cells (45). The Serpina1 family of genes encodes for inhibitors of serine proteases. In humans, it is represented by a single gene, called α1-antitrypsin (AAT) for which expression was enhanced by proinflammatory cytokines in islet cells (46). AAT is an acute-phase protein with anti-inflammatory, tissue-protective, and antiapoptotic properties that is able to prolong islet allograft survival and to inhibit cytokine and STZ-induced β-cell apoptosis (47,48) as well as diabetes development in NOD mice (49). Although decreased functional activity of serum AAT was shown in diabetes, serum levels are variable, probably reflecting changes in the inflammatory status of the disease (50). To what extent these newly identified genes contribute to human disease pathology, however, needs further investigation.

One striking aspect of the IKK2-CAPdx-1 model is the enormous recovery potential from the diabetes status including the resolution of insulitis and the efficient regeneration of insulin-positive β-cells after transgene inactivation. A possible rationale is that a portion of β-cells is not destroyed but, rather, has lost its functional and differentiated state and therefore can regain its functionality upon IKK2-CA inactivation with the consequent resolution of proinflammatory mediators and disappearance of immune cells. Furthermore, the increase of Ki67-positive β-cells during the regeneration phase suggests that proliferation is also involved in the restoration of β-cell mass in this model as found in other models; however, other mechanisms cannot be excluded. This finding implies that the IKK/NF-κB system might be a potential target for clinical intervention. Furthermore, the IKK2-CAPdx-1 mouse model could be valuable for assessing mechanisms and identifying genes involved in β-cell regeneration as well. Since diabetes passes through relapsing-remitting onsets, our model is further potentially useful for evaluating clinical therapeutics at different stages of the human disease presentation through switching the system off and on.

In summary, the IKK2-CAPdx-1 model represents a novel conditional mouse model of immune-mediated diabetes with very distinct checkpoints of β-cell destruction and regeneration. This model phenocopies major aspects of the human disease and may represent a valuable tool for improving preclinical drug assessment for diabetes treatment.

Acknowledgments. The authors thank Ute Leschik, Melanie Gerstenlauer, and Bianca Ries (University of Ulm) for excellent technical assistance and Karlheinz Holzmann (Genomics-Core Facility, University Hospital Ulm) for performing the microarray analysis.

Funding. This work was supported by grants GRK-1041-P3 (Deutsche Forschungsgemeinschaft) and BIU-C6 (Boehringer Ingelheim Ulm University BioCenter) to B.B.

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

Author Contributions. H.H.S. and B.B. designed and performed the experiments, analyzed data, and wrote the manuscript. B.T. and K.F. performed research and analyzed data. H.J.M. contributed to discussion and reviewed and edited the manuscript. R.S. and M.W. contributed to discussion, performed the experiments, and reviewed and edited the manuscript. B.O.B. and T.W. designed research and reviewed and edited the manuscript. H.H.S. and B.B. are the guarantors of this work and, as such, had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.

1.
Bluestone
JA
,
Herold
K
,
Eisenbarth
G
.
Genetics, pathogenesis and clinical interventions in type 1 diabetes
.
Nature
2010
;
464
:
1293
1300
[PubMed]
2.
Eizirik
DL
,
Colli
ML
,
Ortis
F
.
The role of inflammation in insulitis and beta-cell loss in type 1 diabetes
.
Nat Rev Endocrinol
2009
;
5
:
219
226
[PubMed]
3.
Baker
RG
,
Hayden
MS
,
Ghosh
S
.
NF-κB, inflammation, and metabolic disease
.
Cell Metab
2011
;
13
:
11
22
[PubMed]
4.
Baumann
B
,
Salem
HH
,
Boehm
BO
.
Anti-inflammatory therapy in type 1 diabetes
.
Curr Diab Rep
2012
;
12
:
499
509
[PubMed]
5.
Zhao
Y
,
Krishnamurthy
B
,
Mollah
ZU
,
Kay
TW
,
Thomas
HE
.
NF-κB in type 1 diabetes
.
Inflamm Allergy Drug Targets
2011
;
10
:
208
217
[PubMed]
6.
Holland
AM
,
Hale
MA
,
Kagami
H
,
Hammer
RE
,
MacDonald
RJ
.
Experimental control of pancreatic development and maintenance
.
Proc Natl Acad Sci USA
2002
;
99
:
12236
12241
[PubMed]
7.
Baumann
B
,
Wagner
M
,
Aleksic
T
, et al
.
Constitutive IKK2 activation in acinar cells is sufficient to induce pancreatitis in vivo
.
J Clin Invest
2007
;
117
:
1502
1513
[PubMed]
8.
Lattke
M
,
Magnutzki
A
,
Walther
P
,
Wirth
T
,
Baumann
B
.
Nuclear factor κB activation impairs ependymal ciliogenesis and links neuroinflammation to hydrocephalus formation
.
J Neurosci
2012
;
32
:
11511
11523
[PubMed]
9.
Rajasalu
T
,
Brosi
H
,
Schuster
C
, et al
.
Deficiency in B7-H1 (PD-L1)/PD-1 coinhibition triggers pancreatic beta-cell destruction by insulin-specific, murine CD8 T-cells
.
Diabetes
2010
;
59
:
1966
1973
[PubMed]
10.
Bottazzo
GF
,
Dean
BM
,
McNally
JM
,
MacKay
EH
,
Swift
PG
,
Gamble
DR
.
In situ characterization of autoimmune phenomena and expression of HLA molecules in the pancreas in diabetic insulitis
.
N Engl J Med
1985
;
313
:
353
360
[PubMed]
11.
Sarkar
SA
,
Lee
CE
,
Victorino
F
, et al
.
Expression and regulation of chemokines in murine and human type 1 diabetes
.
Diabetes
2012
;
61
:
436
446
[PubMed]
12.
Planas
R
,
Carrillo
J
,
Sanchez
A
, et al
.
Gene expression profiles for the human pancreas and purified islets in type 1 diabetes: new findings at clinical onset and in long-standing diabetes
.
Clin Exp Immunol
2010
;
159
:
23
44
[PubMed]
13.
Eizirik
DL
,
Sammeth
M
,
Bouckenooghe
T
, et al
.
The human pancreatic islet transcriptome: expression of candidate genes for type 1 diabetes and the impact of pro-inflammatory cytokines
.
PLoS Genet
2012
;
8
:
e1002552
[PubMed]
14.
Ylipaasto
P
,
Kutlu
B
,
Rasilainen
S
, et al
.
Global profiling of coxsackievirus- and cytokine-induced gene expression in human pancreatic islets
.
Diabetologia
2005
;
48
:
1510
1522
[PubMed]
15.
Ylipaasto
P
,
Smura
T
,
Gopalacharyulu
P
, et al
.
Enterovirus-induced gene expression profile is critical for human pancreatic islet destruction
.
Diabetologia
2012
;
55
:
3273
3283
[PubMed]
16.
Cnop
M
,
Welsh
N
,
Jonas
JC
,
Jörns
A
,
Lenzen
S
,
Eizirik
DL
.
Mechanisms of pancreatic beta-cell death in type 1 and type 2 diabetes: many differences, few similarities
.
Diabetes
2005
;
54
(
Suppl. 2
):
S97
S107
[PubMed]
17.
Eizirik
DL
,
Cardozo
AK
,
Cnop
M
.
The role for endoplasmic reticulum stress in diabetes mellitus
.
Endocr Rev
2008
;
29
:
42
61
[PubMed]
18.
Tersey
SA
,
Nishiki
Y
,
Templin
AT
, et al
.
Islet β-cell endoplasmic reticulum stress precedes the onset of type 1 diabetes in the nonobese diabetic mouse model
.
Diabetes
2012
;
61
:
818
827
[PubMed]
19.
Marhfour
I
,
Lopez
XM
,
Lefkaditis
D
, et al
.
Expression of endoplasmic reticulum stress markers in the islets of patients with type 1 diabetes
.
Diabetologia
2012
;
55
:
2417
2420
[PubMed]
20.
Kim
SH
,
Cleary
MM
,
Fox
HS
,
Chantry
D
,
Sarvetnick
N
.
CCR4-bearing T cells participate in autoimmune diabetes
.
J Clin Invest
2002
;
110
:
1675
1686
[PubMed]
21.
Richardson
SJ
,
Willcox
A
,
Bone
AJ
,
Morgan
NG
,
Foulis
AK
.
Immunopathology of the human pancreas in type-I diabetes
.
Semin Immunopathol
2011
;
33
:
9
21
[PubMed]
22.
Bertho
N
,
Drénou
B
,
Laupeze
B
, et al
.
HLA-DR-mediated apoptosis susceptibility discriminates differentiation stages of dendritic/monocytic APC
.
J Immunol
2000
;
164
:
2379
2385
[PubMed]
23.
Dotta
F
,
Censini
S
,
van Halteren
AG
, et al
.
Coxsackie B4 virus infection of beta cells and natural killer cell insulitis in recent-onset type 1 diabetic patients
.
Proc Natl Acad Sci USA
2007
;
104
:
5115
5120
[PubMed]
24.
Amrani
A
,
Verdaguer
J
,
Thiessen
S
,
Bou
S
,
Santamaria
P
.
IL-1alpha, IL-1beta, and IFN-gamma mark beta cells for Fas-dependent destruction by diabetogenic CD4(+) T lymphocytes
.
J Clin Invest
2000
;
105
:
459
468
[PubMed]
25.
Phillips
JM
,
Parish
NM
,
Raine
T
, et al
.
Type 1 diabetes development requires both CD4+ and CD8+ T cells and can be reversed by non-depleting antibodies targeting both T cell populations
.
Rev Diabet Stud
2009
;
6
:
97
103
[PubMed]
26.
Eldor
R
,
Yeffet
A
,
Baum
K
, et al
.
Conditional and specific NF-kappaB blockade protects pancreatic beta cells from diabetogenic agents
.
Proc Natl Acad Sci USA
2006
;
103
:
5072
5077
[PubMed]
27.
Ueberberg
S
,
Deutschbein
T
,
Klein
HH
, et al
.
Protection from diabetes development by single-chain antibody-mediated delivery of a NF-κB inhibitor specifically to β-cells in vivo
.
Am J Physiol Endocrinol Metab
2011
;
301
:
E83
E90
[PubMed]
28.
Laybutt
DR
,
Weir
GC
,
Kaneto
H
, et al
.
Overexpression of c-Myc in beta-cells of transgenic mice causes proliferation and apoptosis, downregulation of insulin gene expression, and diabetes
.
Diabetes
2002
;
51
:
1793
1804
[PubMed]
29.
Allagnat
F
,
Fukaya
M
,
Nogueira
TC
, et al
.
C/EBP homologous protein contributes to cytokine-induced pro-inflammatory responses and apoptosis in β-cells
.
Cell Death Differ
2012
;
19
:
1836
1846
[PubMed]
30.
Moore
F
,
Naamane
N
,
Colli
ML
, et al
.
STAT1 is a master regulator of pancreatic beta-cell apoptosis and islet inflammation
.
J Biol Chem
2011
;
286
:
929
941
[PubMed]
31.
Newsholme
P
,
Morgan
D
,
Rebelato
E
, et al
.
Insights into the critical role of NADPH oxidase(s) in the normal and dysregulated pancreatic beta cell
.
Diabetologia
2009
;
52
:
2489
2498
[PubMed]
32.
Rabinovitch
A
,
Sumoski
W
,
Rajotte
RV
,
Warnock
GL
.
Cytotoxic effects of cytokines on human pancreatic islet cells in monolayer culture
.
J Clin Endocrinol Metab
1990
;
71
:
152
156
[PubMed]
33.
Hober
D
,
Sauter
P
.
Pathogenesis of type 1 diabetes mellitus: interplay between enterovirus and host
.
Nat Rev Endocrinol
2010
;
6
:
279
289
[PubMed]
34.
Liu
D
,
Cardozo
AK
,
Darville
MI
,
Eizirik
DL
.
Double-stranded RNA cooperates with interferon-gamma and IL-1 beta to induce both chemokine expression and nuclear factor-kappa B-dependent apoptosis in pancreatic beta-cells: potential mechanisms for viral-induced insulitis and beta-cell death in type 1 diabetes mellitus
.
Endocrinology
2002
;
143
:
1225
1234
[PubMed]
35.
Heinig
M
,
Petretto
E
,
Wallace
C
, et al
Cardiogenics Consortium
.
A trans-acting locus regulates an anti-viral expression network and type 1 diabetes risk
.
Nature
2010
;
467
:
460
464
[PubMed]
36.
Colli
ML
,
Moore
F
,
Gurzov
EN
,
Ortis
F
,
Eizirik
DL
.
MDA5 and PTPN2, two candidate genes for type 1 diabetes, modify pancreatic beta-cell responses to the viral by-product double-stranded RNA
.
Hum Mol Genet
2010
;
19
:
135
146
[PubMed]
37.
Nakayama
T
,
Hieshima
K
,
Nagakubo
D
, et al
.
Selective induction of Th2-attracting chemokines CCL17 and CCL22 in human B cells by latent membrane protein 1 of Epstein-Barr virus
.
J Virol
2004
;
78
:
1665
1674
[PubMed]
38.
Jung
MR
,
Lee
TH
,
Bang
MH
, et al
.
Suppression of thymus- and activation-regulated chemokine (TARC/CCL17) production by 3-O-β-D-glucopyanosylspinasterol via blocking NF-κB and STAT1 signaling pathways in TNF-α and IFN-γ-induced HaCaT keratinocytes
.
Biochem Biophys Res Commun
2012
;
427
:
236
241
[PubMed]
39.
Alferink
J
,
Lieberam
I
,
Reindl
W
, et al
.
Compartmentalized production of CCL17 in vivo: strong inducibility in peripheral dendritic cells contrasts selective absence from the spleen
.
J Exp Med
2003
;
197
:
585
599
[PubMed]
40.
Hartl
D
,
Lee
CG
,
Da Silva
CA
,
Chupp
GL
,
Elias
JA
.
Novel biomarkers in asthma: chemokines and chitinase-like proteins
.
Curr Opin Allergy Clin Immunol
2009
;
9
:
60
66
[PubMed]
41.
Saeki
H
,
Tamaki
K
.
Thymus and activation regulated chemokine (TARC)/CCL17 and skin diseases
.
J Dermatol Sci
2006
;
43
:
75
84
[PubMed]
42.
Romagnani
S
.
Cytokines and chemoattractants in allergic inflammation
.
Mol Immunol
2002
;
38
:
881
885
[PubMed]
43.
Montane
J
,
Bischoff
L
,
Soukhatcheva
G
, et al
.
Prevention of murine autoimmune diabetes by CCL22-mediated Treg recruitment to the pancreatic islets
.
J Clin Invest
2011
;
121
:
3024
3028
[PubMed]
44.
Aso
Y
,
Matsuura
H
,
Momobayashi
A
, et al
.
Profound reduction in T-helper (Th) 1 lymphocytes in peripheral blood from patients with concurrent type 1 diabetes and Graves’ disease
.
Endocr J
2006
;
53
:
377
385
[PubMed]
45.
Gupta
SK
,
Haigh
BJ
,
Griffin
FJ
,
Wheeler
TT
.
The mammalian secreted RNases: mechanisms of action in host defence
.
Innate Immun
2013
;
19
:
86
97
[PubMed]
46.
Bosco
D
,
Meda
P
,
Morel
P
, et al
.
Expression and secretion of alpha1-proteinase inhibitor are regulated by proinflammatory cytokines in human pancreatic islet cells
.
Diabetologia
2005
;
48
:
1523
1533
[PubMed]
47.
Koulmanda
M
,
Bhasin
M
,
Fan
Z
, et al
.
Alpha 1-antitrypsin reduces inflammation and enhances mouse pancreatic islet transplant survival
.
Proc Natl Acad Sci USA
2012
;
109
:
15443
15448
[PubMed]
48.
Zhang
B
,
Lu
Y
,
Campbell-Thompson
M
, et al
.
Alpha1-antitrypsin protects beta-cells from apoptosis
.
Diabetes
2007
;
56
:
1316
1323
[PubMed]
49.
Koulmanda
M
,
Bhasin
M
,
Hoffman
L
, et al
.
Curative and beta cell regenerative effects of alpha1-antitrypsin treatment in autoimmune diabetic NOD mice
.
Proc Natl Acad Sci USA
2008
;
105
:
16242
16247
[PubMed]
50.
Lisowska-Myjak
B
.
AAT as a diagnostic tool
.
Clin Chim Acta
2005
;
352
:
1
13
[PubMed]
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. See http://creativecommons.org/licenses/by-nc-nd/3.0/ for details.

Supplementary data