Endoplasmic reticulum stress–mediated apoptosis may play an important role in the destruction of pancreatic β-cells, thus contributing to the development of type 1 and type 2 diabetes. One of the regulators of endoplasmic reticulum stress–mediated cell death is the CCAAT/enhancer binding protein (C/EBP) homologous protein (Chop). We presently studied the molecular regulation of Chop expression in insulin-producing cells (INS-1E) in response to three pro-apoptotic and endoplasmic reticulum stress–inducing agents, namely the cytokines interleukin-1β + interferon-γ, the free fatty acid palmitate, and the sarcoendoplasmic reticulum pump Ca2+ ATPase blocker cyclopiazonic acid (CPA). Detailed mutagenesis studies of the Chop promoter showed differential regulation of Chop transcription by CPA, cytokines, and palmitate. Whereas palmitate- and cytokine-induced Chop expression was mediated via a C/EBP–activating transcription factor (ATF) composite and AP-1 binding sites, CPA induction required the C/EBP-ATF site and the endoplasmic reticulum stress response element. Cytokines, palmitate, and CPA induced eIF2α phosphorylation in INS-1E cells leading to activation of the transcription factor ATF4. Chop transcription in response to cytokines and palmitate depends on the binding of ATF4 and AP-1 to the Chop promoter, but distinct AP-1 dimers were formed by cytokines and palmitate. These results suggest a differential response of β-cells to diverse endoplasmic reticulum stress inducers, leading to a differential regulation of Chop transcription.

Because of their marked insulin secretory activity, pancreatic β-cells have a highly developed endoplasmic reticulum and are particularly susceptible to endoplasmic reticulum stress (13). Endoplasmic reticulum stress is a cellular response triggered by disruption of the endoplasmic reticulum homeostasis and consequent accumulation of misfolded proteins in its lumen. To restore normal endoplasmic reticulum function, the cells activate a cytoprotective mechanism known as the unfolded protein response (UPR) (4), which causes 1) activation of key transcription factors, such as X-box binding protein-1 and activating transcription factor 6 (ATF6), which together increase the expression of endoplasmic reticulum–resident chaperones; and 2) phosphorylation of the eukaryotic initiation factor (eIF2α) by the kinase PKR-like endoplasmic reticulum kinase (PERK), leading to translational attenuation and activation of the transcription factor ATF4 (4). If the UPR fails to restore normal endoplasmic reticulum function, apoptosis will be triggered. To date, several proteins involved in endoplasmic reticulum stress–induced apoptosis have been identified, including the CCAAT/enhancer binding protein (C/EBP) homologous transcription factor (Chop), the Bcl-2 family members (Bak/Bax), caspase-12, and c-Jun NH2-terminal kinase (JNK) (4).

Endoplasmic reticulum stress seems to contribute to β-cell apoptosis in both type 1 and type 2 diabetes (59). During insulitis in type 1 diabetes, β-cells are exposed to cytokines such as interleukin (IL)-1β, interferon (IFN)-γ, and tumor necrosis factor-α and free radicals such as nitric oxide (NO) (10,11). IL-1β + IFN-γ, via NO production, inhibit expression of the sarcoendoplasmic reticulum pump Ca2+ ATPase (SERCA) 2 and deplete endoplasmic reticulum calcium stores in β-cells, leading to endoplasmic reticulum stress and apoptosis (5). The endoplasmic reticulum stress response induced by cytokines in β-cells is characterized by inositol-requiring endoplasmic reticulum–to–nucleus signal kinase (IRE)-1α activation, Chop, and ATF4 induction (5). The chemical SERCA blocker cyclopiazonic acid (CPA) also induces the endoplasmic reticulum stress response in β-cells, activating all components of the UPR, Chop expression, and apoptosis (12). IFN-γ aggravates endoplasmic reticulum stress, enhancing Chop expression and apoptosis via inhibition of protective endoplasmic reticulum chaperones (12).

The pathogenesis of type 2 diabetes is characterized by both insulin resistance and impaired insulin secretion due to pancreatic β-cell dysfunction and decreased β-cell mass, secondary to increased rates of β-cell apoptosis (13,14). High levels of circulating free fatty acids (FFAs) may contribute to β-cell dysfunction and death (11,13), and the FFA palmitate triggers endoplasmic reticulum stress and induces apoptosis in β-cells (6,7). Palmitate activates the IRE-1α and PERK pathways, leading to the induction of Chop, ATF4, and Bip (6,7).

Chop is a transcription factor of the C/EBP family (15). Its basal expression is low under nonstressed conditions, but it increases markedly in response to a variety of physiological and pharmacological stresses, including conditions that alter protein folding in the endoplasmic reticulum (15,16). Chop has a dual role, acting either as a dominant-negative inhibitor of C/EBPs or as a gene activator (17,18). It can enhance the transcriptional activation of AP-1 by tethering to the AP-1 complex without direct binding to DNA (19). Several studies point to a pro-apoptotic effect of Chop downstream of irremediable endoplasmic reticulum stress (15). Possible mechanisms for Chop-induced apoptosis include 1) translocation of Bax from the cytosol to the mitochondria (20); 2) downregulation of Bcl-2 expression and perturbation the cellular redox state by depletion of cellular glutathione (21); 3) upregulation of the death receptor 5 (22); and 4) interaction with other proteins that remain to be identified (15). Chop can mediate pancreatic β-cell death as suggested by 1) resistance of Chop knockout islet to the toxic effects of chemical NO donors (9) and 2) delayed diabetes onset in Akita mice bearing a homozygous deletion of the Chop gene (23). Regulation of Chop expression is cell and stimulus dependent (2428), which consequently impacts on the outcome of endoplasmic reticulum stress.

Against this background, we presently studied the transcriptional regulation of the Chop gene in INS-1E cells after exposure to different endoplasmic reticulum stress inducers that may contribute to type 1 (cytokines) or type 2 (FFAs) diabetes. For comparative purposes and as a positive control, INS-1E cells were exposed to the reversible SERCA blocker CPA.

Cell culture treatments and NO measurements.

Insulin-producing INS-1E cells were cultured in RPMI-1640 medium (Invitrogen, Paisley, U.K.) supplemented with 5% FCS (29,30). Recombinant rat IFN-γ (R&D Systems, Abingdon, U.K.) was used at 0.18 μg/ml (corresponding to 500 units/ml IFN-γ) and human recombinant IL-1β (gift from Dr. C.W. Reinolds, National Cancer Institute, Bethesda, MD), at 10 units/ml. The inducible nitric oxide synthase (iNOS) blocker aminoguanidine (Sigma, Steinheim, Germany) was used at 2.5 mmol/l, and culture medium was collected for nitrite determination (nitrite is a stable product of NO oxidation) using the Griess method (31). Palmitate (Sigma) was solubilized in ethanol and used at a concentration of 0.5 mmol/l. For exposure to palmitate, RPMI-1640 medium containing 1% BSA and 1% FCS was used (6). CPA (Sigma) was dissolved in DMSO and used at a concentration of 25 μmol/l. The respective control conditions were for cytokines, medium alone; for palmitate, 1% ethanol; and for CPA, 0.25% DMSO. These concentrations of ethanol or DMSO do not affect β-cell viability or gene expression (6,12). Cytokines, aminoguanidine, palmitate, and CPA concentrations were selected based on our previous time course and dose-response studies (data not shown) (6,12,30). The presently used INS-1E cells have a well-preserved insulin release in response to glucose (29) and respond to cytokines (5,30), palmitate (6), and CPA (5,12) in a similar way as primary β-cells.

Real-time RT-PCR analysis.

INS-1E cells, treated as described in the figures, were harvested for mRNA isolation and subsequent reverse transcription (32). Chop mRNA expression was determined by real-time RT-PCR using SYBR Green fluorescence on a LightCycler instrument (Roche, Manheim, Germany) using the standard curve method (6). Glyceraldehyde-3-phosphate dehydrogenase (GAPDH), whose expression is not modified under the present experimental conditions (5,6,12,30) (data not shown), was used as a housekeeping gene. Results are shown as the expression of the gene of interest divided by GAPDH. Primer sequences for Chop and GAPDH are described elsewhere (5). For RNA stability experiments, INS-1E cells were exposed for 16 h to IL-1β + IFN-γ or palmitate to induce Chop mRNA expression. Medium was then replaced and new RNA formation was blocked with actinomycin D (4 ng/ml); culture was then continued for an extra 240 min with actinomycin D in the presence or absence of cytokines or palmitate.

DNA reporter constructs, transfection, and luciferase assays.

The following hamster Chop promoter constructs tethered to a luciferase reporter gene were used 1) wild type (−782 to 21 bp); 2) mutants in which the 5′ 345, 553, and 719 bp of the promoter were selectively deleted; and 3) mutants for C/EBP-ATF site and endoplasmic reticulum stress element (ERSE). Oligonucleotide reporter constructs containing two copies of the Chop C/EBP-ATF composite site or mutants at either the C/EBP or ATF portion of the site were also tested. These constructs, originally cloned in the JymCATO plasmid (27), were extracted by enzymatic digestion as follows 1) wild-type and 5′ deletion mutant of the Chop promoter as well as C/EBP-ATF oligonucleotide reporter construct were cut with SmaI and HindIII and 2) C/EBP-ATF and ERSE mutant were cut with NcoI followed by blunt-end formation using Klenow enzyme and finally digested with HindIII. The fragments obtained were subcloned between the ClaI and HindIII sites of the pGL3 basic luciferase construct (Promega, Madison, WI). The substitution mutation of the AP-1 site was made using the QuickChange II site-directed mutagenesis kit (Stratagene, La Jolla, CA). The AP-1 site TGACTCA was changed to gGACTCA using as template the wild-type and C/EBP-ATF mutant Chop promoter constructs. A construct containing the Chop promoter plus the first exon of the Chop gene was generated by PCR amplification of the exon (−13 to +121 bp) using the following primers: 5′-CGCTGCAGAGGTCAGTAGCCTGAGTCTCACACTTC-3′ (forward) and 5′-CTGAAGCTTCACTTCCTTCTGGAACACTCGCTCC-3′ (reverse). A HindIII restriction site was added at the 5′ extremity of the reverse primer (underlined sequence), and the PCR product was then cloned in the Chop promoter luciferase construct between the PstI and HindIII sites. INS-1E cells were transfected using lipofectamine reagent (Invitrogen) with 250 ng of the different luciferase reporter constructs and the pRL-CMV plasmid (50 ng, with Renilla used as internal control for transfection efficiency) (33). Twenty-four hours after transfection, the cells were exposed for 36 h to IL-1β + IFN-γ or palmitate or for 15 h to CPA. Luciferase activities of cell lysates were then determined and expressed as Firefly/Renilla (relative luciferase activity).

Electrophoretic mobility shift assay.

For the C/EBP-ATF electrophoretic mobility shift assays (EMSAs), 10 μg nuclear extracts were prepared and electrophoresed on 4% Tris glycine EDTA gels as described previously (27). For the AP-1 EMSAs 5 μg nuclear extracts were prepared and electrophoresed in 5% Tris borate EDTA gels (34). For supershift analysis, 2 μg BSA or antibody (ATF4 [sc-200], C/EBP-β [sc-150], c-Fos [sc-52], c-Jun [sc-45], or Jun-B [sc-73]; Santa Cruz Biotechnologies, Santa Cruz, CA) was used. The double-stranded synthetic oligonucleotides probes (C/EBP-ATF, 5′-AGAAACATTGCATCACCC-3′; and AP-1, 5′-AGCGCGCATGACTCACTCAAAT-3′) were labeled using Klenow enzyme and [γ32P]CTP. Specificity was assessed using nonradioactive wild-type or mutant oligonucleotides (C/EBP-ATF mutant, 5′-AGAAAcgcgtcccTCACCC-3′; or AP-1 mutant, 5′-AGCGCGCAgGACTCACTCAAAT-3′).

Western blot analysis.

Cells were lysed with either whole-cell extract buffer (1% NP40, 25 mmol/l HEPES, pH 7.8, 50 mmol/l KCl2, 10 mmol/l NaF, 1 mmol/l Na3VO4, 1 mmol/l phenylmethylsulfonyl fluoride [PMSF], and protease inhibitor cocktail) or phospho–whole-cell extract buffer (1% NP40, 1 mmol/l CaCl2, 1 mmol/l MgCl2, 150 mmol/l NaCl, 10 mmol/l NaF, 1 mmol/l Na3VO4, 1 mmol/l PMSF, and protease inhibitor cocktail [Roche]). Lysates were then subjected to a 10% SDS-PAGE and transferred to a nitrocellulose membrane. Immunoblot analysis was performed with the indicated concentrations of antibodies against Chop (G6916; 4.8 μg/ml; for the cytokines or CPA-treated experiments) from Sigma; ATF4 (sc-200; 0.8 μg/ml), eIF2α (sc-11386; 0.2 μg/ml), and Chop (sc-575; 0.3 μg/ml; for the palmitate Western blot) from Santa Cruz; and phospho-eIF2α (9721; Ser51; 16.7 ng/ml), phospho–c-Jun (9261; Ser63; 9 ng/ml), and β-actin (4967; 8 ng/ml) from Cell Signaling (Danvers, MA). A secondary anti-rabbit horseradish peroxidase–labeled IgG antibody (Santa Cruz Biotechnologies) was used for detection. Western blot pictures were quantified, and values of the protein of interest were corrected for β-actin or total eIF2α considering the maximum value in each experiment as 1.

Statistical analysis.

Data are shown as means ± SE, and comparisons between groups were made by paired t test or by ANOVA followed by t test with the Bonferroni correction, as indicated.

Chop mRNA expression and stability.

Twenty-four–and 36-h treatment of INS-1E cells with cytokines led to a 30- and 15-fold increase, respectively, of Chop mRNA expression (Fig. 1A), whereas palmitate induced a 6- and 2-fold upregulation of Chop transcript, respectively (Fig. 1B). A 22-fold induction of Chop was observed after 15 h of exposure to CPA (Fig. 1C). Western blot time course experiments were also performed to analyze Chop protein induction by cytokines, palmitate, or CPA, indicating clear upregulation in protein expression after 8–16 h (Supplementary Fig. S1 in the online appendix [available at http://dx.doi.org/10.2337/db06-1253]). INS-1E cells were also exposed to cytokines with or without the iNOS blocker aminoguanidine. Aminoguanidine did not affect basal NO production, but it prevented to a large extent cytokine-induced NO formation (Supplementary Fig. S2). Aminoguanidine completely blocked cytokine-induced Chop mRNA expression (Fig. 1A), confirming that the effect of cytokines is mediated via NO formation. Because neither palmitate nor CPA trigger NO production in INS-1E cells (5,6), we did not study the role of NO in Chop induction by these agents. Neither cytokines (Fig. 1D) nor palmitate (Fig. 1E) increased Chop mRNA stability, and after 4 h of actinomycin exposure, Chop mRNA expression was similar in both treated and untreated cells. Thus, Chop mRNA induction by cytokines or palmitate in INS-1E cells results mainly from transcriptional activation.

Different endoplasmic reticulum stress stimuli induce distinct usage of cis-acting elements of the Chop promoter.

To identify the regions that are responsible for the activation of the Chop promoter by cytokines/NO, palmitate, or CPA, we performed transient transfections with luciferase reporter constructs containing progressive 5′ deletions of the Chop promoter (Fig. 2A). There was a basal Chop activity with the −782-bp wild-type promoter that increased two- to threefold after cytokine (Fig. 2B), palmitate (Fig. 2C), or CPA (Fig. 2D) exposure. As observed for Chop mRNA expression (Fig. 1A), activation of the Chop promoter by cytokines was abolished by aminoguanidine (Fig. 2B). Aminoguanidine alone did not modify Chop promoter activity (Supplementary Fig. S3).

It was previously described that the first exon of the Chop gene contains regulatory elements (28). Hence, we prepared a luciferase reporter construct containing the −782 bp of the hamster Chop promoter plus the first exon (120 bp). We did not observe, however, any effect of exon 1 in the induction by IL-1β + IFN-γ or palmitate (Supplementary Fig. S4).

Removal of nucleotides −782 to −437 bp (deletion 1) had no effect in Chop induction by the tested agents (Fig. 2), but deletion of nucleotides −437 to −229 bp (deletion 2) led to a decrease in basal activity (Fig. 2), a partial impairment of CPA induction (Fig. 2D), and abolished induction of the Chop promoter by cytokines (Fig. 2B) and palmitate (Fig. 2C). Deletion of downstream nucleotides −229 to −63 bp (deletion 3) further decreased basal activity (Fig. 2) and completely abolished CPA induction of the Chop promoter (Fig. 2D). Based on these data, we identified one promoter region (−437 to −229 bp) necessary for CPA, cytokine, and palmitate induction of the Chop promoter and another region (−229 to −63 bp) that is mostly involved in induction by CPA. The first region contains a C/EBP-ATF composite and an AP-1 binding site, whereas the second contains an ERSE (Fig. 2A) (24,27). Mutation of the C/EBP-ATF composite site (see scheme on Fig. 3A) decreased basal promoter activity and Chop promoter induction by cytokines (Fig. 3B), palmitate (Fig. 3C), or CPA (Fig. 3D). Mutation of the AP-1 site (Fig. 3A) led to a small increase in basal promoter activity and impaired Chop induction by cytokines (Fig. 3B) and palmitate (Fig. 3C) but had no effect of on promoter induction by CPA (Fig. 3D). A double mutant for both C/EBP-ATF and AP-1 binding sites (Fig. 3A) abolished induction of luciferase activity by cytokines (Fig. 3B) or palmitate (Fig. 3C). Deletion of the ERSE binding site (Fig. 3A) specifically impaired induction by CPA (Fig. 3D), whereas double mutation of C/EBP-ATF site and ERSE greatly decreased CPA-induced promoter activity (Fig. 3D). These results suggest that CPA induces the Chop promoter mainly via the C/EBP-ATF and ERSE binding sites but not through the AP-1 site. Mutation of the ERSE site did not affect induction of Chop promoter activity by cytokines or palmitate (Fig. 3B and C).

Cytokines and palmitate trigger eIF2α phosphorylation and induce the transcription factor ATF4.

In the C/EBP-ATF composite site, the C/EBP (5′-CATTGC) and ATF (5′-ATCATC) binding sequences are present in tandem. To further investigate the role of these two binding sites, we used luciferase reporter constructs containing two copies of the C/EBP-ATF composite site or mutants at either the C/EBP or ATF sites (Fig. 4A). The wild-type construct was induced around twofold in response to cytokines (Fig. 4B) or palmitate (Fig. 4C). Mutations of either the C/EBP or ATF portions of the site severely decreased basal promoter activity (Fig. 4B and C). Mutation of the ATF site, but not of the C/EBP site, inhibited activation of the construct by cytokines (Fig. 4B) or palmitate (Fig. 4C). These results suggest that the ATF portion of the C/EBP-ATF composite site is involved in Chop induction and point to ATF4 as an important transcription factor for Chop activation. Because induction of ATF4 is secondary to eIF2α phosphorylation (4), we evaluated eIF2α phosphorylation and ATF4 induction by cytokines, palmitate, or CPA. Phosphorylation of eIF2α in response to IL-1β + IFN-γ was already observed after 4 h and remained present up to 24 h (Fig. 4D). The induction of ATF4 by cytokines was detected after 16 h of exposure and was still present at 24 h (Fig. 4D). Blocking cytokine-induced NO production by aminoguanidine prevented eIF2α phosphorylation and decreased ATF4 and Chop protein induction (Fig. 4E). Palmitate induced a prolonged (up to 24 h) upregulation in eIF2α phosphorylation and ATF4 expression, already detectable after 4 h and increasing further in the subsequent hours (Fig. 4F). CPA induced an early eIF2α phosphorylation and ATF4 induction that remained detectable until 24 h (Fig. 4G).

Cytokines and palmitate induce an ATF4-containing complex that binds to the C/EBP-ATF composite site.

To identify the cytokine- and palmitate-induced protein complexes binding to the C/EBP-ATF composite site of the Chop promoter, EMSA was performed using a probe containing the wild-type C/EBP-ATF site. Under control conditions, two different complexes were identified (Fig. 5, complexes α and β). Competition assays with 20-fold excess cold wild-type or mutated probes demonstrated the specificity of these complexes (Fig. 5A, lanes11–14, and D, lanes 11 and 12). Complex β was induced in response to cytokines or palmitate after 16 and 24 h of exposure (Fig. 5A and D, lanes 8 and 10). Blocking NO production with aminoguanidine prevented cytokine-mediated induction of complex β (Fig. 5B, compare lanes 2 and 4). An ATF4 antibody completely supershifted the cytokine- and palmitate-induced complex β (Fig. 5C and E, lane 4). On the other hand, the C/EBPβ antibody partially supershifted the constitutive complex α but did not affect complex β (Fig. 5C and E, lanes 5 and 6).

Cytokines and palmitate induce different protein complexes binding to the AP-1 site of the Chop promoter.

Both cytokines and palmitate induced a specific complex that binds to the radiolabeled probe containing the AP-1 binding site (Fig. 6A and C). This complex is barely detectable under control conditions but is induced after 4 h of cytokine or palmitate exposure (Fig. 6A and C). In cytokine- or palmitate-treated cells, there was a maximum AP-1 binding at 8 and 4–8 h, respectively, that remained detectable up to 24 h (Fig. 6A and C). Specificity of the complex was confirmed by competition assays using a 20-fold excess of cold wild-type or mutant AP-1 cold probes (Fig. 6A, lanes 11 and 12, and C, lanes 11–14). Cytokine-induced formation of the AP-1 complex was mildly reduced (∼20–30%) when the cells were treated with cytokines plus aminoguanidine (Fig. 6B). In INS-1E cells treated with either cytokines or palmitate, the c-Fos antibody induced a shift in the AP-1 complex (Fig. 7A and B, lane 4). The c-Jun antibody induced a shift in cells treated with cytokines (Fig. 7A, lane 10) but not in cells treated with palmitate (Fig. 7B, lane 8). As observed in Fig. 7A (lane 8) and B (lane 10), Jun-B antibody shifted both the cytokine- and palmitate-induced AP-1 complexes. To confirm the activation of c-Jun by cytokines, we examined c-Jun phosphorylation on serine 63 (Fig. 7C). Cytokines induced c-Jun phosphorylation already after 4 h of exposure, with a peak at 8 h and return to basal levels by 24 h. In agreement with the supershift experiments (Fig. 7B), palmitate did not trigger c-Jun phosphorylation in INS-1E cells (data not shown). These results indicate that cytokines induce AP-1 dimers composed of c-Fos, c-Jun, and/or Jun-B, whereas palmitate induces AP-1 dimers composed preferentially of c-Fos and Jun-B.

Several reports have shown that Chop may be induced at the transcriptional and/or posttranscriptional level by a variety of agents that cause cellular stress (15). The elements controlling Chop transcription vary depending on the cell type and the kind of stress induced (2428). This report provides a comprehensive analysis of the regulation of Chop mRNA expression in β-cells by different pro-apoptotic endoplasmic reticulum stressors, namely a chemical (CPA) and two physiological endoplasmic reticulum stress inducers (cytokines and palmitate).

In agreement with previous experiments (5,6,12), we observed that Chop mRNA expression is induced in insulin-producing cells by IL-1β + IFN-γ (via NO formation), palmitate, and CPA. This increase is due to augmented gene transcription, because both cytokines and palmitate increased activity of the Chop promoter without modifying Chop mRNA stability. We identified the C/EBP-ATF composite and AP-1 sites as key regulatory elements for Chop induction in response to cytokines/NO and palmitate. Differently, Chop regulation by the chemical endoplasmic reticulum stress inducer CPA involves the C/EBP-ATF and ERSE binding sites but not the AP-1 site. The SERCA blocker thapsigargin was previously shown to regulate Chop mRNA transcription in fibroblasts in a similar fashion (27). By using a luciferase reporter construct containing wild-type and mutated forms of the C/EBP-ATF composite site, we identified the ATF portion of the site as responsible for both cytokine and palmitate induction. In agreement with this, EMSA identified ATF4 as the main transcription factor of the inducible complex activated by both agents. In endoplasmic reticulum–stressed fibroblasts, two inducible complexes involving ATF4 were observed: one formed by ATF4 only and another containing both ATF4 and C/EBPβ transcription factors (27). At variance with these observations, we did not observe C/EBPβ in the complexes induced by cytokines or palmitate: C/EBPβ was only involved in a constitutive complex (Fig. 5, complex α). There was a good correlation between the kinetics of ATF4 protein induction and binding to the C/EBP-ATF site in response to cytokines, but we observed a discrepancy between the early ATF4 protein induction and its late binding to the C/EBP-ATF site in response to palmitate. This suggests that another transcription factor besides C/EBPβ is necessary for the binding of ATF4 to the C/EBP-ATF site. One possible candidate is ATF2, which binds to this site in amino acid–starved Hela cells (35). Another possibility is that posttranslational regulation of ATF4, such as phosphorylation (36), contributes to this time lag.

Induction of ATF4 protein occurs downstream of eIF2α phosphorylation (4,37). We have presently shown that cytokines (Fig. 4D), palmitate (Fig. 4F), and CPA (Fig. 4G) induce eIF2α phosphorylation in INS-1E cells. Four kinases, namely PERK, general control nonderepressible-2, heme-regulated inhibitor, and double-stranded RNA–induced protein kinase can phosphorylate eIF2α protein (37), but PERK is the main mediator of this effect during endoplasmic reticulum stress. Palmitate activates PERK kinase leading to eIF2α phosphorylation and ATF4 induction (7). Of note, cytokine-induced eIF2α phosphorylation (present data) may explain the early decrease in (pro)insulin and total protein biosynthesis in β-cells exposed to IL-1β + IFN-γ (38).

Analysis of the AP-1 site by EMSA revealed an early binding (4 h) induced by both cytokines and palmitate; this was maintained up to 24 h but at a reduced level. Supershift experiments indicated that the AP-1 dimer binding to the Chop promoter after palmitate exposure is composed of Jun-B/c-Fos, whereas cytokine exposure induces Jun-B/c-Fos and c-Jun/c-Fos in INS-1E cells. Jun-B/Jun-B and Jun-B/c-Jun are mostly transcription repressors, and c-Jun/c-Jun homodimers bind weakly to DNA (39). IL-1β induces a rapid (30 min) increase in c-Jun and c-Fos expression peaking at 3–5 h (4042); this peak coincides with the AP-1 binding to the Chop promoter observed in the present experiments. The transactivating potential of c-Jun is regulated both at the level of expression and by posttranslational modifications (39), such as phosphorylation of c-Jun on Ser-63 and Ser-73 by JNK, which enhances its transactivating activity (43). We observed that cytokines, but not palmitate, induce Ser-63 phosphorylation of c-Jun in INS-1E cells. This lack of palmitate effects on Ser-63 phosphorylation correlates with the lack of c-Jun supershift. Phosphorylation of c-Jun at Ser-63 is pro-apoptotic in neuroblastoma cells (44), and IL-1β–induced JNK activation contributes to β-cell death (45,46). During prolonged endoplasmic reticulum stress, IRE1 activates JNK via TRAF2 (47,48). Cytokine-induced JNK activation in INS-1E cells, however, is an early (1 h) and NO-independent event, whereas the endoplasmic reticulum stress response induced by cytokines in these cells is an NO-dependent and late (>4–6 h) phenomenon (5). Additional studies are required to clarify the role of the JNK–AP-1 pathway in cytokine-induced β-cell apoptosis.

Cytokines, via NO production, induce a progressive increase of Chop transcript in INS-1E cells, starting at 8 h and peaking at 24 h (5). Chop mRNA induction by palmitate increases later, ∼12 h (4.9 ± 1.1–fold increase vs. control, n = 4), with peak at 24 h (6; present data). Both cytokines and palmitate induce AP-1 binding at early time points (starting at 4 h), whereas binding of ATF4 to the C/EBP-ATF occurs later (16 h). Based on the present findings, Fig. 8 provides an overview of our current understanding of Chop induction in insulin-producing cells. The early induction of Chop mRNA by cytokines is mediated by AP-1 transcription factors, whose effects are enhanced by the subsequent binding of ATF4 to C/EBP-ATF site. The early binding of AP-1 to the Chop promoter is NO independent, whereas the subsequent cytokine induction of ATF4 and its binding to the C/EBP-ATF binding site is NO dependent. In the case of palmitate, occupation of both AP-1 and C/EBP-ATF sites are necessary for promoter activation.

In conclusion, we show that both AP-1 and ATF4 transcription factors are involved in Chop induction in INS-1E cells in response to cytokines and palmitate and that at variance with CPA treatment, neither of these agents uses the ERSE. Although ATF4 is probably activated via the UPR/PERK pathway for both cytokines and palmitate, induction of AP-1 by cytokines seems to be endoplasmic reticulum stress independent (Fig. 8).

It is puzzling that a branch of the UPR, namely the PERK/ATF4 pathway, usually considered as a β-cell survival mechanism (4) also induces the pro-apoptotic transcription factor Chop. Chop action occurs essentially via nuclear translocation and subsequent regulation of target genes (15,1719). Chop cannot homodimerize (18), suggesting that a lack of suitable partners in the nucleus might prevent its effects. Posttranslational modification of Chop can also affect its action, because phosphorylation by p38 kinase enhances Chop transcriptional activity (49), whereas phosphorylation by Casein kinase-2 has an inhibitory effect (50). Additional studies are thus required to clarify the activation and the role of Chop in endoplasmic reticulum stress–induced β-cell apoptosis.

The present observations indicate similarities but also subtle differences in the way endoplasmic reticulum stress caused by different agents activates Chop in β-cells. Further understanding of the signal transduction of cytokines and palmitate will hopefully allow the development of specific maneuvers to modulate the pro-apoptotic components of the UPR response while boosting the UPR “defense” components.

FIG. 1.

Induction of Chop mRNA by different endoplasmic reticulum stressors in INS-1E cells. A–C: Real-time PCR for Chop mRNA. INS-1E cells were incubated with IL-1β (10 units/ml) + IFN-γ (0.18 μg/ml) (Ctk) in the presence or absence of aminoguanidine (AG) (2.5 mmol/l) (A); palmitate (0.5 mmol/l) (B), CPA (25 μmol/l) (C); or respective controls (white bars). Data shown are means of four to six experiments, corrected for GAPDH expression. **P < 0.01 and ***P < 0.001 vs. control; ###P < 0.001 vs. Ctk. D–E: Analysis of Chop mRNA stability. INS-1E cells were exposed for 15 h to IL-1β (10 units/ml) + IFN-γ (0.18 μg/ml) (D) or palmitate (0.5 mmol/l) (E). The cells were then washed and treated with actinomycin D (4 ng/ml) in the presence (full line) or absence (dotted line) of cytokines (D) or palmitate (E) for up to 240 min. Chop per GAPDH mRNA expression is expressed relative to the expression level at time 0. The graphs are representative of two to three similar experiments.

FIG. 1.

Induction of Chop mRNA by different endoplasmic reticulum stressors in INS-1E cells. A–C: Real-time PCR for Chop mRNA. INS-1E cells were incubated with IL-1β (10 units/ml) + IFN-γ (0.18 μg/ml) (Ctk) in the presence or absence of aminoguanidine (AG) (2.5 mmol/l) (A); palmitate (0.5 mmol/l) (B), CPA (25 μmol/l) (C); or respective controls (white bars). Data shown are means of four to six experiments, corrected for GAPDH expression. **P < 0.01 and ***P < 0.001 vs. control; ###P < 0.001 vs. Ctk. D–E: Analysis of Chop mRNA stability. INS-1E cells were exposed for 15 h to IL-1β (10 units/ml) + IFN-γ (0.18 μg/ml) (D) or palmitate (0.5 mmol/l) (E). The cells were then washed and treated with actinomycin D (4 ng/ml) in the presence (full line) or absence (dotted line) of cytokines (D) or palmitate (E) for up to 240 min. Chop per GAPDH mRNA expression is expressed relative to the expression level at time 0. The graphs are representative of two to three similar experiments.

Close modal
FIG. 2.

Identification of the promoter regions responsible for Chop transcription in INS-1E cells after exposure to cytokines, palmitate, or CPA. A: Schematic representation of the deletions (Del.) performed in the Chop promoter. Potentially important binding sites for C/EBP-ATF (○), AP-1 (▵), and ERSE (□) are indicated. INS-1E cells were transfected with wild-type or deletion mutant Chop promoter luciferase reporter plasmids and the internal control pRL-CMV. Cells were treated or not (control, white bars) with IL-1β (10 units/ml) + IFN-γ (0.18 μg/ml) (black bars) in the presence of absence of aminoguanidine (2.5 mmol/l, striped bars) for 36 h (B), palmitate (0.5 mmol/l, hatched bars) for 36 h (C), or CPA for 15 h (25 μmol/l, dotted bars) (D). Left panels represent relative luciferase activity (RLU). **P < 0.01 and ***P < 0.001 vs. respective control; ‡‡‡P < 0.001 vs. wild-type control. Right panels represent fold variation of luciferase activity for each construct relative to its control. ##P < 0.01 and ###P < 0.001 vs. wild type. Results are means ± SE of 6–16 experiments.

FIG. 2.

Identification of the promoter regions responsible for Chop transcription in INS-1E cells after exposure to cytokines, palmitate, or CPA. A: Schematic representation of the deletions (Del.) performed in the Chop promoter. Potentially important binding sites for C/EBP-ATF (○), AP-1 (▵), and ERSE (□) are indicated. INS-1E cells were transfected with wild-type or deletion mutant Chop promoter luciferase reporter plasmids and the internal control pRL-CMV. Cells were treated or not (control, white bars) with IL-1β (10 units/ml) + IFN-γ (0.18 μg/ml) (black bars) in the presence of absence of aminoguanidine (2.5 mmol/l, striped bars) for 36 h (B), palmitate (0.5 mmol/l, hatched bars) for 36 h (C), or CPA for 15 h (25 μmol/l, dotted bars) (D). Left panels represent relative luciferase activity (RLU). **P < 0.01 and ***P < 0.001 vs. respective control; ‡‡‡P < 0.001 vs. wild-type control. Right panels represent fold variation of luciferase activity for each construct relative to its control. ##P < 0.01 and ###P < 0.001 vs. wild type. Results are means ± SE of 6–16 experiments.

Close modal
FIG. 3.

Differential usage of cis-acting elements in the Chop promoter by cytokines, palmitate, or CPA. A: Schematic representation of the mutations performed in the Chop promoter (×, substitution; ▪, deletion): C/EBP-ATF (○), AP-1 (▵), and ERSE (□) sites. INS-1E cells were transfected with wild-type or mutant Chop promoter constructs shown in A and treated with IL-1β (10 units/ml) + IFN-γ (0.18 μg/ml) (black bars) for 36 h (B); palmitate (0.5 mmol/l, hatched bars) for 36 h (C); CPA (25 μmol/l, dotted bars) for 15 h (D); or respective controls (white bars). Left panels represent the relative luciferase activity (RLU) of 4–16 experiments. *P < 0.05, **P < 0.01, and ***P < 0.001 vs. respective control; ‡P < 0.05, ‡‡P < 0.01, and ‡‡‡P < 0.001 vs. wild-type control. Right panels represent fold variation of luciferase activity for each construct in response to the respective treatment. #P < 0.05 and ###P < 0.001 vs. wild type. db mu., double mutant.

FIG. 3.

Differential usage of cis-acting elements in the Chop promoter by cytokines, palmitate, or CPA. A: Schematic representation of the mutations performed in the Chop promoter (×, substitution; ▪, deletion): C/EBP-ATF (○), AP-1 (▵), and ERSE (□) sites. INS-1E cells were transfected with wild-type or mutant Chop promoter constructs shown in A and treated with IL-1β (10 units/ml) + IFN-γ (0.18 μg/ml) (black bars) for 36 h (B); palmitate (0.5 mmol/l, hatched bars) for 36 h (C); CPA (25 μmol/l, dotted bars) for 15 h (D); or respective controls (white bars). Left panels represent the relative luciferase activity (RLU) of 4–16 experiments. *P < 0.05, **P < 0.01, and ***P < 0.001 vs. respective control; ‡P < 0.05, ‡‡P < 0.01, and ‡‡‡P < 0.001 vs. wild-type control. Right panels represent fold variation of luciferase activity for each construct in response to the respective treatment. #P < 0.05 and ###P < 0.001 vs. wild type. db mu., double mutant.

Close modal
FIG. 4.

Activation of the eIF2α/ATF4 pathway by cytokines or palmitate in INS-1E cells. A–C: Analysis of the C/EBP-ATF composite site. A: Schematic representation of the wild-type luciferase construct containing two copies of the C/EBP-ATF composite site from the Chop promoter. INS-1E cells were transfected with the wild-type or mutant C/EBP-ATF oligonucleotide reporter constructs (i.e., with mutations at either C/EBP or ATF) and treated with IL-1β (10 units/ml) + IFN-γ (0.18 μg/ml) (black bars) (B), palmitate (0.5 mmol/l, hatched bars) (C), or respective controls (white bars) for 36 h. Left panels represent relative luciferase activity of six to seven experiments. *P < 0.05 and ***P < 0.001 vs. respective control. Right panels represent fold variation of luciferase activity for each construct in response to the respective treatment. #P < 0.05 and ##P < 0.01 vs. wild type. D–G: Evaluation of eIF2α/ATF4 pathway activation by Western blot analysis. INS-1E cells were treated with IL-1β + IFN-γ (Ctk) (D); Ctk in the presence or absence of aminoguanidine (AG) (2.5 mmol/l) (E); palmitate (F); or CPA (G) for the indicated time points. Total eIF2α and β-actin serve as control for protein loading. Quantifications of the Western blots are shown as protein of interest optical density corrected per β-actin. Molecular weights of the proteins are also indicated. ND, not detectable. The figures are representative of two to three similar experiments.

FIG. 4.

Activation of the eIF2α/ATF4 pathway by cytokines or palmitate in INS-1E cells. A–C: Analysis of the C/EBP-ATF composite site. A: Schematic representation of the wild-type luciferase construct containing two copies of the C/EBP-ATF composite site from the Chop promoter. INS-1E cells were transfected with the wild-type or mutant C/EBP-ATF oligonucleotide reporter constructs (i.e., with mutations at either C/EBP or ATF) and treated with IL-1β (10 units/ml) + IFN-γ (0.18 μg/ml) (black bars) (B), palmitate (0.5 mmol/l, hatched bars) (C), or respective controls (white bars) for 36 h. Left panels represent relative luciferase activity of six to seven experiments. *P < 0.05 and ***P < 0.001 vs. respective control. Right panels represent fold variation of luciferase activity for each construct in response to the respective treatment. #P < 0.05 and ##P < 0.01 vs. wild type. D–G: Evaluation of eIF2α/ATF4 pathway activation by Western blot analysis. INS-1E cells were treated with IL-1β + IFN-γ (Ctk) (D); Ctk in the presence or absence of aminoguanidine (AG) (2.5 mmol/l) (E); palmitate (F); or CPA (G) for the indicated time points. Total eIF2α and β-actin serve as control for protein loading. Quantifications of the Western blots are shown as protein of interest optical density corrected per β-actin. Molecular weights of the proteins are also indicated. ND, not detectable. The figures are representative of two to three similar experiments.

Close modal
FIG. 5.

Cytokines (via NO production) or palmitate induce ATF4 binding to the C/EBP-ATF composite site of the Chop promoter. EMSA and supershift assays were performed using an oligonucleotide spanning the C/EBP-ATF composite site of the Chop promoter (nucleotides −335 to −323). Nuclear extracts were prepared from INS-1E cells treated with IL-1β (10 units/ml) + IFN-γ (0.18 μg/ml) (Ctk) in the presence or absence of aminoguanidine (AG) (2.5 mmol/l) (AC) or palmitate (0.5 mmol/l) (D and E). Twenty times excess wild-type or mutant competitor oligonucleotides were used to determine specificity of the complexes (A, lanes1114; D, lanes 11 and 12). C and E: Antibodies for ATF4 (lanes3 and 4) and C/EBPβ (lanes5 and 6) or BSA (2 μg/reaction) were used to supershift the complexes. Arrows α and β indicate specific complexes, whereas shifted complexes are indicated by “Shift”. The figures are representative of two to three similar experiments.

FIG. 5.

Cytokines (via NO production) or palmitate induce ATF4 binding to the C/EBP-ATF composite site of the Chop promoter. EMSA and supershift assays were performed using an oligonucleotide spanning the C/EBP-ATF composite site of the Chop promoter (nucleotides −335 to −323). Nuclear extracts were prepared from INS-1E cells treated with IL-1β (10 units/ml) + IFN-γ (0.18 μg/ml) (Ctk) in the presence or absence of aminoguanidine (AG) (2.5 mmol/l) (AC) or palmitate (0.5 mmol/l) (D and E). Twenty times excess wild-type or mutant competitor oligonucleotides were used to determine specificity of the complexes (A, lanes1114; D, lanes 11 and 12). C and E: Antibodies for ATF4 (lanes3 and 4) and C/EBPβ (lanes5 and 6) or BSA (2 μg/reaction) were used to supershift the complexes. Arrows α and β indicate specific complexes, whereas shifted complexes are indicated by “Shift”. The figures are representative of two to three similar experiments.

Close modal
FIG. 6.

Cytokines or palmitate induce AP-1 binding to the Chop promoter. EMSAs were performed using an oligonucleotide spanning the AP-1 site of the Chop promoter (nucleotides −253 to −236). Nuclear extracts were prepared from INS-1E cells treated with IL-1β (10 units/ml) + IFN-γ (0.18 μg/ml) (Ctk) (A), Ctk in the presence of absence of aminoguanidine (AG) (2.5 mmol/l) (B), or palmitate (0.5 mmol/l) (C). Twenty times excess wild-type or mutant oligonucleotide competitor was used to determine specificity of the complexes (A, lanes11 and 12; B, lanes 11–14). The specific complex is indicated by an arrowhead; ns, nonspecific complexes. The figures are representative of two to four similar experiments.

FIG. 6.

Cytokines or palmitate induce AP-1 binding to the Chop promoter. EMSAs were performed using an oligonucleotide spanning the AP-1 site of the Chop promoter (nucleotides −253 to −236). Nuclear extracts were prepared from INS-1E cells treated with IL-1β (10 units/ml) + IFN-γ (0.18 μg/ml) (Ctk) (A), Ctk in the presence of absence of aminoguanidine (AG) (2.5 mmol/l) (B), or palmitate (0.5 mmol/l) (C). Twenty times excess wild-type or mutant oligonucleotide competitor was used to determine specificity of the complexes (A, lanes11 and 12; B, lanes 11–14). The specific complex is indicated by an arrowhead; ns, nonspecific complexes. The figures are representative of two to four similar experiments.

Close modal
FIG. 7.

Differential composition of the AP-1 complex binding to the Chop promoter in response to cytokines or palmitate. A and B: EMSAs were performed as described in Fig. 5. For supershift assays, nuclear extracts of INS-1E cells treated with IL-1β (10 units/ml) + IFN-γ (0.18 μg/ml) (Ctk) for 8 h (A) or palmitate (0.5 mmol/l) for 16 h (B) were incubated with BSA (control) or antibodies against c-Fos, Jun-B, and c-Jun. The arrowhead indicates the specific complexes; “Shift” indicates shifted complexes; ns, nonspecific complex. C: Phospho–c-Jun Western blot of INS-1E cells treated or not with IL-1β (10 units/ml) + IFN-γ (0.18 μg/ml) (Ctk) for the indicated times. β-Actin serves as a control for protein loading. Molecular weight of phospho–c-Jun and β-actin are indicated. The figures are representative of two to three similar experiments.

FIG. 7.

Differential composition of the AP-1 complex binding to the Chop promoter in response to cytokines or palmitate. A and B: EMSAs were performed as described in Fig. 5. For supershift assays, nuclear extracts of INS-1E cells treated with IL-1β (10 units/ml) + IFN-γ (0.18 μg/ml) (Ctk) for 8 h (A) or palmitate (0.5 mmol/l) for 16 h (B) were incubated with BSA (control) or antibodies against c-Fos, Jun-B, and c-Jun. The arrowhead indicates the specific complexes; “Shift” indicates shifted complexes; ns, nonspecific complex. C: Phospho–c-Jun Western blot of INS-1E cells treated or not with IL-1β (10 units/ml) + IFN-γ (0.18 μg/ml) (Ctk) for the indicated times. β-Actin serves as a control for protein loading. Molecular weight of phospho–c-Jun and β-actin are indicated. The figures are representative of two to three similar experiments.

Close modal
FIG. 8.

Proposed model for transcriptional regulation of the Chop promoter in response to cytokines, palmitate, or CPA in β-cells. Description and discussion of the pathways is provided in the text.

FIG. 8.

Proposed model for transcriptional regulation of the Chop promoter in response to cytokines, palmitate, or CPA in β-cells. Description and discussion of the pathways is provided in the text.

Close modal

Additional information for this article can be found in an online appendix at http://dx.doi.org/10.2337/db06-1253.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

A.K.C. is the recipient of a postdoctoral fellowship from the Juvenile Diabetes Research Foundation and is presently a Research Associate of the Fonds National de la Recherche Scientifique (FNRS; Belgium). This work has received grants from the European Union (Integrated Project EuroDia LSHM-CT-2006-518153 in the Framework Programme 6 of the European Community), the FNRS, and the Communauté française de Belgique-Actions de Recherche Concertées.

We thank the personnel from the Laboratory of Experimental Medicine, ULB, especially M.A. Neef, G. Vandenbroeck, and R. Leeman for excellent technical support and Drs. Z. Dogusan and P. Hekerman for advice and help with phospho–Western blot analysis.

1.
Harding HP, Zeng H, Zhang Y, Jungries R, Chung P, Plesken H, Sabatini DD, Ron D: Diabetes mellitus and exocrine pancreatic dysfunction in perk−/− mice reveals a role for translational control in secretory cell survival.
Mol Cell
7
:
1153
–1163,
2001
2.
Scheuner D, Mierde DV, Song B, Flamez D, Creemers JW, Tsukamoto K, Ribick M, Schuit FC, Kaufman RJ: Control of mRNA translation preserves endoplasmic reticulum function in beta cells and maintains glucose homeostasis.
Nat Med
757
–764,
2005
3.
Ladiges WC, Knoblaugh SE, Morton JF, Korth MJ, Sopher BL, Baskin CR, MacAuley A, Goodman AG, LeBoeuf RC, Katze MG: Pancreatic β-cell failure and diabetes in mice with a deletion mutation of the endoplasmic reticulum molecular chaperone gene P58IPK.
Diabetes
54
:
1074
–1081,
2005
4.
Wu J, Kaufman RJ: From acute ER stress to physiological roles of the unfolded protein response.
Cell Death Differ
13
:
374
–384,
2006
5.
Cardozo AK, Ortis F, Storling J, Feng YM, Rasschaert J, Tonnesen M, Van Eylen F, Mandrup-Poulsen T, Herchuelz A, Eizirik DL: Cytokines downregulate the sarcoendoplasmic reticulum pump Ca2+ ATPase 2b and deplete endoplasmic reticulum Ca2+, leading to induction of endoplasmic reticulum stress in pancreatic β-cells.
Diabetes
54
:
452
–461,
2005
6.
Kharroubi I, Ladriere L, Cardozo AK, Dogusan Z, Cnop M, Eizirik DL: Free fatty acids and cytokines induce pancreatic beta-cell apoptosis by different mechanisms: role of nuclear factor-kappaB and endoplasmic reticulum stress.
Endocrinology
145
:
5087
–5096,
2004
7.
Karaskov E, Scott C, Zhang L, Teodoro T, Ravazzola M, Volchuk A: Chronic palmitate but not oleate exposure induces endoplasmic reticulum stress, which may contribute to INS-1 pancreatic β-Cell apoptosis.
Endocrinology
147
:
3398
–3407,
2006
8.
Contreras JL, Smyth CA, Bilbao G, Eckstein C, Young CJ, Thompson JA, Curiel DT, Eckhoff DE: Coupling endoplasmic reticulum stress to cell death program in isolated human pancreatic islets: effects of gene transfer of Bcl-2.
Transpl Int
16
:
537
–542,
2003
9.
Oyadomari S, Takeda K, Takiguchi M, Gotoh T, Matsumoto M, Wada I, Akira S, Araki E, Mori M: Nitric oxide-induced apoptosis in pancreatic beta cells is mediated by the endoplasmic reticulum stress pathway.
Proc Natl Acad Sci U S A
98
:
10845
–10850,
2001
10.
Eizirik DL, Mandrup-Poulsen T: A choice of death-the signal-transduction of immune-mediated beta-cell apoptosis.
Diabetologia
44
:
2115
–2133,
2001
11.
Cnop M, Welsh N, Jonas JC, Jorns A, Lenzen S, Eizirik DL: Mechanisms of pancreatic β-cell death in type 1 and type 2 diabetes: many differences, few similarities.
Diabetes
54 (Suppl. 2)
:
S97
–S107,
2005
12.
Pirot P, Eizirik DL, Cardozo AK: Interferon-gamma potentiates endoplasmic reticulum stress-induced death by reducing pancreatic beta cell defence mechanisms.
Diabetologia
49
:
1229
–1236,
2006
13.
Butler AE, Janson J, Bonner-Weir S, Ritzel R, Rizza RA, Butler PC: β-Cell deficit and increased β-cell apoptosis in humans with type 2 diabetes.
Diabetes
52
:
102
–110,
2003
14.
Rhodes CJ: Type 2 diabetes: a matter of beta-cell life and death?
Science
307
:
380
–384,
2005
15.
Oyadomari S, Mori M: Roles of CHOP/GADD153 in endoplasmic reticulum stress.
Cell Death Differ
11
:
381
–389,
2004
16.
Eizirik DL, Bjorklund A, Cagliero E: Genotoxic agents increase expression of growth arrest and DNA damage-inducible genes gadd 153 and gadd 45 in rat pancreatic islets.
Diabetes
42
:
738
–745,
1993
17.
Ron D, Habener JF: CHOP, a novel developmentally regulated nuclear protein that dimerizes with transcription factors C/EBP and LAP and functions as a dominant-negative inhibitor of gene transcription.
Genes Dev
6
:
439
–453,
1992
18.
Ubeda M, Wang XZ, Zinszner H, Wu I, Habener JF, Ron D: Stress-induced binding of the transcriptional factor CHOP to a novel DNA control element.
Mol Cell Biol
16
:
1479
–1489,
1996
19.
Ubeda M, Vallejo M, Habener JF: CHOP enhancement of gene transcription by interactions with Jun/Fos AP-1 complex proteins.
Mol Cell Biol
19
:
7589
–7599,
1999
20.
Gotoh T, Terada K, Oyadomari S, Mori M: Hsp70-DnaJ chaperone pair prevents nitric oxide- and CHOP-induced apoptosis by inhibiting translocation of Bax to mitochondria.
Cell Death Differ
11
:
390
–402,
2004
21.
McCullough KD, Martindale JL, Klotz LO, Aw TY, Holbrook NJ: Gadd153 sensitizes cells to endoplasmic reticulum stress by down- regulating Bcl2 and perturbing the cellular redox state.
Mol Cell Biol
21
:
1249
–1259,
2001
22.
Yamaguchi H, Wang HG: CHOP is involved in endoplasmic reticulum stress-induced apoptosis by enhancing DR5 expression in human carcinoma cells.
J Biol Chem
279
:
45495
–45502,
2004
23.
Oyadomari S, Koizumi A, Takeda K, Gotoh T, Akira S, Araki E, Mori M: Targeted disruption of the Chop gene delays endoplasmic reticulum stress-mediated diabetes.
J Clin Invest
109
:
525
–532,
2002
24.
Guyton KZ, Xu Q, Holbrook NJ: Induction of the mammalian stress response gene GADD153 by oxidative stress: role of AP-1 element.
Biochem J
314
:
547
–554,
1996
25.
Yoshida H, Okada T, Haze K, Yanagi H, Yura T, Negishi M, Mori K: ATF6 activated by proteolysis binds in the presence of NF-Y (CBF) directly to the cis-acting element responsible for the mammalian unfolded protein response.
Mol Cell Biol
20
:
6755
–6767,
2000
26.
Bruhat A, Jousse C, Wang XZ, Ron D, Ferrara M, Fafournoux P: Amino acid limitation induces expression of CHOP, a CCAAT/enhancer binding protein-related gene, at both transcriptional and post-transcriptional levels.
J Biol Chem
272
:
17588
–17593,
1997
27.
Ma Y, Brewer JW, Diehl JA, Hendershot LM: Two distinct stress signaling pathways converge upon the CHOP promoter during the mammalian unfolded protein response.
J Mol Biol
318
:
1351
–1365,
2002
28.
Schmitt-Ney M, Habener JF: CHOP/GADD153 gene expression response to cellular stresses inhibited by prior exposure to ultraviolet light wavelength band C (UVC): inhibitory sequence mediating the UVC response localized to exon 1.
J Biol Chem
275
:
40839
–40845,
2000
29.
Merglen A, Theander S, Rubi B, Chaffard G, Wollheim CB, Maechler P: Glucose sensitivity and metabolism-secretion coupling studied during two-year continuous culture in INS-1E insulinoma cells.
Endocrinology
145
:
667
–678,
2004
30.
Kutlu B, Cardozo AK, Darville MI, Kruhoffer M, Magnusson N, Orntoft T, Eizirik DL: Discovery of gene networks regulating cytokine-induced dysfunction and apoptosis in insulin-producing INS-1 cells.
Diabetes
52
:
2701
–2719,
2003
31.
Green LC, Wagner DA, Glogowski J, Skipper PL, Wishnok JS, Tannenbaum SR: Analysis of nitrate, nitrite, and [15N]nitrate in biological fluids.
Anal Biochem
126
:
131
–138,
1982
32.
Chen MC, Schuit F, Eizirik DL: Identification of IL-1β-induced messenger RNAs in rat pancreatic beta cells by differential display of messenger RNA.
Diabetologia
42
:
1199
–1203,
1999
33.
Darville MI, Eizirik DL: Regulation by cytokines of the inducible nitric oxide synthase promoter in insulin-producing cells.
Diabetologia
41
:
1101
–1108,
1998
34.
Schreiber E, Matthias P, Muller MM, Schaffner W: Rapid detection of octamer binding proteins with ‘mini-extracts’, prepared from a small number of cells.
Nucleic Acid Res
17
:
6419
,
1989
35.
Averous J, Bruhat A, Jousse C, Carraro V, Thiel G, Fafournoux P: Induction of CHOP expression by amino acid limitation requires both ATF4 expression and ATF2 phosphorylation.
J Biol Chem
279
:
5288
–5297,
2004
36.
Yang X, Matsuda K, Bialek P, Jacquot S, Masuoka HC, Schinke T, Li L, Brancorsini S, Sassone-Corsi P, Townes TM, Hanauer A, Karsenty G: ATF4 is a substrate of RSK2 and an essential regulator of osteoblast biology; implication for Coffin-Lowry Syndrome.
Cell
117
:
387
–398,
2004
37.
Wek RC, Jiang HY, Anthony TG: Coping with stress: eIF2 kinases and translational control.
Biochem Soc Trans
34
:
7
–11,
2006
38.
Eizirik DL: Interleukin-1 beta induces an early decrease in insulin release, (pro)insulin biosynthesis and insulin mRNA in mouse pancreatic islets by a mechanism dependent on gene transcription and protein synthesis.
Autoimmunity
10
:
107
–113,
1991
39.
Herdegen T, Leah JD: Inducible and constitutive transcription factors in the mammalian nervous system: control of gene expression by Jun, Fos and Krox, and CREB/ATF proteins.
Brain Res Brain Res Rev
28
:
370
–490,
1998
40.
Kwon G, Corbett JA, McDaniel ML: Interleukin 1-induced Fos and Jun do not regulate inducible nitric oxide synthase in rat islets of Langerhans and RINm5F cells.
Endocrinology
137
:
825
–830,
1996
41.
Eizirik DL, Leijerstam F: The inducible form of nitric oxide synthase (iNOS) in insulin-producing cells.
Diabetes Metab
20
:
116
–122,
1994
42.
Hughes JH, Watson MA, Easom RA, Turk J, McDaniel ML: Interleukin-1 induces rapid and transient expression of the c-fos proto-oncogene in isolated pancreatic islets and in purified beta-cells.
FEBS Lett
266
:
33
–36,
1990
43.
Morton S, Davis RJ, McLaren A, Cohen P: A reinvestigation of the multisite phosphorylation of the transcription factor c-Jun.
EMBO J
22
:
3876
–3886,
2003
44.
Li L, Feng Z, Porter AG: JNK-dependent phosphorylation of c-Jun on serine 63 mediates nitric oxide-induced apoptosis of neuroblastoma cells.
J Biol Chem
279
:
4058
–4065,
2004
45.
Bonny C, Oberson A, Negri S, Sauser C, Schorderet DF: Cell-permeable peptide inhibitors of JNK: novel blockers of β-cell death.
Diabetes
50
:
77
–82,
2001
46.
Nikulina MA, Sandhu N, Shamim Z, Andersen NA, Oberson A, Dupraz P, Thorens B, Karlsen AE, Bonny C, Mandrup-Poulsen T: The JNK binding domain of islet-brain 1 inhibits IL-1 induced JNK activity and apoptosis but not the transcription of key proapoptotic or protective genes in insulin-secreting cell lines.
Cytokine
24
:
13
–24,
2003
47.
Urano F, Wang X, Bertolotti A, Zhang Y, Chung P, Harding HP, Ron D: Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1.
Science
287
:
664
–666,
2000
48.
Nishitoh H, Matsuzawa A, Tobiume K, Saegusa K, Takeda K, Inoue K, Hori S, Kakizuka A, Ichijo H: ASK1 is essential for endoplasmic reticulum stress-induced neuronal cell death triggered by expanded polyglutamine repeats.
Genes Dev
16
:
1345
–1355,
2002
49.
Maytin EV, Ubeda M, Lin JC, Habener JF: Stress-inducible transcription factor CHOP/gadd153 induces apoptosis in mammalian cells via p38 kinase-dependent and -independent mechanisms.
Exp Cell Res
267
:
193
–204,
2001
50.
Ubeda M, Habener JF: CHOP transcription factor phosphorylation by casein kinase 2 inhibits transcriptional activation.
J Biol Chem
278
:
40514
–40520,
2003

Supplementary data