当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 糖尿病学杂志 > 2005年 > 第8期 > 正文
编号:11256704
Disruption of the -Interferon Signaling Pathway at the Level of Signal Transducer and Activator of Transcription-1 Prevents Immune Destructi
     1 Laboratory of Experimental Medicine and Endocrinology, UZ Gasthuisberg O&N, Katholieke Universiteit Leuven, Leuven, Belgium

    2 Laboratory of Experimental Medicine, Universitee Libre de Bruxelles, Brussels, Belgium

    3 Department of Pathology and Kaplan Cancer Center, New York University School of Medicine, New York, New York

    4 Laboratory of Immunobiology, Rega Institute for Medical Research, Katholieke Universiteit Leuven, Leuven, Belgium

    ABSTRACT

    -Cells under immune attack are destroyed by the aberrant activation of key intracellular signaling cascades. The aim of the present study was to evaluate the contribution of the signal transducer and activator of transcription (STAT)-1 pathway for -cell apoptosis by studying the sensitivity of -cells from STAT-1 knockout (eC/eC) mice to immune-mediated cell death in vitro and in vivo. Whole islets from STAT-1eC/eC mice were completely resistant to interferon (IFN)- (studied in combination with interleukin [IL]-1)-mediated cell death (92 ± 4% viable cells in STAT-1eC/eC mice vs. 56 ± 3% viable cells in wild-type controls, P 0.001) and had preserved insulin release after exposure to IL-1 and IFN-. Moreover, analysis of cell death in cytokine-exposed purified -cells confirmed that protection was due to absence of STAT-1 in the -cells themselves. Deficiency of STAT-1 in islets completely prevented cytokine-induced upregulation of IL-15, interferon inducible protein 10, and inducible nitric oxide synthase transcription but did not interfere with monocyte chemoattractant protein 1 and macrophage inflammatory protein 3 expression. In vivo, STAT-1eC/eC mice were partially resistant to development of diabetes after multiple low-dose streptozotocin injections as reflected by mean blood glucose at 12 days after first injection (159 ± 28 vs. 283 ± 81 mg/dl in wild-type controls, P 0.05) and diabetes incidence at the end of the follow-up period (39 vs. 73% in wild-type controls, P 0.05). In conclusion, the present results indicate that STAT-1 is a crucial transcription factor in the process of IFN-eCmediated -cell death and the subsequent development of immune-mediated diabetes.

    The main focus in studies on the pathogenesis of autoimmune type 1 diabetes has been on the role of the immune system, with much less attention directed to the target of the immune assault, namely the insulin-secreting -cells. -Cells under inflammatory attack have massive changes in gene expression, as evaluated by microarray analysis (1,2). These genes are controlled by families of transcription-regulating signaling molecules, such as signal transducer and activator of transcription (STAT) and nuclear factor-B (NF-B) (3,4), which become activated after exposure to proinflammatory cytokines. Blocking one of these transcription factors, namely NF-B, prevents cytokine-induced -cell death (3,5). These observations suggest that aberrant activation of key intracellular signaling pathways in the -cells ultimately determine -cell survival or death.

    Pancreatic -cells are exposed to many inflammatory assaults such as cytokines during insulitis (6,7), and in vitro studies indicate that interferon (IFN)- is of particular importance to sensitize human and rodent islets for -cell death induced by another cytokine, namely interleukin (IL)-1 (8). Indeed, although IL-1 alone does not suffice to induce the death of human and rodent -cells, a combination of IL-1 plus IFN- kills half of the cells after 6eC9 days (9,10). Moreover, IFN-, in combination with double-stranded RNA produced during viral infections, is a potent inducer of -cell dysfunction and death (11). Although IL-1 exerts its main effects through the NF-B pathway, IFN- acts mostly via janus kinase (JAK) activation, with the intracellular transcription factor STAT-1 playing a central role in the downstream pathway. Upon IFN- ligand-receptor coupling, STAT-1 is recruited to receptor phosphotyrosine residues that provide docking sites for the Src-homology-2 domain of STAT-1. Next, STAT-1 phosphorylation on tyrosine-701 leads to STAT-1 dimerization and translocation to the nucleus where it regulates gene expression by binding to -activated sequences in the promoter of target genes (12). Depending on the cell type and on the parallel induction of other signaling pathways, activation of STAT-1 may lead to either pro- or antiapoptotic effects (14).

    Several genes with putative roles in -cell death are activated by IFN-, including major histocompatibility complex class I (15eC17), the adhesion molecule intracellular adhesion molecule 1 (18,19), inducible NO synthase (iNOS) (20), the death receptor Fas (21), caspase-1 (22,23), the chemokine interferon inducible protein (IP)-10, and the cytokine IL-15 (7) as well as interferon-regulated factor (IRF)-1 (24,25) but whether regulation of all of these genes in primary -cells is dependent on STAT-1 activation is still under investigation. Of note, we have previously shown that inhibition of IRF-1, a transcription factor located immediately downstream of STAT-1, does not prevent cytokine-induced apoptosis (9).

    The aim of the present study was to investigate whether disruption of STAT-1 prevents cytokine-mediated -cell death in vitro and in vivo. For this purpose, we isolated islets and purified primary -cells from wild-type mice or mice lacking STAT-1 and exposed them to cytokines in vitro. There was a complete protection against -cell death, as well as a modified chemokine and cytokine expression profile. In line with these in vitro observations, STAT-1 knockout mice were less susceptible to multiple low-dose streptozotocin (MLDS)-induced diabetes in vivo. Our results indicate that STAT-1 plays a central role in inflammation-induced -cell death. Disrupting IFN- signaling at this level but not at the level of IRF-1 confers -cell protection.

    RESEARCH DESIGN AND METHODS

    The STAT-1eC/eC mutation was produced by deleting 5.7 kb of the STAT-1 gene, resulting in a total inability to produce STAT-1 in homozygous mutant animals (26). These mice have been backcrossed for 10 generations in the C57BL/6 background. STAT-1eC/eC mice were kept under specific pathogen free conditions. C57BL/6 mice (Harlan, Zeist, the Netherlands) were used as wild-type controls. All experimental designs were conducted with the approval of the Animal Ethics Committees of the Katholieke Universiteit Leuven (in vivo studies) and Universitee Libre de Bruxelles (in vitro studies).

    Isolation, culture, and cytokine treatment of whole islets and primary -cells.

    Islets were obtained from 10-week-old male and female wild-type control and STAT-1eC/eC mice as previously described (9,27). To obtain purified -cells, islets were dissociated and sorted by FACStar flow cytometer (Becton Dickinson, Sunnyvale, CA), as previously described for rat -cells (28). The preparations used in the present experiments contained 84 ± 3% and 84 ± 4% -cells for wild-type and STAT-1eC/eC mice, respectively. Cell culture was performed in Ham’s F-10 medium supplemented with 10 mmol/l glucose, 50 eol/l isobutylmethylxanthine, 0.5% BSA (Boehringer Mannheim, Mannheim, Germany), and 2 mmol/l L-glutamine (Life Technologies, Paisley, Scotland). These culture conditions, originally developed for the maintenance of rat -cells, have been validated for mouse islet or -cell culture (10).

    Whole islets were cultured in suspension, whereas fluorescence-activated cell sorter (FACS)eCpurified single -cells were cultured attached to polylysine-coated dishes. The effect of cytokines was examined after 2 and 5 days of culture in the presence of recombinant murine IFN- (1,000 units/ml, 107 units/mg; PeproTech) and recombinant human IL-1 (50 units/ml, 38 units/ng; kind gift of Dr. C.W. Reynolds from the National Cancer Institute, Bethesda, MD). These concentrations of cytokines were selected based on our previous studies with mouse pancreatic islets and -cells (9,10,29).

    Assessment of -cell viability and medium insulin release.

    The viability of islet cell preparations was assessed after 2 and 5 days exposure to cytokines. Whole islets or isolated -cells were incubated for 15 min with propidium iodide (10 mg/ml) and Hoechst 342 (20 mg/ml). The approximate percentage of dead islet cells was estimated by two individual observers, with one of them unaware of the sample identity (9,10). Evaluation of cell death in whole islets is complicated by the superposition between cells, and these observations should thus be considered as semiquantitative. The percentages of apoptotic cells in the single -cell preparations were assessed by propidium iodide and Hoechst after 2 or 5 days exposure to cytokines, as previously described for mouse -cells (9,10). This fluorescence assay for single -cells is quantitative and has been validated by systematic comparisons with electron microscopy observations (30). The apoptosis and necrosis indexes were calculated as [(percent apoptotic or necrotic cells in experimental condition eC percent apoptotic or necrotic cells in control)/(100 eC percent dead cells in control)] x 100. The percent viability in control single -cells (not exposed to cytokines) was in the range of 55eC70%. These values were similar after 2 or 5 days of culture and were not significantly different between wild-type and STAT-1eC/eC -cells (data not shown).

    Culture media were collected after 24 and 48 h, stored at eC20°C and assayed for immune-reactive insulin. Insulin release in the culture media was determined by enzyme-linked immunosorbent assay (ELISA).

    Intraperitoneal glucose tolerance test.

    Intraperitoneal glucose tolerance tests (IPGTTs) were carried out in 8-week-old male wild-type and STAT-1eC/eC mice after an overnight fast. Briefly, mice were injected intraperitoneally with 2 g/kg body wt D-glucose dissolved in sterile PBS. Blood samples were drawn at different time points (0, 15, 30, 60, 90, and 120 min) from the tail vein. Plasma glucose concentrations were assayed by a glucose oxidase method using an automatic glucometer (Glucocard, Menarini).

    Streptozotocin-induced diabetes.

    For MLDS induction, 8-week-old male STAT-1eC/eC and wild-type mice were injected intraperitoneally with freshly dissolved streptozotocin (50 mg/kg) (Sigma, St. Louis, MO) for 5 consecutive days. Development of glucosuria was monitored with Clinistix (Bayer Diagnostics) and glucose concentrations of venous blood were determined using a glucometer (Glucocard, Menarini) as described above. Clinical diabetes was defined by positive glucosuria and hyperglycemia (blood glucose levels >250 mg/dl) on 2 consecutive days in nonfasted animals. Mice were followed until 40 days after the last injection of streptozotocin. At that time point, animals were killed and pancreata were removed for histology and insulin content measurement. In a separate set of wild-type and STAT-1eC/eC animals, the pancreas was removed 3 days (experimental day 8) after the start of streptozotocin for histology.

    Histology and insulin content determination.

    Pancreata were removed from killed mice and dissected free of adipose tissue and lymph nodes in PBS at 4°C under a stereomicroscope. Half of each pancreas was snap-frozen in liquid nitrogen-chilled isopentane and stored at eC20°C until processing. For histology, serial cryostat sections (5-e thickness) were cut and stained with hematoxylin and eosin to assess pancreatic islet integrity and immune cell infiltration. The remaining half of the pancreas was used for insulin content determination. Briefly, protein was extracted overnight at 4°C in acideCethanol. Insulin concentrations in the extracts were measured using an ultrasensitive rat insulin ELISA kit (Mercodia, Uppsala, Sweden) with rat insulin as standard as previously described (31).

    Real-time PCR analysis.

    Cultured islet cells (24 h of cytokine exposure) were snap-frozen in liquid nitrogen and stored at eC80°C until RNA extraction (TRIzol reagent, Invitrogen). cDNA was created using Superscript II RT (Invitrogen) and quantitative PCR analysis was performed with the ABI prism 7700 Sequence detector (Applied Biosystems, Foster City, CA) as previously described (32). PCR conditions were 2 min at 50°C and 10 min at 94°C, followed by a total of 40eC45 two-temperature cycles (15 s at 94°C and 1 min at 60°C). Primer and probe sequences for the determination of mouse cDNAs for housekeeping gene -actin (33), IL-1 (33), IL-15 (33), IFN- (33), iNOS (33), MCP-1 (34), IP-10 (34), and MIP-3 (34) were as described in the indicated references. The target cDNAs present in each sample were corrected for the respective -actin values. The primer and probe sequences used for determination of mouse cDNA for STAT-1 were: FW, 5'-CACCAGAACCGATGGAGCTT-3'; RV, 5'-TCCGGGACATCTCATCAAACT-3'; and TP, 5'-CACCCTTCTAGACTTCAGACCACAGACAACCT-3'.

    Statistical analysis.

    The statistical software NCSS 2000 (Kaysville, UT) was used for statistical analysis. Data are expressed as means ± SE. Areas under the curve were determined for analysis of IPGTT, whereas the 2 test was performed for the incidence of MLDS-induced diabetes. ANOVA and Student’s t test were used for multiple comparisons whenever appropriate. Significance was defined as P 0.05. Unless indicated otherwise, the graphs show triplicate determinations from at least three independent experiments.

    RESULTS

    Cell viability and insulin release of whole islets isolated from wild-type and STAT-1eC/eC mice after exposure to a combination of cytokines.

    In an initial series of experiments, we confirmed the STAT-1eCdeficient state of the pancreatic islets from knockout mice. Thus, there was no STAT-1 mRNA expression under basal conditions of cell culture or after exposure to IL-1 plus IFN- in islets from STAT-1eC/eC mice (Fig. 1). In contrast, STAT-1 expression was clearly upregulated in islets from wild-type mice after stimulation with IL-1 + IFN- (CYTK) (Fig. 1).

    Next, we confirmed that treatment with IL-1 in combination with IFN- significantly decreased cell viability of wild-type islets after 48 h of cytokine exposure (Fig. 2A). After 5 days of cytokine exposure, only 22 ± 13% wild-type viable cells were left compared with 93 ± 1% viable cells in control conditions (Fig. 2B). Islets from STAT-1eC/eC mice showed resistance to cytokine-induced islet destruction, as reflected by fully preserved islet viability after 48 h (Fig. 2A) and 5 days of cell culture (Fig. 2B). In parallel, insulin release into the medium by wild-type islets decreased significantly upon exposure to the cytokine combination IL-1 plus IFN-, whereas insulin secretion was fully preserved in STAT-1eC/eC islets exposed to cytokines (Fig. 2C and D).

    Cell viability of primary -cells isolated from wild-type and STAT-1eC/eC mice after exposure to a combination of cytokines.

    To discriminate between apoptosis and necrosis and to investigate the importance of the absence of STAT-1 in the -cells themselves in the observed protection against cytokine-induced islet cell death, FACS-purified -cells from wild-type and STAT-1eC/eC mice were studied (Table 1). In accord with the results obtained in whole islets, exposure of wild-type -cells to cytokines induced a clear increase in the apoptotic index at both 2 and 5 days (Table 1), but there was no increase in the necrosis index (data not shown), confirming that apoptosis is the main form of cytokine-induced -cell death in this model (8). On the other hand, STAT-1eC/eC purified -cells were totally protected against cytokine-induced apoptosis, with apoptotic indexes around zero after 2 and 5 days (Table 1).

    Gene expression in whole islets isolated from wild-type and STAT-1eC/eC mice after exposure to a combination of cytokines.

    Under basal conditions, wild-type and STAT-1eC/eC islets expressed IL-1, MCP-1, MIP-3, IP-10, and iNOS mRNAs at comparable levels (Fig. 3), but there was no expression for IL-15 in STAT-1eC/eC islets in basal culture conditions.

    Exposure of wild-type islets to inflammatory cytokines induced a marked upregulation of several genes encoding for chemokines and iNOS. A similar upregulation was observed in STAT-1eC/eC islets for MCP-1 and MIP-3, but induction of IL-15, IP-10, and iNOS was completely prevented by the STAT-1 disruption (Fig. 3).

    Streptozotocin-induced diabetes.

    We next examined the effects of STAT-1 disruption in vivo in a model of immune-mediated -cell destruction, namely MLDS. To assure that the basal conditions were similar between wild-type and STAT-1eC/eC mice, we first demonstrated that deletion of STAT-1 does not affect normal glucose tolerance. IPGTTs, performed in wild-type and STAT-1eC/eC mice 8 weeks of age, showed similar responses as reflected by the area under the glucose curve (Fig. 4). In addition, no differences were noted between control and knockout pancreatic insulin content (164 ± 15 vs. 148 ± 46 pmol/mg pancreas in wild-type and STAT-1eC/eC mice, respectively).

    Second, we excluded differences in the sensitivity to a single high dose of streptozotocin (150 mg/kg). Blood glucose levels and diabetes incidence were similar in wild-type and STAT-1eC/eC mice 48 h after high-dose streptozotocin (394 ± 52 mg/dl [100%, 6/6] vs. 386 ± 24 mg/dl [100%, 6/6], respectively; NS). Further follow-up of the animals up to 7 days confirmed stable high blood glucose level in wild-type andeC/eC mice (data not shown).

    A clear difference in sensitivity to MLDS was, however, observed between wild-type and STAT-1eC/eC mice. At the start of the five daily injections of streptozotocin, plasma glucose levels were 98 ± 17 mg/dl in wild-type mice and 95 ± 6 mg/dl in STAT-1eC/eC mice (NS). On experimental day 10, glucose levels were significantly higher in wild types than in STAT-1eC/eC mice and increased rapidly on day 17, continuing to rise until reaching a plateau level after 30 days. On the other hand, glucose levels remained virtually constant in streptozotocin-treated STAT-1eC/eC mice until day 45 (Fig. 5A).

    Although 73% of wild-type mice developed diabetes (defined as positive glucosuria and blood glucose levels >250 mg/dl on 2 consecutive days) by 40 days after the last streptozotocin injection, only 39% of STAT-1eC/eC mice reached the diabetic level at the end of the follow-up period (P 0.05 vs. controls) (Fig. 5B), suggesting that absence of STAT-1 provides a partial protection against development of diabetes. That this protection was only partial is confirmed by the pancreatic insulin content determination at the end of the experiment, where a clearly higher insulin level is still present in the STAT-1eC/eC pancreata versus controls (21.8 ± 3.9 vs. 5.8 ± 1.7 pmol/mg pancreas, P 0.01), although a 85% decrease was observed compared with the levels present before streptozotocin administration (see above). This was also reflected by the histological image at the end of the study where mostly small islets were present in both groups (data not shown).

    DISCUSSION

    Pancreatic -cells are particularly susceptible to the cytotoxic effects of inflammatory cytokines, secreted by infiltrating immune cells during the course of insulitis (8). Cytokine-mediated -cell death results from changes in the global gene expression profile of -cells (1,2). Therefore, autoimmune diabetes can be seen as a failure of intracellular signaling pathways in the -cells, pushing these cells toward death instead of survival and making these molecular pathways increasingly relevant as therapeutic targets. The cytokine IFN-, in synergy with other stimuli, triggers in the -cells the expression and secretion of a large number of proinflammatory cytokines, chemokines, costimulatory molecules, and enzymes, which are known to control cellular immune reactions that contribute to -cell destruction (1,11). For many of these genes, mRNA expression is regulated at the transcriptional level by binding of IFN--activated STAT-1 (itself upregulated in -cells after costimulation with inflammatory cytokines) to respective binding sites (-activated sequences) within the target promoters (1,11).

    In the present study, we used mice with a knockout of the transcription factor STAT-1 to determine whether JAK-STAT activation is essential for cytokine-stimulated gene expression and -cell death in vitro and to investigate whether these findings are translated into protection against in vivo -cell destruction. We found that absence of STAT-1 in whole islets protects them against IFN- (studied in combination with IL-1)eCinduced cell death. Moreover, the observation of complete protection of FACS-purified -cells from STAT-1eC/eC mice against cytokine-induced apoptosis confirms that the protection is due to the absence of STAT-1 in the -cells themselves and not to its absence in putative passenger leukocytes. In line with our observations, other studies have recently shown that -cell-specific overexpression of suppressor of cytokine signaling (SOCS) proteins 1 and 3, which block IFN- activation of the JAK-STAT signaling cascade, prevents diabetes in different mouse models (35eC37). Overexpression of SOCS-1, however, impairs both STAT-1 tyrosine phosphorylation and toll-like receptor signaling (38), whereas upregulation of SOCS-3 blocks NF-B activation (39) and modifies signaling via the insulin receptor (40) in -cells. Significantly, our data demonstrate that interfering exclusively with IFN--activated JAK-STAT signaling protects insulin-secreting -cells from cytokine-elicited inflammatory responses and death in vitro. We have previously shown that IL-1 contributes to -cell death via activation of NF-B (3,5). Taken together with the present data, we suggest that STAT-1 and NF-B are key transcription factors regulating the gene networks that trigger -cell apoptosis.

    We have previously demonstrated that expression of IL-15, IP-10, and iNOS is regulated by IFN-, whereas the expression of MCP-1 and MIP-3 is IL-1eCdependent (7). In the case of IFN-, many responsive genes are induced through interaction of phosphorylated STAT-1 homodimers and an inducible enhancer termed -activated sequences. Other potential downstream effectors responsible for gene induction by IFN- are IRF-1 (41), the extracellular signal-regulated kinases 1 and 2 (42), NF-B (p65/p50) (43), and CCAAT/enhancer-binding protein-. An important observation in the present study is that STAT-1eCdeficient islets, in contrast to wild-type islets, are unable to upregulate expression of mRNAs for IL-15, IP-10, and the enzyme iNOS after cytokine exposure, suggesting that JAK/STAT activation is crucial for such responses. On the other hand, no inhibitory effects of STAT-1 deficiency were observed on cytokine-elicited MCP-1 and MIP-3 expression in -cells.

    STAT-1 actions are cell- and context-dependent and may lead to opposite outcomes, such as increasing survival or apoptosis (44). Studies using cells and mice deficient in STAT-1 have shown that the transcription factor mediates most immune/inflammatory effects of IFN-, including the induction of immune effector and inflammatory genes (26,45,46) and both pro- and antiapoptotic genes (14). Of note, recent studies have identified several IFN-eCinduced but STAT-1eCindependent genes in diverse cell types (47eC49), leading to a currently ongoing reevaluation of the role of STAT-1 for IFN- signaling. We found that a targeted deletion of STAT-1 protein in mice did not affect the -cell mass homeostasis, as demonstrated by normal IPGTT and comparable pancreatic insulin content to wild-type controls, but causes a partial protection against -cell destruction by multiple low doses of streptozotocin. On the other hand, no direct protection was observed against the toxic effects of a high dose of streptozotocin on -cells, suggesting that absence of STAT-1 prevents -cell death in MLDS-induced diabetes by interfering with immune-mediated -cell death, not by interfering with the alkylating actions of streptozotocin. Because these mice were deficient for STAT-1 at both the -cell and immune-system levels, the present data do not allow us to conclude whether the observed protection was due to beneficial effects of STAT-1 absence on the immune system or -cells or, more probably, in both. Of note, the degree of immune-cell infiltration in the islets after MLDS treatment was similar between wild-type and STAT-1eC/eC mice, suggesting that under similar immune attack -cells from STAT-1eC/eC mice are more resistant to cell death.

    These data, together with our previous observations, suggest a dual role for IFN- signaling in -cell destruction. Thus, we have previously shown that disruption of the transcription factor IRF-1 not only failed to prevent cytokine-induced -cell death in vitro (9), but it also aggravated MLDS-induced diabetes in vivo (50). This is in marked contrast with the present findings and suggests that although STAT-1eCregulated genes have mainly a proapoptotic role in -cells, genes regulated by the downstream transcription factor IRF-1 have a protective role against -cell destruction. New experiments, including microarray analysis, are now required to identify the nature of these STAT-1eCand/or IRF-1eCregulated genes.

    In conclusion, we have demonstrated a central role of STAT-1 in inflammation-induced -cell death. Disruption of the STAT-1eCmediated signaling cascade prevents upregulation of several chemokines and iNOS and completely prevents IFN- (studied in combination with IL-1)-mediated -cell death in vitro. Moreover, blocking STAT-1 partially protects -cells against destruction in vivo. These data suggest that STAT-1 may become a therapeutic target in our efforts to increase -cell resistance against immune destruction.

    ACKNOWLEDGMENTS

    This work was supported by the JDRF Center for Prevention of Beta Cell Destruction in Europe Grant 4-2002-457; the Flemish Research Foundation Fonds Voor Wetenschappelijk Onderzoek (FWO) Grants G.0084.02 and G.0233.04, a doctoral scholarship for H.C., a clinical fellowship for C.M, and a postdoctoral fellowship for C.G. and P.M.; the Belgian Research Foundation Fonds National de la Recherche Scientifique; the Belgian Program on Interuniversity Poles of Attraction initiated by the Belgian State Grant IUAP P5/17; the Katholieke Universiteit Leuven Grant GOA 2004/10; and an Action de Recherche Concerteee de la Communautee franaise de Belgique.

    The authors thank the personnel of the laboratory of Experimental Medicine and Endocrinology at KUL (in particular J. Depovere, D. Valckx, and J. Laureys) and of the Laboratory of Experimental Medicine at ULB (in particular M. Neef, J. Schoonheydt, and M. Urbain) for their excellent technical support and constructive discussions.

    FOOTNOTES

    C.A.G. and L.L. contributed equally to this study.

    ELISA, enzyme-linked immunosorbent assay; FACS, fluorescence-activated cell sorter; IFN, interferon; IL, interleukin; iNOS, inducible nitric oxide synthase; IP, interferon-inducible protein; IPGTT, intraperitoneal glucose tolerance test; IRF, interferon regulatory factor; JAK, janus kinase; MIP, macrophage inflammatory protein; MLDS, multiple low-dose streptozotocin; NF-B, nuclear factor-B; SOCS, suppressor of cytokine signaling; STAT, signal transducer and activator of transcription

    REFERENCES

    Cardozo AK, Kruhoffer M, Leeman R, Orntoft T, Eizirik DL: Identification of novel cytokine-induced genes in pancreatic -cells by high-density oligonucleotide arrays. Diabetes50 :909 eC920,2001

    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. Diabetes52 :2701 eC2719,2003

    Cardozo AK, Heimberg H, Heremans Y, Leeman R, Kutlu B, Kruhoffer M, Orntoft T, Eizirik DL: A comprehensive analysis of cytokine-induced and nuclear factor-B-dependent genes in primary rat pancreatic -cells. J Biol Chem276 :48879 eC48886,2001

    Eizirik DL, Kutlu B, Rasschaert J, Darville M, Cardozo AK: Use of microarray analysis to unveil transcription factor and gene networks contributing to cell dysfunction and apoptosis. Ann N Y Acad Sci1005 :55 eC74,2003

    Heimberg H, Heremans Y, Jobin C, Leemans R, Cardozo AK, Darville M, Eizirik DL: Inhibition of cytokine-induced NF-B activation by adenovirus-mediated expression of a NF-B super-repressor prevents -cell apoptosis. Diabetes50 :2219 eC2224,2001

    Thomas HE, Kay TW: How beta cells die in type 1 diabetes. Curr Dir Autoimmun4 :144 eC170,2001

    Cardozo AK, Proost P, Gysemans C, Chen MC, Mathieu C, Eizirik DL: IL-1 and IFN- induce the expression of diverse chemokines and IL-15 in human and rat pancreatic islet cells, and in islets from pre-diabetic NOD mice. Diabetologia46 :255 eC266,2003

    Eizirik DL, Mandrup-Poulsen T: A choice of death—the signal-transduction of immune-mediated -cell apoptosis. Diabetologia44 :2115 eC2133,2001

    Pavlovic D, Chen MC, Gysemans CA, Mathieu C, Eizirik DL: The role of interferon regulatory factor-1 in cytokine-induced mRNA expression and cell death in murine pancreatic -cells. Eur Cytokine Netw10 :403 eC412,1999

    Liu D, Pavlovic D, Chen MC, Flodstrom M, Sandler S, Eizirik DL: Cytokines induce apoptosis in -cells isolated from mice lacking the inducible isoform of nitric oxide synthase (iNOSeC/eC). Diabetes49 :1116 eC1122,2000

    Rasschaert J, Liu D, Kutlu B, Cardozo AK, Kruhoffer M, Orntoft TF, Eizirik DL: Global profiling of double stranded RNA- and IFN--induced genes in rat pancreatic cells. Diabetologia46 :1641 eC1657,2003

    Levy DE, Darnell JE Jr: Stats: transcriptional control and biological impact. Nat Rev Mol Cell Biol3 :651 eC662,2002

    Kovarik P, Mangold M, Ramsauer K, Heidari H, Steinborn R, Zotter A, Levy DE, Muller M, Decker T: Specificity of signaling by STAT1 depends on SH2 and C-terminal domains that regulate Ser727 phosphorylation, differentially affecting specific target gene expression. EMBO J20 :91 eC100,2001

    Stephanou A, Latchman DS: STAT-1: a novel regulator of apoptosis. Int J Exp Pathol84 :239 eC244,2003

    Yamada K, Miyajima E, Nonaka K: Inhibition of cytokine-induced MHC class II but not class I molecule expression on mouse islet cells by niacinamide and 3-aminobenzamide. Diabetes39 :1125 eC1130,1990

    Thomas HE, Parker JL, Schreiber RD, Kay TW: IFN- action on pancreatic cells causes class I MHC upregulation but not diabetes. J Clin Invest102 :1249 eC1257,1998

    Lee CK, Gimeno R, Levy DE: Differential regulation of constitutive major histocompatibility complex class I expression in T and B lymphocytes. J Exp Med190 :1451 eC1464,1999

    Prieto J, Kaaya EE, Juntti-Berggren L, Berggren PO, Sandler S, Biberfeld P, Patarroyo M: Induction of intercellular adhesion molecule-1 (CD54) on isolated mouse pancreatic cells by inflammatory cytokines. Clin Immunol Immunopathol65 :247 eC253,1992

    Walter MJ, Look DC, Tidwell RM, Roswit WT, Holtzman MJ: Targeted inhibition of interferon--dependent intercellular adhesion molecule-1 (ICAM-1) expression using dominant-negative Stat1. J Biol Chem272 :28582 eC28589,1997

    Darville MI, Eizirik DL: Regulation by cytokines of the inducible nitric oxide synthase promoter in insulin-producing cells. Diabetologia41 :1101 eC1108,1998

    Darville MI, Liu D, Chen MC, Eizirik DL: Molecular regulation of Fas expression in -cells. Diabetes50 :S83 ,2001

    Kumar A, Commane M, Flickinger TW, Horvath CM, Stark GR: Defective TNF--induced apoptosis in STAT1-null cells due to low constitutive levels of caspases. Science278 :1630 eC1632,1997

    Karlsen AE, Pavlovic D, Nielsen K, Jensen J, Andersen HU, Pociot F, Mandrup-Poulsen T, Eizirik DL, Nerup J: Interferon- induces interleukin-1 converting enzyme expression in pancreatic islets by an interferon regulatory factor-1-dependent mechanism. J Clin Endocrinol Metab85 :830 eC836,2000

    Li X, Leung S, Qureshi S, Darnell JE Jr, Stark GR: Formation of STAT1-STAT2 heterodimers and their role in the activation of IRF-1 gene transcription by interferon-alpha. J Biol Chem271 :5790 eC5794,1996

    Flodstrom M, Eizirik DL: Interferon--induced interferon regulatory factor-1 (IRF-1) expression in rodent and human islet cells precedes nitric oxide production. Endocrinology138 :2747 eC2753,1997

    Durbin JE, Hackenmiller R, Simon MC, Levy DE: Targeted disruption of the mouse Stat1 gene results in compromised innate immunity to viral disease. Cell84 :443 eC450,1996

    Gysemans CA, Waer M, Valckx D, Laureys JM, Mihkalsky D, Bouillon R, Mathieu C: Early graft failure of xenogeneic islets in NOD mice is accompanied by high levels of interleukin-1 and low levels of transforming growth factor- wild-type factor- mRNA in the grafts. Diabetes49 :1992 eC1997,2000

    Pipeleers DG, in’t Veld PA, Van de WM, Maes E, Schuit FC, Gepts W: A new in vitro model for the study of pancreatic A and B cells. Endocrinology117 :806 eC816,1985

    Chen MC, Proost P, Gysemans C, Mathieu C, Eizirik DL: Monocyte chemoattractant protein-1 is expressed in pancreatic islets from prediabetic NOD mice and in interleukin-1 -exposed human and rat islet cells. Diabetologia44 :325 eC332,2001

    Hoorens A, Van de CM, Kloppel G, Pipeleers D: Glucose promotes survival of rat pancreatic cells by activating synthesis of proteins which suppress a constitutive apoptotic program. J Clin Invest98 :1568 eC1574,1996

    Gysemans C, Waer M, Laureys J, Depovere J, Pipeleers D, Bouillon R, Mathieu C: Islet xenograft destruction in the hu-PBL-severe combined immunodeficient (SCID) mouse necessitates anti-CD3 preactivation of human immune cells. Clin Exp Immunol121 :557 eC565,2000

    Overbergh L, Giulietti A, Valckx D, Decallonne R, Bouillon R, Mathieu C: The use of real-time reverse transcriptase PCR for the quantification of cytokine gene expression. J Biomol Tech14 :33 eC43,2003

    Overbergh L, Valckx D, Waer M, Mathieu C: Quantification of murine cytokine mRNAs using real time quantitative reverse transcriptase PCR. Cytokine11 :305 eC312,1999

    Giulietti A, Overbergh L, Valckx D, Decallonne B, Bouillon R, Mathieu C: An overview of real-time quantitative PCR: applications to quantify cytokine gene expression. Methods25 :386 eC401,2001

    Chong MM, Thomas HE, Kay TW: -Interferon signaling in pancreatic -cells is persistent but can be terminated by overexpression of suppressor of cytokine signaling-1. Diabetes50 :2744 eC2751,2001

    Karlsen AE, Ronn SG, Lindberg K, Johannesen J, Galsgaard ED, Pociot F, Nielsen JH, Mandrup-Poulsen T, Nerup J, Billestrup N: Suppressor of cytokine signaling 3 (SOCS-3) protects -cells against interleukin-1- and interferon--mediated toxicity. Proc Natl Acad Sci U S A98 :12191 eC12196,2001

    Flodstrom-Tullberg M, Yadav D, Hagerkvist R, Tsai D, Secrest P, Stotland A, Sarvetnick N: Target cell expression of suppressor of cytokine signaling-1 prevents diabetes in the NOD mouse. Diabetes52 :2696 eC2700,2003

    Baetz A, Frey M, Heeg K, Dalpke AH: Suppressor of cytokine signaling (SOCS) proteins indirectly regulate toll-like receptor signaling in innate immune cells. J Biol Chem24 :54708 eC54715,2004

    Karlsen AE, Heding PE, Frobose H, Ronn SG, Kruhoffer M, Orntoft TF, Darville M, Eizirik DL, Pociot F, Nerup J, Mandrup-Poulsen T, Billestrup N: Suppressor of cytokine signalling (SOCS)-3 protects cells against IL-1-mediated toxicity through inhibition of multiple nuclear factor-B-regulated proapoptotic pathways. Diabetologia47 :1998 eC2011,2004

    Emanuelli B, Glondu M, Filloux C, Peraldi P, Van Obberghen E: The potential role of SOCS-3 in the interleukin-1-induced desensitization of insulin signaling in pancreatic -cells. Diabetes53 :S97 eCS103,2004

    Tanaka N, Kawakami T, Taniguchi T: Recognition DNA sequences of interferon regulatory factor 1 (IRF-1) and IRF-2, regulators of cell growth and the interferon system. Mol Cell Biol13 :4531 eC4538,1993

    Hu J, Roy SK, Shapiro PS, Rodig SR, Reddy SP, Platanias LC, Schreiber RD, Kalvakolanu DV: ERK1 and ERK2 activate CCAAAT/enhancer-binding protein--dependent gene transcription in response to interferon-gamma. J Biol Chem276 :287 eC297,2001

    Deb A, Haque SJ, Mogensen T, Silverman RH, Williams BR: RNA-dependent protein kinase PKR is required for activation of NF-B by IFN- in a STAT1-independent pathway. J Immunol166 :6170 eC6180,2001

    Chesler DA, Dodard C, Lee GY, Levy DE, Reiss CS: Interferon--induced inhibition of neuronal vesicular stomatitis virus infection is STAT1 dependent. J Neurovirol10 :57 eC63,2004

    Lee CK, Rao DT, Gertner R, Gimeno R, Frey AB, Levy DE: Distinct requirements for IFNs and STAT1 in NK cell function. J Immunol165 :3571 eC3577,2000

    Lee CK, Smith E, Gimeno R, Gertner R, Levy DE: STAT1 affects lymphocyte survival and proliferation partially independent of its role downstream of IFN-. J Immunol164 :1286 eC1292,2000

    Ramana CV, Grammatikakis N, Chernov M, Nguyen H, Goh KC, Williams BR, Stark GR: Regulation of c-myc expression by IFN- through Stat1-dependent and independent pathways. EMBO J19 :263 eC272,2000

    Gil MP, Bohn E, O’Guin AK, Ramana CV, Levine B, Stark GR, Virgin HW, Schreiber RD: Biologic consequences of Stat1-independent IFN signaling. Proc Natl Acad Sci U S A98 :6680 eC6685,2001

    Ramana CV, Gil MP, Schreiber RD, Stark GR: Stat1-dependent and -independent pathways in IFN--dependent signaling. Trends Immunol23 :96 eC101,2002

    Gysemans CA, Pavlovic D, Bouillon R, Eizirik DL, Mathieu C: Dual role of interferon- signalling pathway in sensitivity of pancreatic cells to immune destruction. Diabetologia44 :567 eC574,2001(Conny A. Gysemans, Lauren)