Islet β-cell endoplasmic reticulum stress precedes the onset of type 1 diabetes in the nonobese diabetic mouse model

Sarah A Tersey, Yurika Nishiki, Andrew T Templin, Susanne M Cabrera, Natalie D Stull, Stephanie C Colvin, Carmella Evans-Molina, Jenna L Rickus, Bernhard Maier, Raghavendra G Mirmira, Sarah A Tersey, Yurika Nishiki, Andrew T Templin, Susanne M Cabrera, Natalie D Stull, Stephanie C Colvin, Carmella Evans-Molina, Jenna L Rickus, Bernhard Maier, Raghavendra G Mirmira

Abstract

Type 1 diabetes is preceded by islet β-cell dysfunction, but the mechanisms leading to β-cell dysfunction have not been rigorously studied. Because immune cell infiltration occurs prior to overt diabetes, we hypothesized that activation of inflammatory cascades and appearance of endoplasmic reticulum (ER) stress in β-cells contributes to insulin secretory defects. Prediabetic nonobese diabetic (NOD) mice and control diabetes-resistant NOD-SCID and CD1 strains were studied for metabolic control and islet function and gene regulation. Prediabetic NOD mice were relatively glucose intolerant and had defective insulin secretion with elevated proinsulin:insulin ratios compared with control strains. Isolated islets from NOD mice displayed age-dependent increases in parameters of ER stress, morphologic alterations in ER structure by electron microscopy, and activation of nuclear factor-κB (NF-κB) target genes. Upon exposure to a mixture of proinflammatory cytokines that mimics the microenvironment of type 1 diabetes, MIN6 β-cells displayed evidence for polyribosomal runoff, a finding consistent with the translational initiation blockade characteristic of ER stress. We conclude that β-cells of prediabetic NOD mice display dysfunction and overt ER stress that may be driven by NF-κB signaling, and strategies that attenuate pathways leading to ER stress may preserve β-cell function in type 1 diabetes.

Figures

FIG. 1.
FIG. 1.
Glucose homeostasis in female CD1, NOD-SCID, and prediabetic NOD mice. A: Diabetes incidence in female NOD mice by age. Diabetes was defined as blood glucose >300 mg/dL on two consecutive measurements. n = 57 mice. B: Blood glucose values, as measured following an overnight fast in 6-, 8-, and 10-week-old mice. C, E, and G: Results of glucose tolerance tests and their corresponding area under the curve (AUC) analyses in mice at 6 (C), 8 (E), and 10 (G) weeks of age following intraperitoneal injections of glucose (2 mg/kg). n = 5–6 mice per group. D, F, and H: Serum insulin values in mice at 6 (D), 8 (F), and 10 (H) weeks of age at the indicated times following an intraperitoneal injection of glucose (2 g/kg). n = 3–4 mice per group. *P < 0.05 compared with the corresponding time value for CD1 mice. #P < 0.05 compared with the corresponding time value for NOD-SCID mice.
FIG. 2.
FIG. 2.
GSIS from islets of female CD1, NOD-SCID, and prediabetic NOD mice. Shown are results of GSIS studies from islets of female CD1, NOD-SCID, and prediabetic NOD mice at 6 (A), 8 (B), and 10 (C) weeks of age exposed to low (2.5 mmol/L) and high (25 mmol/L) glucose concentrations. n = 3 independent, pooled islet isolations per group. *P < 0.05 compared with the corresponding glucose value for CD1 mice. #P < 0.05 compared with the corresponding glucose value for NOD-SCID mice.
FIG. 3.
FIG. 3.
Secreted hormone levels and electron microscopic imaging of female prediabetic mice. A: Insulin levels in nonfasting 6-, 8-, and 10-week-old female CD1, NOD-SCID, and prediabetic NOD mice. B: Proinsulin levels in nonfasting 6-, 8-, and 10-week-old CD1, NOD-SCID, and prediabetic NOD mice. C: Proinsulin:insulin ratios in nonfasting 6-, 8-, and 10-week-old CD1, NOD-SCID, and prediabetic NOD mice. D: Representative low- and high-magnification transmission electron microscopic images of β-cells from 10-week-old NOD-SCID and prediabetic NOD mice. Arrows indicate ER. N, nucleus; SG, secretory granule. n = 4–5 mice in panels AC. *P < 0.05 compared with the corresponding value for CD1 mice. #P < 0.05 compared with the corresponding value for NOD-SCID mice.
FIG. 4.
FIG. 4.
Transcript levels of ER stress–responsive genes in islets of female CD1, NOD-SCID, and prediabetic NOD mice. Islets from 6-, 8-, and 10-week-old female CD1, NOD-SCID, and prediabetic NOD mice were isolated and processed for total RNA. Total RNA from each age and strain was then subjected to quantitative real-time RT-PCR for selected genes; the values were corrected for Actb mRNA levels and are displayed relative to 6-week-old CD1 islet data. Shown are results for Pdx1 (A), Ins1/2 (B), pre-Ins2 (C), Bip (D), Xbp1 (E), Xbp1s (F), Chop (G), Atf4 (H), Wfs1 (I), and Serca2b (J). n = 3 independent, pooled islet isolations per group. *P < 0.05 compared with the corresponding value for CD1 mice. #P < 0.05 compared with the corresponding value for NOD-SCID mice.
FIG. 5.
FIG. 5.
CHOP expression in islets and pancreata of prediabetic NOD mice. A: Results of immunoblot analysis for glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and CHOP from islet extracts from each of two 10-week-old female CD1, NOD-SCID, and prediabetic NOD mice. B: Fixed pancreatic sections from 10-week-old female NOD-SCID and prediabetic NOD mice were subjected to immunofluorescence staining for insulin (green) and CHOP (red). Shown are representative islets from each strain. Arrows in the right panel identify CHOP+/insulin+ cells. (A high-quality digital representation of this figure is available in the online issue.)
FIG. 6.
FIG. 6.
Transcript levels of NF-κB target genes in islets of female NOD-SCID and prediabetic NOD mice. Islets from 10-week-old female NOD-SCID and prediabetic NOD mice were isolated and processed for total RNA. Total RNA from each strain was then subjected to quantitative real-time RT-PCR for the NF-κB target genes shown at the top of each panel; the values were corrected for Actb mRNA levels and are displayed relative to NOD-SCID islet data. n = 3 independent, pooled islet isolations per group.
FIG. 7.
FIG. 7.
Translational profiling of MIN6 cells. MIN6 cells were incubated with thapsigargin for 6 h or cytokines for the indicated times, then cellular extracts were harvested and subjected to centrifugation through a 10–50% sucrose gradient. Profiles through the gradient were measured by absorbance at 254 nm. A: Profiles of MIN6 cells untreated or exposed to thapsigargin. Positions of the 40S and 60S subunits, 80S monosome, and polyribosomes are indicated, and the P/M ratios are shown (as calculated from areas under the respective curves). B: Profiles of MIN6 cells untreated or exposed to a mixture of cytokines (IL-1β, TNF-α, and IFN-γ) for the indicated times are shown, as are the respective P/M ratios. C: Results of SDS-PAGE analysis of 35S-labeled protein in MIN6 cells that were untreated or exposed to cytokines for the indicated times. D: Profiles of MIN6 cells untreated and exposed to a mixture of cytokines or to a mixture of cytokines + 100 μmol/L l-NMMA. All data shown are from representative experiments performed on 2–3 occasions.

References

    1. Charré S, Rosmalen JG, Pelegri C, et al. Abnormalities in dendritic cell and macrophage accumulation in the pancreas of nonobese diabetic (NOD) mice during the early neonatal period. Histol Histopathol 2002;17:393–401
    1. McDuffie M, Maybee NA, Keller SR, et al. Nonobese diabetic (NOD) mice congenic for a targeted deletion of 12/15-lipoxygenase are protected from autoimmune diabetes. Diabetes 2008;57:199–208
    1. Lehuen A, Diana J, Zaccone P, Cooke A. Immune cell crosstalk in type 1 diabetes. Nat Rev Immunol 2010;10:501–513
    1. Atkinson MA, Bluestone JA, Eisenbarth GS, et al. How does type 1 diabetes develop?: the notion of homicide or β-cell suicide revisited. Diabetes 2011;60:1370–1379
    1. Strandell E, Eizirik DL, Sandler S. Reversal of beta-cell suppression in vitro in pancreatic islets isolated from nonobese diabetic mice during the phase preceding insulin-dependent diabetes mellitus. J Clin Invest 1990;85:1944–1950
    1. Leiter EH, Prochazka M, Coleman DL. The non-obese diabetic (NOD) mouse. Am J Pathol 1987;128:380–383
    1. Keskinen P, Korhonen S, Kupila A, et al. First-phase insulin response in young healthy children at genetic and immunological risk for type I diabetes. Diabetologia 2002;45:1639–1648
    1. Ferrannini E, Mari A, Nofrate V, Sosenko JM, Skyler JS; DPT-1 Study Group Progression to diabetes in relatives of type 1 diabetic patients: mechanisms and mode of onset. Diabetes 2010;59:679–685
    1. Sreenan S, Pick AJ, Levisetti M, Baldwin AC, Pugh W, Polonsky KS. Increased beta-cell proliferation and reduced mass before diabetes onset in the nonobese diabetic mouse. Diabetes 1999;48:989–996
    1. Ize-Ludlow D, Lightfoot YL, Parker M, et al. Progressive erosion of β-cell function precedes the onset of hyperglycemia in the NOD mouse model of type 1 diabetes. Diabetes 2011;60:2086–2091
    1. Chambers KT, Unverferth JA, Weber SM, Wek RC, Urano F, Corbett JA. The role of nitric oxide and the unfolded protein response in cytokine-induced beta-cell death. Diabetes 2008;57:124–132
    1. Oyadomari S, Takeda K, Takiguchi M, et al. Nitric oxide-induced apoptosis in pancreatic beta cells is mediated by the endoplasmic reticulum stress pathway. Proc Natl Acad Sci USA 2001;98:10845–10850
    1. Cardozo AK, Ortis F, Storling J, et al. Cytokines downregulate the sarcoendoplasmic reticulum pump Ca2+ ATPase 2b and deplete endoplasmic reticulum Ca2+, leading to induction of endoplasmic reticulum stress in pancreatic beta-cells. Diabetes 2005;54:452–461
    1. Satoh T, Abiru N, Kobayashi M, et al. CHOP deletion does not impact the development of diabetes but suppresses the early production of insulin autoantibody in the NOD mouse. Apoptosis 2011;16:438–448
    1. Maier B, Ogihara T, Trace AP, et al. The unique hypusine modification of eIF5A promotes islet beta cell inflammation and dysfunction in mice. J Clin Invest 2010;120:2156–2170
    1. Evans-Molina C, Robbins RD, Kono T, et al. Peroxisome proliferator-activated receptor gamma activation restores islet function in diabetic mice through reduction of ER stress and maintenance of euchromatin structure. Mol Cell Biol 2009;29:2053–2067
    1. Iype T, Francis J, Garmey JC, et al. Mechanism of insulin gene regulation by the pancreatic transcription factor Pdx-1: application of pre-mRNA analysis and chromatin immunoprecipitation to assess formation of functional transcriptional complexes. J Biol Chem 2005;280:16798–16807
    1. Lipson KL, Fonseca SG, Ishigaki S, et al. Regulation of insulin biosynthesis in pancreatic beta cells by an endoplasmic reticulum-resident protein kinase IRE1. Cell Metab 2006;4:245–254
    1. Kulkarni RN, Roper MG, Dahlgren G, et al. Islet secretory defect in insulin receptor substrate 1 null mice is linked with reduced calcium signaling and expression of sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA)-2b and -3. Diabetes 2004;53:1517–1525
    1. Sachdeva MM, Claiborn KC, Khoo C, et al. Pdx1 (MODY4) regulates pancreatic beta cell susceptibility to ER stress. Proc Natl Acad Sci USA 2009;106:19090–19095
    1. Robbins RD, Tersey SA, Ogihara T, et al. Inhibition of deoxyhypusine synthase enhances islet beta cell function and survival in the setting of endoplasmic reticulum stress and type 2 diabetes. J Biol Chem 2010;285:39943–39952
    1. Palam LR, Baird TD, Wek RC. Phosphorylation of eIF2 facilitates ribosomal bypass of an inhibitory upstream ORF to enhance CHOP translation. J Biol Chem 2011;286:10939–10949
    1. Evans-Molina C, Garmey JC, Ketchum RJ, Brayman KL, Deng S, Mirmira RG. Glucose regulation of insulin gene transcription and pre-mRNA processing in human islets. Diabetes 2007;56:827–835
    1. Yoshida H, Matsui T, Yamamoto A, Okada T, Mori K. XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell 2001;107:881–891
    1. Calfon M, Zeng H, Urano F, et al. IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA. Nature 2002;415:92–96
    1. Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol 2007;8:519–529
    1. Fonseca SG, Ishigaki S, Oslowski CM, et al. Wolfram syndrome 1 gene negatively regulates ER stress signaling in rodent and human cells. J Clin Invest 2010;120:744–755
    1. Riggs AC, Bernal-Mizrachi E, Ohsugi M, et al. Mice conditionally lacking the Wolfram gene in pancreatic islet beta cells exhibit diabetes as a result of enhanced endoplasmic reticulum stress and apoptosis. Diabetologia 2005;48:2313–2321
    1. Hatanaka M, Tanabe K, Yanai A, et al. Wolfram syndrome 1 gene (WFS1) product localizes to secretory granules and determines granule acidification in pancreatic beta-cells. Hum Mol Genet 2011;20:1274–1284
    1. Jiang HY, Wek RC. Phosphorylation of the alpha-subunit of the eukaryotic initiation factor-2 (eIF2alpha) reduces protein synthesis and enhances apoptosis in response to proteasome inhibition. J Biol Chem 2005;280:14189–14202
    1. Back SH, Scheuner D, Han J, et al. Translation attenuation through eIF2alpha phosphorylation prevents oxidative stress and maintains the differentiated state in beta cells. Cell Metab 2009;10:13–26
    1. Teske BF, Baird TD, Wek RC. Methods for analyzing eIF2 kinases and translational control in the unfolded protein response. Methods Enzymol 2011;490:333–356
    1. Steffes MW, Sibley S, Jackson M, Thomas W. Beta-cell function and the development of diabetes-related complications in the diabetes control and complications trial. Diabetes Care 2003;26:832–836
    1. Hotamisligil GS. Endoplasmic reticulum stress and the inflammatory basis of metabolic disease. Cell 2010;140:900–917
    1. Wek RC, Jiang HY, Anthony TG. Coping with stress: eIF2 kinases and translational control. Biochem Soc Trans 2006;34:7–11
    1. Pirot P, Ortis F, Cnop M, et al. Transcriptional regulation of the endoplasmic reticulum stress gene chop in pancreatic insulin-producing cells. Diabetes 2007;56:1069–1077
    1. Akerfeldt MC, Howes J, Chan JY, et al. Cytokine-induced beta-cell death is independent of endoplasmic reticulum stress signaling. Diabetes 2008;57:3034–3044
    1. Meldolesi J, Pozzan T. The endoplasmic reticulum Ca2+ store: a view from the lumen. Trends Biochem Sci 1998;23:10–14
    1. Bygrave FL, Benedetti A. What is the concentration of calcium ions in the endoplasmic reticulum? Cell Calcium 1996;19:547–551
    1. Subasinghe W, Syed I, Kowluru A. Phagocyte-like NADPH oxidase promotes cytokine-induced mitochondrial dysfunction in pancreatic β-cells: evidence for regulation by Rac1. Am J Physiol Regul Integr Comp Physiol 2011;300:R12–R20
    1. Dahlén E, Dawe K, Ohlsson L, Hedlund G. Dendritic cells and macrophages are the first and major producers of TNF-alpha in pancreatic islets in the nonobese diabetic mouse. J Immunol 1998;160:3585–3593

Source: PubMed

3
구독하다