β-cell dysfunction due to increased ER stress in a stem cell model of Wolfram syndrome

Linshan Shang, Haiqing Hua, Kylie Foo, Hector Martinez, Kazuhisa Watanabe, Matthew Zimmer, David J Kahler, Matthew Freeby, Wendy Chung, Charles LeDuc, Robin Goland, Rudolph L Leibel, Dieter Egli, Linshan Shang, Haiqing Hua, Kylie Foo, Hector Martinez, Kazuhisa Watanabe, Matthew Zimmer, David J Kahler, Matthew Freeby, Wendy Chung, Charles LeDuc, Robin Goland, Rudolph L Leibel, Dieter Egli

Abstract

Wolfram syndrome is an autosomal recessive disorder caused by mutations in WFS1 and is characterized by insulin-dependent diabetes mellitus, optic atrophy, and deafness. To investigate the cause of β-cell failure, we used induced pluripotent stem cells to create insulin-producing cells from individuals with Wolfram syndrome. WFS1-deficient β-cells showed increased levels of endoplasmic reticulum (ER) stress molecules and decreased insulin content. Upon exposure to experimental ER stress, Wolfram β-cells showed impaired insulin processing and failed to increase insulin secretion in response to glucose and other secretagogues. Importantly, 4-phenyl butyric acid, a chemical protein folding and trafficking chaperone, restored normal insulin synthesis and the ability to upregulate insulin secretion. These studies show that ER stress plays a central role in β-cell failure in Wolfram syndrome and indicate that chemical chaperones might have therapeutic relevance under conditions of ER stress in Wolfram syndrome and other forms of diabetes.

Figures

Figure 1
Figure 1
iPS cells from Wolfram subjects differentiated into insulin-producing cells. (A) Diagram of WFS1 structure showing the mutation sites and Sanger sequencing profiles in the four Wolfram subjects described here. Arrows indicate the four deleted nucleotides (CTCT). (B) Immunostaining of Wolfram cultures differentiated into endoderm (SOX17), pancreatic endoderm (PDX1), and C-peptide–positive cells. Scale bar, 50 μm. (C) Differentiation efficiency in controls and WFS1 mutant cells determined by imaging (n = 10 for each of three independent experiments). (D) Immunostaining of WFS1, glucagon, and C-peptide in iPS-derived pancreatic cell cultures at d15 of differentiation. Scale bar, 20 μm.
Figure 2
Figure 2
UPR-dependent reduction of insulin content in Wolfram β-cells. (A) Insulin mRNA levels in control and Wolfram β-cells normalized to TBP (TATA-binding protein) mRNA levels and to the number of C-peptide–positive cells used for analysis. (B) Insulin protein content in the β-cells normalized to the number of C-peptide–positive cells. Error bars represent three independent experiments with three replicates in each experiment. *P < 0.05 for vehicle of WS vs. control cells. (C) Transmission electron microscope images of β-cells (scale bar, 2 μm). (D) Quantification of granule numbers per cell. Three independent differentiation experiments with n = 20 sections for each subject of each experiment. *P < 0.05 for WS-1 vs. control or WS-1 cells treated with 4PBA for 7 days or WS-1 cells carrying wild-type WFS1 expression vector. Ctrl, control.
Figure 3
Figure 3
Increased UPR pathway activity and increased sensitivity of the ER-to-ER stress in Wolfram β-cells. (A) Proinsulin-to-insulin ratio determined by ELISA; n = 6 for each of two independent experiments. (BF) Quantification of components of the UPR displayed as fold change compared with vehicle-treated control cells (set to 1). Western blot analysis and quantification for (B) nuclear ATF6α (n = 3) and (C) phosphorylated eIF2α (n = 3) in β-cells. (DF) Quantitative PCR for mRNA levels of sXBP-1, ATF4 in β-cells, and GRP78 in iPS cells (n = 3 for each experiment). *P < 0.05. (G) Transmission electron microscope image showing ER morphology in control and Wolfram cells after 12-h treatment with 10 nmol/L TG. Arrows point to the ER. Scale bar, 500 nm. β-Cells were treated with TG 10 nmol/L for 12 h, and 1 mmol/L 4PBA or 1 mmol/L TUDCA was added 1 h prior to and during TG treatment. 4PBA 7d refers to treatment from day 9 to day 15 of β-cell differentiation.
Figure 4
Figure 4
Differential effect of ER stress on insulin secretion in Wolfram and control cells. (A) Fold change of human C-peptide secretion in response to indicated secretagogues. Cells were incubated in 5.6 mmol/L glucose for 1 h followed by 16.9 mmol/L glucose, 15 mmol/L arginine, 30 mmol/L potassium, or 1 mmol/L DBcAMP + 16.9 mmol/L glucose for an additional hour. Fold change of human C-peptide secretion was calculated as the ratio of C-peptide concentration occurring in secretagogue-stimulated media over the concentration in 5.6 mmol/L glucose. Results represent three independent experiments, with n = 3 for each experiment. *P < 0.05 of TG vs. vehicle; #P < 0.05 of TG + 4PBA vs. TG. (B) Fold change of human C-peptide secretion to 16.9 mmol/L glucose stimulation; n = 3 for each of two independent experiments. (C) Fold change of human C-peptide secretion in response to 30 mmol/L potassium stimulation upon TM treatment. Results represent three independent experiments, with n = 3 for each experiment. (D) Fold change of human C-peptide and glucagon in control and Wolfram cells under indicated conditions; n = 3 for each of three independent experiments. Arg, arginine.
Figure 5
Figure 5
ER stress and reduced glucose-stimulated insulin secretion of Wolfram cells in vivo. (A) Immunohistochemistry for insulin and urocortin 3 in transplants. Scale bar, 20 μm. (B) Fold change of human C-peptide in the sera of mice transplanted with human islets and control and Wolfram cells after intraperitoneal glucose injection. Bars show the median. (C) Human C-peptide levels in the sera of transplanted mice over a 1-month time period. Error bars show SD. (D) Fold change of human C-peptide in the sera of transplanted mice over a 1-month time period. Error bars show SD. Immunohistochemistry for insulin and (E) ATF6α and (F) CHOP in transplants. Scale bar, 20 μm. C-PEP, C-peptide.

References

    1. Inoue H, Tanizawa Y, Wasson J, et al. A gene encoding a transmembrane protein is mutated in patients with diabetes mellitus and optic atrophy (Wolfram syndrome). Nat Genet 1998;20:143–148
    1. Barrett TG, Bundey SE. Wolfram (DIDMOAD) syndrome. J Med Genet 1997;34:838–841
    1. Karasik A, O’Hara C, Srikanta S, et al. Genetically programmed selective islet beta-cell loss in diabetic subjects with Wolfram’s syndrome. Diabetes Care 1989;12:135–138
    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. Ishihara H, Takeda S, Tamura A, et al. Disruption of the WFS1 gene in mice causes progressive beta-cell loss and impaired stimulus-secretion coupling in insulin secretion. Hum Mol Genet 2004;13:1159–1170
    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. Takeda K, Inoue H, Tanizawa Y, et al. WFS1 (Wolfram syndrome 1) gene product: predominant subcellular localization to endoplasmic reticulum in cultured cells and neuronal expression in rat brain. Hum Mol Genet 2001;10:477–484
    1. Takei D, Ishihara H, Yamaguchi S, et al. WFS1 protein modulates the free Ca(2+) concentration in the endoplasmic reticulum. FEBS Lett 2006;580:5635–5640
    1. Yurimoto S, Hatano N, Tsuchiya M, et al. Identification and characterization of wolframin, the product of the wolfram syndrome gene (WFS1), as a novel calmodulin-binding protein. Biochemistry 2009;48:3946–3955
    1. Fonseca SG, Urano F, Weir GC, Gromada J, Burcin M. Wolfram syndrome 1 and adenylyl cyclase 8 interact at the plasma membrane to regulate insulin production and secretion. Nat Cell Biol 2012;14:1105–1112
    1. Fonseca SG, Fukuma M, Lipson KL, et al. WFS1 is a novel component of the unfolded protein response and maintains homeostasis of the endoplasmic reticulum in pancreatic beta-cells. J Biol Chem 2005;280:39609–39615
    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. Fusaki N, Ban H, Nishiyama A, Saeki K, Hasegawa M. Efficient induction of transgene-free human pluripotent stem cells using a vector based on Sendai virus, an RNA virus that does not integrate into the host genome. Proc Jpn Acad, Ser B, Phys Biol Sci 2009;85:348–362
    1. Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007;131:861–872
    1. Hua H, Shang L, Martinez H, et al. iPSC-derived β cells model diabetes due to glucokinase deficiency. J Clin Invest 2013;123:3146–3153
    1. Lee JH, Won SM, Suh J, et al. Induction of the unfolded protein response and cell death pathway in Alzheimer’s disease, but not in aged Tg2576 mice. Exp Mol Med 2010;42:386–394
    1. Merquiol E, Uzi D, Mueller T, et al. HCV causes chronic endoplasmic reticulum stress leading to adaptation and interference with the unfolded protein response. PLoS ONE 2011;6:e24660.
    1. Lu J, Wang Q, Huang L, et al. Palmitate causes endoplasmic reticulum stress and apoptosis in human mesenchymal stem cells: prevention by AMPK activator. Endocrinology 2012;153:5275–5284
    1. Szot GL, Koudria P, Bluestone JA. Transplantation of pancreatic islets into the kidney capsule of diabetic mice. J Vis Exp 2007;9:404.
    1. Colosimo A, Guida V, Rigoli L, et al. Molecular detection of novel WFS1 mutations in patients with Wolfram syndrome by a DHPLC-based assay. Hum Mutat 2003;21:622–629
    1. Chen AE, Egli D, Niakan K, et al. Optimal timing of inner cell mass isolation increases the efficiency of human embryonic stem cell derivation and allows generation of sibling cell lines. Cell Stem Cell 2009;4:103–106
    1. Hetz C. The unfolded protein response: controlling cell fate decisions under ER stress and beyond. Nat Rev Mol Cell Biol 2012;13:89–102
    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. Wong WL, Brostrom MA, Kuznetsov G, Gmitter-Yellen D, Brostrom CO. Inhibition of protein synthesis and early protein processing by thapsigargin in cultured cells. Biochem J 1993;289:71–79
    1. Kozutsumi Y, Segal M, Normington K, Gething MJ, Sambrook J. The presence of malfolded proteins in the endoplasmic reticulum signals the induction of glucose-regulated proteins. Nature 1988;332:462–464
    1. Ma Y, Hendershot LM. ER chaperone functions during normal and stress conditions. J Chem Neuroanat 2004;28:51–65
    1. Yam GH, Gaplovska-Kysela K, Zuber C, Roth J. Sodium 4-phenylbutyrate acts as a chemical chaperone on misfolded myocilin to rescue cells from endoplasmic reticulum stress and apoptosis. Invest Ophthalmol Vis Sci 2007;48:1683–1690
    1. de Almeida SF, Picarote G, Fleming JV, Carmo-Fonseca M, Azevedo JE, de Sousa M. Chemical chaperones reduce endoplasmic reticulum stress and prevent mutant HFE aggregate formation. J Biol Chem 2007;282:27905–27912
    1. Berger E, Haller D. Structure-function analysis of the tertiary bile acid TUDCA for the resolution of endoplasmic reticulum stress in intestinal epithelial cells. Biochem Biophys Res Commun 2011;409:610–615
    1. Ashcroft SJ. Glucoreceptor mechanisms and the control of insulin release and biosynthesis. Diabetologia 1980;18:5–15
    1. Herchuelz A, Lebrun P, Boschero AC, Malaisse WJ. Mechanism of arginine-stimulated Ca2+ influx into pancreatic B cell. Am J Physiol 1984;246:E38–E43
    1. Furman B, Ong WK, Pyne NJ. Cyclic AMP signaling in pancreatic islets. Adv Exp Med Biol 2010;654:281–304
    1. Matthews EK, Shotton PA. Efflux of 86Rb from rat and mouse pancreatic islets: the role of membrane depolarization. Br J Pharmacol 1984;83:831–839
    1. Oyadomari S, Mori M. Roles of CHOP/GADD153 in endoplasmic reticulum stress. Cell Death Differ 2004;11:381–389
    1. Oslowski CM, Hara T, O’Sullivan-Murphy B, et al. Thioredoxin-interacting protein mediates ER stress-induced β cell death through initiation of the inflammasome. Cell Metab 2012;16:265–273
    1. Bertuzzi F, Berra C, Socci C, Davalli AM, Pozza G, Pontiroli AE. Insulin and glucagon release of human islets in vitro: effects of chronic exposure to glucagon. J Endocrinol 1997;152:239–243
    1. Blum B, Hrvatin SS, Schuetz C, Bonal C, Rezania A, Melton DA. Functional beta-cell maturation is marked by an increased glucose threshold and by expression of urocortin 3. Nat Biotechnol 2012;30:261–264
    1. Tersey SA, Nishiki Y, Templin AT, et al. Islet β-cell endoplasmic reticulum stress precedes the onset of type 1 diabetes in the nonobese diabetic mouse model. Diabetes 2012;61:818–827
    1. Marhfour I, Lopez XM, Lefkaditis D, et al. Expression of endoplasmic reticulum stress markers in the islets of patients with type 1 diabetes. Diabetologia 2012;55:2417–2420
    1. Ozcan U, Cao Q, Yilmaz E, et al. Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science 2004;306:457–461
    1. Laybutt DR, Preston AM, Akerfeldt MC, et al. Endoplasmic reticulum stress contributes to beta cell apoptosis in type 2 diabetes. Diabetologia 2007;50:752–763
    1. Shah SC, Malone JI, Simpson NE. A randomized trial of intensive insulin therapy in newly diagnosed insulin-dependent diabetes mellitus. N Engl J Med 1989;320:550–554
    1. Gastaldelli A, Ferrannini E, Miyazaki Y, Matsuda M, Mari A, DeFronzo RA. Thiazolidinediones improve beta-cell function in type 2 diabetic patients. Am J Physiol Endocrinol Metab 2007;292:E871–E883
    1. Dixon JB, Dixon AF, O’Brien PE. Improvements in insulin sensitivity and beta-cell function (HOMA) with weight loss in the severely obese. Homeostatic model assessment. Diabet Med 2003;20:127–134
    1. Sandhu MS, Weedon MN, Fawcett KA, et al. Common variants in WFS1 confer risk of type 2 diabetes. Nat Genet 2007;39:951–953

Source: PubMed

3
S'abonner