Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Apr 1.
Published in final edited form as: Curr Opin Cell Biol. 2010 Dec 16;23(2):207–215. doi: 10.1016/j.ceb.2010.11.005

The binary switch that controls the life and death decisions of ER stressed beta cells

Christine M Oslowski 1, Fumihiko Urano 1,2
PMCID: PMC3078183  NIHMSID: NIHMS256555  PMID: 21168319

Abstract

Diabetes mellitus is a group of common metabolic disorders defined by hyperglycemia. One of the most important factors contributing to hyperglycemia is dysfunction and death of β cells. Increasing experimental, clinical, and genetic evidence indicates that endoplasmic reticulum (ER) stress plays an important role in β cell dysfunction and death during the progression of type 1 and type 2 diabetes as well as genetic forms of diabetes such as Wolfram syndrome. The mechanisms of ER stress-mediated β cell dysfunction and death are complex and not homogenous. Here we review the recent key findings on the role of ER stress and the unfolded protein response (UPR) in β cells and the mechanisms of ER stress mediated β cell dysfunction and death. Complete understanding of these mechanisms will lead to novel therapeutic modalities for diabetes.

I. Introduction

Diabetes mellitus is a group of common metabolic disorders defined by hyperglycemia. One of the most important factors contributing to hyperglycemia is dysfunction and death of β cells [1,2]. Increasing experimental, clinical, and genetic evidence indicates that endoplasmic reticulum (ER) stress plays a role in β cell dysfunction and death during the progression of type 1 and type 2 diabetes as well as genetic forms of diabetes such as Wolfram syndrome [36]. ER stress activates a network of signaling pathways collectively termed the Unfolded Protein Response (UPR). The UPR primarily functions to mitigate ER stress, maintain β cell function, and promote β cell survival. However; in the disease state, the UPR initiates β cell dysfunction and apoptosis. In this article, we review recent key findings on ER stress in the β cell and the potential mechanisms of ER stress-mediated β cell dysfunction and death.

II. ER stress and the role of the Unfolded Protein Response in β cells

ER in the β cell

Pancreatic β cells produce and secrete insulin to control blood glucose levels. The ER of β cells plays a key role in the regulation of insulin production. The ER houses a specialized chemical and protein environment as well as quality control mechanisms to ensure the proper folding and processing of secretory proteins including insulin, establishing ER homeostasis. ER homeostasis is defined as the balance between the ER protein load and the ER folding capacity to handle this load. β cells often undergo conditions that stimulate insulin production and therefore a disruption to ER homeostasis. This disruption leads to the accumulation of unfolded and misfolded proinsulin in the ER lumen causing ER stress.

Three distinct functions of the UPR in β cells

ER stress is sensed by the luminal domains of three ER transmembrane proteins: Inositol Requiring 1 (IRE1), PKR-like ER kinase (PERK), and Activating Transcription Factor 6 (ATF6). These stress sensors subsequently become activated regulating a complex signaling cascade termed the unfolded protein response (UPR) [7]. The UPR regulates several downstream effectors with the following functions: adaptive response, feedback control, and cell fate regulation [3] (Figure 1). Initially the UPR triggers the adaptive response upregulating the expression of molecular chaperones and protein processing enzymes to increase folding and handling efficiency, attenuating translation and mediating mRNA degradation to reduce ER workload and prevent further accumulation of unfolded proteins, and increasing the expression of ER-associated protein degradation (ERAD) and autophagy components to promote clearance of unwanted proteins. These responses are important for β cells to mitigitate ER stress and restore ER homeostasis to ensure the proper production of high quality proteins such as insulin. As ER stress is attenuated, the UPR has built in feedback control mechanisms to turn off the UPR master regulators and their downstream effectors to prevent harmful UPR hyperactivation. The UPR also regulates the expression or activation of both survival and death factors [3]. The ultimate cell fate decision determined by the UPR will be discussed in detail later.

Figure 1. Homeostatic, Feedback, and Cell Fate Regulation by the UPR.

Figure 1

ER stress is sensed by three master regulators of the UPR: IRE1, PERK, and ATF6. These master regulators subsequently become activated, regulating a complex signaling cascade termed the unfolded protein response (UPR). The UPR regulates several downstream effectors with three distinct functions: adaptive response, feedback control, and cell fate regulation.

Causes of ER stress in β cells

ER stress is triggered by a variety of stimuli and subsequently activates the UPR in a unique manner for a given cell type. There are several physiological, environmental, and genetic causes of ER stress specific for β cells (Figure 2). Under high glucose conditions, increased insulin biosynthesis overwhelms the ER folding capacity leading to ER stress. Both acute (1–3 hours) and chronic (≥ 24 hours) high glucose (≥ 16.7 mM) induces IRE1α phosphorylation in β cells, leading to upregulation of some ER stress markers [8]. Transient activation of IRE1α by acute high glucose enhances proinsulin biosynthesis with minimal XBP-1 splicing, whereas prolonged activation of IRE1α by chronic high glucose leads to insulin mRNA degradation, XBP-1 splicing, and induction of other effectors leading to β cell dysfunction and death [8,9].

Figure 2. β cell specific inducers of ER stress.

Figure 2

There are several physiological, environmental, and genetic causes of ER stress specific for β cells. High glucose conditions cause an increase in proinsulin ER load. Free fatty acids and cytokines decrease ER calcium levels inhibiting the functions of ER chaperones and folding enzymes. Human IAPP spontaneously forms ER membrane-damaging sheets of amyloid. Mutant insulin misfolds and accumulates in the ER.

It has recently been shown that free fatty acids (FFAs), specifically long-chain FFAs, also induce ER stress in β cells perhaps by depleting ER calcium levels. [10,11] (Figure 2). Treatment of β cell lines with the saturated FFA, palmitate causes ER distension and activation of all three UPR stress sensors in β cell lines. The UPR may contribute to FFA-induced β cell death through CHOP upregulation and activation of JNK and caspase-12 [12].

Inflammatory cytokines, interleukin-1β (IL-1β) in combination with γ-interferon (IFN-γ), cause ER stress and UPR activation in β cells [13]. IL-1β with IFN-γ has been known to induce nitric oxide (NO) in β cells, leading to β cell dysfunction and death in type 1 diabetes [14]. It has been shown that NO-induced β cell apoptosis is mediated by ER stress [15]. NO production decreases expression of the sarcoendoplasmic reticulum pump Ca2+ ATPase 2b (SERCA2b), leading to a decrease in Ca2+ in the ER. This Ca2+ depletion causes a high level of ER stress in β cells [13,16]. This ER stress may have a role in cytokine induced-cell death in Type 1 diabetes.

Proinsulin misfolding causes ER stress in β cells. The Akita diabetes mouse model carries a cysteine 96 to tyrosine substitution in the insulin 2 gene [17], This mutation disrupts a disulfide bond of mature insulin causing incorrect folding of proinsulin in the ER, leading to ER stress and subsequent death of β cells [18,19]. It has recently been shown that mutations in the human insulin gene, specifically mutations occurring in critical regions of proinsulin folding can also cause this kind of pathology [20]. This is an autosomal dominant disorder, leading to permanent neonatal diabetes.

Islet amyloid polypetide (IAPP) is secreted along with insulin in β cells. Islet amyloid, composed of a 37-residue amyloidogenic polypeptide derived from IAPP, is commonly found in the pancreatic islets of Type 2 diabetes patients and may play a role in β cell dysfunction and death during the progression of the disease [21]. IAPP spontaneously forms ER membrane-damaging sheets of amyloid [22]. It has been shown that high expression of human IAPP induces ER stress and subsequent death of β cells [23,24]. Therefore, IAPP could be a link between ER stress and β cell death in type 2 diabetes.

Tolerable and Unresolvable ER stress in β cells

We propose that there are two types of ER stress conditions: tolerable and unresolvable. Under tolerable ER stress conditions, the UPR promotes β cell adaptation and survival. In contrast, under unresolvable ER stress conditions, the UPR induces β cell death (Figure 3A).

Figure 3. Tolerable and Unresolvable ER stress in β cells.

Figure 3

  1. There are two types of ER stress conditions: tolerable and unresolvable. Tolerable ER stress can be physiological, acute, and/or mild. Under tolerable ER stress conditions, the UPR is properly activated and regulated reducing ER stress and promoting β cell adaptation and survival. In contrast, unresolvable ER stress conditions are pathological, chronic, and/or severe. Under unresolvable ER stress conditions, the UPR is hyperactivated inducing β cell dysfunction and death.
  2. When β cells are exposed to physiological stimuli that induces tolerable ER stress, the UPR triggers transcription of adaptive and survival genes, attenuates translation, and promotes mild mRNA degradation to reduce ER stress. The UPR also promotes proinsulin biosynthesis. As ER homeostasis is restablished, BiP, Gadd34, P58IPK, RACK1 and WFS1 properly turn off different components of the UPR.
  3. When β cells are under pathological conditions that induce unresolvable ER stress, the UPR is hyperactivated triggering the activation of apoptotic pathways and upregulating apoptotic genes. The UPR also mediates degradation of mRNAs encoding proinsulin and ER homeostatic proteins. Under these conditions, the UPR bypasses feedback regulation.

β cells are exposed to physiological conditions that induce mild ER stress such as transient high glucose. These ER stress conditions are tolerable and the UPR can restore ER homeostasis, leveraging physiological functions and promoting cell survival. Therefore the UPR has a very important role in β cell physiology (Figure 3B).

Under unresolvable conditions in which the UPR fails to attenuate ER stress and restore ER homeostasis, the UPR induces β cell dysfunction and apoptosis. This unresolvable ER stress can be caused by genetic mutations such as insulin gene mutations as well as environmental factors such as inflammatory cytokines, free fatty acids and chronic high glucose (Figure 3C).

III. The UPR and the regulation of β cell fate under tolerable and unresolvable ER stress conditions

UPR as a binary switch regulating the life and death of β cells

The UPR determines β cell fate by behaving like a binary switch between life and death. The outcome of this switch depends on the nature of the ER stress condition, whether it is tolerable or unresolvable, the activation and regulation of the UPR stress sensors, and the balance of UPR regulated death and survival components (Figure 4). The mechanisms of this switch are not completely understood but recent findings have shed some clues. It is important to emphasize that the β cell UPR is unique and tailored to the needs of the β cell. Here we discuss the UPR signaling network and its regulation of cell fate under tolerable and unresolvable ER stress conditions in the context of the β cell.

Figure 4. The UPR determines β cell fate by behaving like a binary switch.

Figure 4

The UPR determines β cell fate by behaving like a binary switch between life and death. The outcome of this switch depends on the nature of the ER stress conditions, the activation and regulation of the UPR stress sensors, and the balance of UPR regulated death and survival components.

Regulation of Survival and Death effectors by the UPR master regulators

IRE1, PERK and ATF6 regulate both survival/adaptive and death effectors. Some of these effectors are simultaneously expressed and the balance between the components determines life and death. Other effectors are specifically turned on or upregulated depending on the ER stress conditions.

IRE1 is the most characterized master regulator of the UPR. Its baseline expression is higher in β cells compared to other cell types [8]. In response to tolerable ER stress conditions, IRE1 autophosphorylates and activates its RNase domain. This activated RNase domain cleaves X-box binding protein 1 (XBP1) mRNA into an active transcription factor. [2527]. Active XBP-1 regulates expression levels of homeostatic components of the UPR such as molecular chaperones and ERAD enzymes [28,29]. IRE1 also cleaves ER associated mRNAs to presumably reduce ER workload. Furthermore IRE1 has a key role in insulin biosynthesis. Under acute high glucose conditions, IRE1 is activated and enhances proinsulin biosynthesis, while disruption of IRE1 signaling leads to a suppression of proinsulin biosynthesis. Thus, IRE1 activation has a role in β cell function and promotes adaptation to physiological ER stress conditions.

In contrast, under unresolvable conditions, IRE1 becomes hyperactivated and as a result have several harmful outputs. IRE1 hyperactivation directly activates apoptotic pathways. In the presence of unresolvable ER stress, IRE1 recruits TRAF2 and phosphorylates ASK1. Activated ASK1 phorphorylates JNK which regulates BCL2 family members to induce apoptosis. [3033]. Prolonged IRE1 activation also induces the decay of mRNAs encoding ER homeostatic proteins, including PDI, and BiP, therefore decreasing the ER folding capacity promoting apoptosis [9,3436].

PERK was discovered as the second master regulator of the UPR [37]. Expression level of PERK is higher in β cells than in other cells. PERK has a role in β cell function under physiological ER stress conditions. PERK has been shown to regulate β cell proliferation and development [38,39]. Tissue- and cell-specific knockouts of the PERK gene in mice cause severe defects in fetal and neonatal β cell proliferation and differentiation, leading to low β cell mass, defects in proinsulin trafficking and insulin secretion, and diabetes [38,39]. PERK also plays a role in producing proper amount of proinsulin in response to hyperglycemia. In PERK deficient islets proinsulin production is markedly increased in response to transient high glucose as compared to control islets [40]. This unregulated production of proinsulin leads to β cell death and subsequent diabetes.

PERK has been shown to protect β cells from ER stress-mediated cell death [40,41]. Under tolerable ER stress conditions, activated PERK phosphorylates eIF2α which leads to attenuation of global mRNA translation reducing ER protein work load and promoting adaption. Meanwhile phosphorylated eIF2α favors the translation of selected mRNAs such as activating transcription factor 4 (ATF4) [42]. ATF4 regulates genes involved in restoring ER homeostasis as well as genes involved in antioxidative stress response and amino acid biosynthesis. We have recently found that PERK upregulates a novel anti-apoptotic effector, apoptosis antagonizing transcription factor (AATF) and mediates β cell survival in part through the transcriptional regulation of AKT1 [43]. Under unresolvable ER stress conditions, continuous eIF2α phosphorylation induces β cell death [11]. In these conditions, ATF4 induces the expression of the pro-apoptotic transcription factor CHOP [15,42,4447]. CHOP regulates the expression of BCL-2 family members as well as other apoptotic components promoting cell death.

ATF6α is the third master regulator of the UPR. Upon ER stress conditions, ATF6α transits to the golgi apparatus and is cleaved into an active transcription factor [48]. This processed form of ATF6α translocates to the nucleus to increase expression of homeostatic effectors involved in protein folding, processing, and degradation [49,50]. However, hyperactivation of ATF6α leads to β cell dysfunction and death. The effectors responsible for this phenotype are currently unknown[51]. In addition, it has been shown that ATF6α hyperactivation suppresses insulin gene expression. Under unresolvable ER stress, ATF6 is activated, leading to a decrease in insulin gene expression [51].

The apoptotic and survival components of the UPR are also regulated at the post-transcriptional level. It has been shown that survival is favored during mild and tolerable ER stress as a consequence of the intrinsic instabilities of mRNAs and proteins that promote apoptosis such as CHOP compared to those that facilitate protein folding and adaptation such as BiP. As a consequence, the expression of apoptotic proteins is short-lived as cells adapt to stress [52]. This observation indicates that post-transcriptional mechanisms regulating the ratio between survival and apoptotic effectors are important for controlling life and death of β cells.

Regulation of the UPR master regulators

As we discussed so far, the UPR regulates both survival and death effectors. The ultimate cell fate consequence regulated by these effectors also depends on the tight regulation of the UPR master regulators. Under tolerable ER stress conditions, the UPR master regulators and downstream components are properly turned off as ER homeostasis is reestablished. However under unresolvable ER stress conditions, the UPR master regulators bypass this feedback regulation, causing harmful hyperacitvation, which favors the activation of apoptotic pathways and the upregulation of death effectors.

Some of the feedback effectors involved in the regulation of the UPR activation include BiP, Gadd34, P58IPK, RACK1 and WFS1. Under normal conditions, BiP binds to the UPR master regulators and prevents their premature activation [5355]. Under ER stress conditions, BiP binds to accumulating unfolded proteins leading to the activation of the UPR mater regulator. Gadd34 dephosphorylates eIF2α by recruiting phosphophatase 1 (PP1) in order to restore protein translation as ER homeostasis is achieved. GADD34 is induced by ER stress under the PERK-eIF2α-ATF4 pathway [56]. P58IPK is another negative regulator of PERK signaling and functions in maintaining ER homeostasis in β cells. P58IPK expression is induced by ER stress mediated by ATF6 and is highly expressed in the pancreas. It has been shown that P58IPK interacts with the kinase domain of PERK and inhibits its activity [57]. P58IPK knockout mice show a gradual onset of glucosuria and hyperglycemia associated with increased apoptosis of islet cells [58]. Recently it has been discovered that IRE1α activation is tightly regulated by the adaptor protein RACK1. Under physiological ER stress conditions such as acute glucose, RACK1 binds to IRE1α and recruits protein phosphatese 2A (PP2A) to turn off IRE1 activation, preventing its hyperactivation [59].

We have recently discovered that WFS1, a causative gene for Wolfram syndrome, is a key negative regulator of the UPR [5]. Wolfram syndrome is a rare autosomal recessive disorder characterized by childhood onset of diabetes mellitus, followed by optic atrophy, deafness and death from neurodegeneration in the third or fourth decades [6062]. Postmortem studies reveal a non-autoimmune-linked selective loss of β cells [63]. Mutations in the WFS1 gene causes Wolfram syndrome [6471]. WFS1 is localized at the ER and has a protective function against ER stress; however, the mechanisms were unclear. Our group found that the WFS1 gene plays an important role in regulating ATF6α activation [5]. In healthy cells, WFS1 prevents activation of ATF6 signaling by recruiting ATF6α to HRD1 and the proteasome for ubiquitin-mediated degradation under non-ER stress conditions. Under ER stress conditions, ATF6α is released from WFS1 and is free to regulate stress signaling targets in the nucleus. As ER homeostasis is reestablished, WFS1 is induced by ER stress, which would cause the eventual degradation of ATF6α. In patients with Wolfram syndrome, ATF6α constantly escapes from WFS1-mediated degradation and is hyperactivated regardless of the ER stress conditions. This ATF6α hyperactivation induces β cell death. Recent genome-wide associate studies have suggested that dysfunctional WFS1 may also have a role in the progression of type 2 diabetes [7274].

Taken together, under tolerable ER stress conditions the UPR is properly turned off favoring β cell survival. However; under unresolvable ER stress conditions, the UPR master regulators are hyperactivated bypassing negative regulation therefore tipping the balance towards β cell apoptosis.

IV. Concluding remarks

Increasing clinical, experimental, and genetic evidence indicates that ER stress and the UPR has a role in β cell dysfunction and death during the progression of type 1, type 2 and genetic forms of diabetes. It is currently believed that the UPR regulates β cell fate by behaving as a binary switch between life and death. The complete understanding of this UPR switch will provide us new insights into the mechanisms of β cell death during diabetes and shed light on future diabetes prevention or treatment.

Acknowledgements

Work in the laboratory of F. Urano is supported by grants from NIH-NIDDK (R01DK067493), the Diabetes and Endocrinology Research Center at the University of Massachusetts Medical School (5 P30 DK32520), and the Juvenile Diabetes Research Foundation International (1-2008-593 and 40-2011-14). We apologize to those colleagues whose publications could not be cited owing to space limitations.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Donath MY, Halban PA. Decreased beta-cell mass in diabetes: significance, mechanisms and therapeutic implications. Diabetologia. 2004;47:581–589. doi: 10.1007/s00125-004-1336-4. [DOI] [PubMed] [Google Scholar]
  • 2. Butler AE, Janson J, Bonner-Weir S, Ritzel R, Rizza RA, Butler PC. Beta-cell deficit and increased beta-cell apoptosis in humans with type 2 diabetes. Diabetes. 2003;52:102–110. doi: 10.2337/diabetes.52.1.102. References 1 and 2 propose that β cell death is a critical pathogenic component in both type 1 and type 2 diabetes.
  • 3. Oslowski CM, Urano F. The binary switch between life and death of endoplasmic reticulum-stressed beta cells. Curr Opin Endocrinol Diabetes Obes. 2010 doi: 10.1097/MED.0b013e3283372843. This is the first article proposing that the UPR acts as a bianary switch regulating life and death of ER stressed β cells.
  • 4.Eizirik DL, Cardozo AK, Cnop M. The role for endoplasmic reticulum stress in diabetes mellitus. Endocr Rev. 2008;29:42–61. doi: 10.1210/er.2007-0015. [DOI] [PubMed] [Google Scholar]
  • 5. Fonseca SG, Ishigaki S, Oslowski CM, Lu S, Lipson KL, Ghosh R, Hayashi E, Ishihara H, Oka Y, Permutt MA, et al. Wolfram syndrome 1 gene negatively regulates ER stress signaling in rodent and human cells. J Clin Invest. 2010 doi: 10.1172/JCI39678. This article reports that β cell death in Wolfram syndrome is caused by the hyperactivation of ER stress signaling.
  • 6.Fonseca SG, Burcin M, Gromada J, Urano F. Endoplasmic reticulum stress in beta-cells and development of diabetes. Curr Opin Pharmacol. 2009 doi: 10.1016/j.coph.2009.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 2007;8:519–529. doi: 10.1038/nrm2199. [DOI] [PubMed] [Google Scholar]
  • 8.Lipson KL, Fonseca SG, Ishigaki S, Nguyen LX, Foss E, Bortell R, Rossini AA, Urano F. Regulation of insulin biosynthesis in pancreatic beta cells by an endoplasmic reticulum-resident protein kinase IRE1. Cell Metab. 2006;4:245–254. doi: 10.1016/j.cmet.2006.07.007. [DOI] [PubMed] [Google Scholar]
  • 9. Lipson KL, Ghosh R, Urano F. The Role of IRE1alpha in the Degradation of Insulin mRNA in Pancreatic beta-Cells. PLoS ONE. 2008;3:e1648. doi: 10.1371/journal.pone.0001648. Reference 8 and 9 report that IRE1α is acivated by hyperglycemia and plays a pivotal role in the regulation of insulin production.
  • 10.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 beta-cell apoptosis. Endocrinology. 2006;147:3398–3407. doi: 10.1210/en.2005-1494. [DOI] [PubMed] [Google Scholar]
  • 11.Cnop M, Ladriere L, Hekerman P, Ortis F, Cardozo AK, Dogusan Z, Flamez D, Boyce M, Yuan J, Eizirik DL. Selective inhibition of eukaryotic translation initiation factor 2 alpha dephosphorylation potentiates fatty acid-induced endoplasmic reticulum stress and causes pancreatic beta-cell dysfunction and apoptosis. J Biol Chem. 2007;282:3989–3997. doi: 10.1074/jbc.M607627200. [DOI] [PubMed] [Google Scholar]
  • 12.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. 2004;145:5087–5096. doi: 10.1210/en.2004-0478. [DOI] [PubMed] [Google Scholar]
  • 13.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 beta-cells. Diabetes. 2005;54:452–461. doi: 10.2337/diabetes.54.2.452. [DOI] [PubMed] [Google Scholar]
  • 14.Eizirik DL, Flodstrom M, Karlsen AE, Welsh N. The harmony of the spheres: inducible nitric oxide synthase and related genes in pancreatic beta cells. Diabetologia. 1996;39:875–890. doi: 10.1007/BF00403906. [DOI] [PubMed] [Google Scholar]
  • 15.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. Proceedings of the National Academy of Sciences of the United States of America. 2001;98:10845–10850. doi: 10.1073/pnas.191207498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.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-kappa B-dependent genes in primary rat pancreatic beta-cells. J Biol Chem. 2001;276:48879–48886. doi: 10.1074/jbc.M108658200. [DOI] [PubMed] [Google Scholar]
  • 17.Wang J, Takeuchi T, Tanaka S, Kubo SK, Kayo T, Lu D, Takata K, Koizumi A, Izumi T. A mutation in the insulin 2 gene induces diabetes with severe pancreatic beta-cell dysfunction in the Mody mouse. Journal of Clinical Investigation. 1999;103:27–37. doi: 10.1172/JCI4431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Yoshioka M, Kayo T, Ikeda T, Koizumi A. A novel locus, Mody4, distal to D7Mit189 on chromosome 7 determines early-onset NIDDM in nonobese C57BL/6 (Akita) mutant mice. Diabetes. 1997;46:887–894. doi: 10.2337/diab.46.5.887. [DOI] [PubMed] [Google Scholar]
  • 19.Kayo T, Koizumi A. Mapping of murine diabetogenic gene mody on chromosome 7 at D7Mit258 and its involvement in pancreatic islet and beta cell development during the perinatal period. Journal of Clinical Investigation. 1998;101:2112–2118. doi: 10.1172/JCI1842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Stoy J, Edghill EL, Flanagan SE, Ye H, Paz VP, Pluzhnikov A, Below JE, Hayes MG, Cox NJ, Lipkind GM, et al. Insulin gene mutations as a cause of permanent neonatal diabetes. Proc Natl Acad Sci U S A. 2007;104:15040–15044. doi: 10.1073/pnas.0707291104. Refeference 28 reports that mutations in the insulin gene cause β cell death and diabetes in humans.
  • 21.Ritzel RA, Meier JJ, Lin CY, Veldhuis JD, Butler PC. Human islet amyloid polypeptide oligomers disrupt cell coupling, induce apoptosis, and impair insulin secretion in isolated human islets. Diabetes. 2007;56:65–71. doi: 10.2337/db06-0734. [DOI] [PubMed] [Google Scholar]
  • 22.Sawaya MR, Sambashivan S, Nelson R, Ivanova MI, Sievers SA, Apostol MI, Thompson MJ, Balbirnie M, Wiltzius JJ, McFarlane HT, et al. Atomic structures of amyloid cross-beta spines reveal varied steric zippers. Nature. 2007;447:453–457. doi: 10.1038/nature05695. [DOI] [PubMed] [Google Scholar]
  • 23.Huang CJ, Haataja L, Gurlo T, Butler AE, Wu X, Soeller WC, Butler PC. Induction of endoplasmic reticulum stress-induced beta-cell apoptosis and accumulation of polyubiquitinated proteins by human islet amyloid polypeptide. Am J Physiol Endocrinol Metab. 2007;293:E1656–E1662. doi: 10.1152/ajpendo.00318.2007. [DOI] [PubMed] [Google Scholar]
  • 24.Matveyenko AV, Gurlo T, Daval M, Butler AE, Butler PC. Successful versus failed adaptation to high-fat diet-induced insulin resistance: the role of IAPP-induced beta-cell endoplasmic reticulum stress. Diabetes. 2009;58:906–916. doi: 10.2337/db08-1464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.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. doi: 10.1016/s0092-8674(01)00611-0. [DOI] [PubMed] [Google Scholar]
  • 26.Shen X, Ellis RE, Lee K, Liu CY, Yang K, Solomon A, Yoshida H, Morimoto R, Kurnit DM, Mori K, et al. Complementary signaling pathways regulate the unfolded protein response and are required for C. elegans development. Cell. 2001;107:893–903. doi: 10.1016/s0092-8674(01)00612-2. [DOI] [PubMed] [Google Scholar]
  • 27.Calfon M, Zeng H, Urano F, Till JH, Hubbard SR, Harding HP, Clark SG, Ron D. IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA. Nature. 2002;415:92–96. doi: 10.1038/415092a. [DOI] [PubMed] [Google Scholar]
  • 28.Yoshida H, Matsui T, Hosokawa N, Kaufman RJ, Nagata K, Mori K. A time-dependent phase shift in the mammalian unfolded protein response. Dev Cell. 2003;4:265–271. doi: 10.1016/s1534-5807(03)00022-4. [DOI] [PubMed] [Google Scholar]
  • 29.Lee AH, Iwakoshi NN, Glimcher LH. XBP-1 regulates a subset of endoplasmic reticulum resident chaperone genes in the unfolded protein response. Mol Cell Biol. 2003;23:7448–7459. doi: 10.1128/MCB.23.21.7448-7459.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.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. 2000;287:664–666. doi: 10.1126/science.287.5453.664. [DOI] [PubMed] [Google Scholar]
  • 31.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. 2002;16:1345–1355. doi: 10.1101/gad.992302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Maundrell K, Antonsson B, Magnenat E, Camps M, Muda M, Chabert C, Gillieron C, Boschert U, Vial-Knecht E, Martinou JC, et al. Bcl-2 undergoes phosphorylation by c-Jun N-terminal kinase/stress-activated protein kinases in the presence of the constitutively active GTP-binding protein Rac1. J Biol Chem. 1997;272:25238–25242. doi: 10.1074/jbc.272.40.25238. [DOI] [PubMed] [Google Scholar]
  • 33.Park J, Kim I, Oh YJ, Lee K, Han PL, Choi EJ. Activation of c-Jun N-terminal kinase antagonizes an anti-apoptotic action of Bcl-2. J Biol Chem. 1997;272:16725–16728. doi: 10.1074/jbc.272.27.16725. [DOI] [PubMed] [Google Scholar]
  • 34.Han D, Lerner AG, Vande Walle L, Upton JP, Xu W, Hagen A, Backes BJ, Oakes SA, Papa FR. IRE1alpha kinase activation modes control alternate endoribonuclease outputs to determine divergent cell fates. Cell. 2009;138:562–575. doi: 10.1016/j.cell.2009.07.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Hollien J, Lin JH, Li H, Stevens N, Walter P, Weissman JS. Regulated Ire1-dependent decay of messenger RNAs in mammalian cells. J Cell Biol. 2009;186:323–331. doi: 10.1083/jcb.200903014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Hollien J, Weissman JS. Decay of endoplasmic reticulum-localized mRNAs during the unfolded protein response. Science. 2006;313:104–107. doi: 10.1126/science.1129631. [DOI] [PubMed] [Google Scholar]
  • 37.Harding HP, Zhang Y, Ron D. Protein translation and folding are coupled by an endoplasmic-reticulum-resident kinase. Nature. 1999;397:271–274. doi: 10.1038/16729. [DOI] [PubMed] [Google Scholar]
  • 38.Zhang P, McGrath B, Li S, Frank A, Zambito F, Reinert J, Gannon M, Ma K, McNaughton K, Cavener DR. The PERK eukaryotic initiation factor 2 alpha kinase is required for the development of the skeletal system, postnatal growth, and the function and viability of the pancreas. Molecular & Cellular Biology. 2002;22:3864–3874. doi: 10.1128/MCB.22.11.3864-3874.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Zhang W, Feng D, Li Y, Iida K, McGrath B, Cavener DR. PERK EIF2AK3 control of pancreatic beta cell differentiation and proliferation is required for postnatal glucose homeostasis. Cell Metab. 2006;4:491–497. doi: 10.1016/j.cmet.2006.11.002. [DOI] [PubMed] [Google Scholar]
  • 40. 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. Molecular Cell. 2001;7:1153–1163. doi: 10.1016/s1097-2765(01)00264-7. References 34 and 35 report that PERK plays a critical role in β cell proliferation, survival, and regulation of insulin production.
  • 41.Harding HP, Zhang Y, Bertolotti A, Zeng H, Ron D. Perk is essential for translational regulation and cell survival during the unfolded protein response. Mol Cell. 2000;5:897–904. doi: 10.1016/s1097-2765(00)80330-5. [DOI] [PubMed] [Google Scholar]
  • 42.Harding HP, Novoa I, Zhang Y, Zeng H, Wek R, Schapira M, Ron D. Regulated translation initiation controls stress-induced gene expression in mammalian cells. Mol Cell. 2000;6:1099–1108. doi: 10.1016/s1097-2765(00)00108-8. [DOI] [PubMed] [Google Scholar]
  • 43.Ishigaki S, Fonseca SG, Oslowski CM, Jurczyk A, Shearstone JR, Zhu LJ, Permutt MA, Greiner DL, Bortell R, Urano F. AATF mediates an antiapoptotic effect of the unfolded protein response through transcriptional regulation of AKT1. Cell Death Differ. 2010;17:774–786. doi: 10.1038/cdd.2009.175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.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. 1992;6:439–453. doi: 10.1101/gad.6.3.439. [DOI] [PubMed] [Google Scholar]
  • 45.Zinszner H, Kuroda M, Wang X, Batchvarova N, Lightfoot RT, Remotti H, Stevens JL, Ron D. CHOP is implicated in programmed cell death in response to impaired function of the endoplasmic reticulum. Genes Dev. 1998;12:982–995. doi: 10.1101/gad.12.7.982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.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. Journal of Clinical Investigation. 2002;109:525–532. doi: 10.1172/JCI14550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Song B, Scheuner D, Ron D, Pennathur S, Kaufman RJ. Chop deletion reduces oxidative stress, improves beta cell function, and promotes cell survival in multiple mouse models of diabetes. J Clin Invest. 2008;118:3378–3389. doi: 10.1172/JCI34587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Ye J, Rawson RB, Komuro R, Chen X, Dave UP, Prywes R, Brown MS, Goldstein JL. ER stress induces cleavage of membrane-bound ATF6 by the same proteases that process SREBPs. Mol Cell. 2000;6:1355–1364. doi: 10.1016/s1097-2765(00)00133-7. [DOI] [PubMed] [Google Scholar]
  • 49.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. 2000;20:6755–6767. doi: 10.1128/mcb.20.18.6755-6767.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Haze K, Yoshida H, Yanagi H, Yura T, Mori K. Mammalian transcription factor ATF6 is synthesized as a transmembrane protein and activated by proteolysis in response to endoplasmic reticulum stress. Mol Biol Cell. 1999;10:3787–3799. doi: 10.1091/mbc.10.11.3787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Seo HY, Kim YD, Lee KM, Min AK, Kim MK, Kim HS, Won KC, Park JY, Lee KU, Choi HS, et al. Endoplasmic reticulum stress-induced activation of activating transcription factor 6 decreases insulin gene expression via up-regulation of orphan nuclear receptor small heterodimer partner. Endocrinology. 2008;149:3832–3841. doi: 10.1210/en.2008-0015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Rutkowski DT, Arnold SM, Miller CN, Wu J, Li J, Gunnison KM, Mori K, Sadighi Akha AA, Raden D, Kaufman RJ. Adaptation to ER stress is mediated by differential stabilities of pro-survival and pro-apoptotic mRNAs and proteins. PLoS Biol. 2006;4:e374. doi: 10.1371/journal.pbio.0040374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Bertolotti A, Zhang Y, Hendershot LM, Harding HP, Ron D. Dynamic interaction of BiP and ER stress transducers in the unfolded-protein response. Nature Cell Biology. 2000;2:326–332. doi: 10.1038/35014014. [DOI] [PubMed] [Google Scholar]
  • 54.Okamura K, Kimata Y, Higashio H, Tsuru A, Kohno K. Dissociation of Kar2p/BiP from an ER sensory molecule, Ire1p, triggers the unfolded protein response in yeast. Biochem Biophys Res Commun. 2000;279:445–450. doi: 10.1006/bbrc.2000.3987. [DOI] [PubMed] [Google Scholar]
  • 55.Shen J, Chen X, Hendershot L, Prywes R. ER stress regulation of ATF6 localization by dissociation of BiP/GRP78 binding and unmasking of Golgi localization signals. Dev Cell. 2002;3:99–111. doi: 10.1016/s1534-5807(02)00203-4. [DOI] [PubMed] [Google Scholar]
  • 56.Novoa I, Zeng H, Harding HP, Ron D. Feedback inhibition of the unfolded protein response by GADD34-mediated dephosphorylation of eIF2alpha. J Cell Biol. 2001;153:1011–1022. doi: 10.1083/jcb.153.5.1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Yan W, Frank CL, Korth MJ, Sopher BL, Novoa I, Ron D, Katze MG. Control of PERK eIF2alpha kinase activity by the endoplasmic reticulum stress-induced molecular chaperone P58IPK. Proc Natl Acad Sci U S A. 2002;99:15920–15925. doi: 10.1073/pnas.252341799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ladiges WC, Knoblaugh SE, Morton JF, Korth MJ, Sopher BL, Baskin CR, MacAuley A, Goodman AG, LeBoeuf RC, Katze MG. Pancreatic beta-cell failure and diabetes in mice with a deletion mutation of the endoplasmic reticulum molecular chaperone gene P58IPK. Diabetes. 2005 doi: 10.2337/diabetes.54.4.1074. [DOI] [PubMed] [Google Scholar]
  • 59.Qiu Y, Mao T, Zhang Y, Shao M, You J, Ding Q, Chen Y, Wu D, Xie D, Lin X, et al. A crucial role for RACK1 in the regulation of glucose-stimulated IRE1alpha activation in pancreatic beta cells. Sci Signal. 2010;3 doi: 10.1126/scisignal.2000514. ra7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Wolfram DJ, Wagener HP. Diabetes mellitus and simple optic atrophy among siblings: report of four cases. May Clin Proc. 1938;1:715–718. [Google Scholar]
  • 61.Barrett TG, Bundey SE. Wolfram (DIDMOAD) syndrome. J Med Genet. 1997;34:838–841. doi: 10.1136/jmg.34.10.838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Barrett TG, Bundey SE, Macleod AF. Neurodegeneration and diabetes: UK nationwide study of Wolfram (DIDMOAD) syndrome. Lancet. 1995;346:1458–1463. doi: 10.1016/s0140-6736(95)92473-6. [DOI] [PubMed] [Google Scholar]
  • 63.Karasik A, O'Hara C, Srikanta S, Swift M, Soeldner JS, Kahn CR, Herskowitz RD. Genetically programmed selective islet beta-cell loss in diabetic subjects with Wolfram's syndrome. Diabetes Care. 1989;12:135–138. doi: 10.2337/diacare.12.2.135. [DOI] [PubMed] [Google Scholar]
  • 64.Inoue H, Tanizawa Y, Wasson J, Behn P, Kalidas K, Bernal-Mizrachi E, Mueckler M, Marshall H, Donis-Keller H, Crock P, et al. A gene encoding a transmembrane protein is mutated in patients with diabetes mellitus and optic atrophy (Wolfram syndrome) Nature Genetics. 1998;20:143–148. doi: 10.1038/2441. [DOI] [PubMed] [Google Scholar]
  • 65. Strom TM, Hortnagel K, Hofmann S, Gekeler F, Scharfe C, Rabl W, Gerbitz KD, Meitinger T. Diabetes insipidus, diabetes mellitus, optic atrophy and deafness (DIDMOAD) caused by mutations in a novel gene (wolframin) coding for a predicted transmembrane protein. Hum Mol Genet. 1998;7:2021–2028. doi: 10.1093/hmg/7.13.2021. References 64 and 65 report that WFS1 is a causative gene for Wolfram syndrome.
  • 66.Khanim F, Kirk J, Latif F, Barrett TG. WFS1/wolframin mutations, Wolfram syndrome, and associated diseases. Hum Mutat. 2001;17:357–367. doi: 10.1002/humu.1110. [DOI] [PubMed] [Google Scholar]
  • 67.Takeda K, Inoue H, Tanizawa Y, Matsuzaki Y, Oba J, Watanabe Y, Shinoda K, Oka Y. 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. doi: 10.1093/hmg/10.5.477. [DOI] [PubMed] [Google Scholar]
  • 68.Hofmann S, Philbrook C, Gerbitz KD, Bauer MF. Wolfram syndrome: structural and functional analyses of mutant and wild-type wolframin, the WFS1 gene product. Hum Mol Genet. 2003;12:2003–2012. doi: 10.1093/hmg/ddg214. [DOI] [PubMed] [Google Scholar]
  • 69.Fonseca SG, Fukuma M, Lipson KL, Nguyen LX, Allen JR, Oka Y, Urano F. 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. doi: 10.1074/jbc.M507426200. [DOI] [PubMed] [Google Scholar]
  • 70.Ishihara H, Takeda S, Tamura A, Takahashi R, Yamaguchi S, Takei D, Yamada T, Inoue H, Soga H, Katagiri H, 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. doi: 10.1093/hmg/ddh125. [DOI] [PubMed] [Google Scholar]
  • 71.Riggs AC, Bernal-Mizrachi E, Ohsugi M, Wasson J, Fatrai S, Welling C, Murray J, Schmidt RE, Herrera PL, Permutt MA. 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. doi: 10.1007/s00125-005-1947-4. [DOI] [PubMed] [Google Scholar]
  • 72.Minton JA, Hattersley AT, Owen K, McCarthy MI, Walker M, Latif F, Barrett T, Frayling TM. Association studies of genetic variation in the WFS1 gene and type 2 diabetes in U.K. populations. Diabetes. 2002;51:1287–1290. doi: 10.2337/diabetes.51.4.1287. [DOI] [PubMed] [Google Scholar]
  • 73.Sandhu MS, Weedon MN, Fawcett KA, Wasson J, Debenham SL, Daly A, Lango H, Frayling TM, Neumann RJ, Sherva R, et al. Common variants in WFS1 confer risk of type 2 diabetes. Nat Genet. 2007;39:951–953. doi: 10.1038/ng2067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Franks PW, Rolandsson O, Debenham SL, Fawcett KA, Payne F, Dina C, Froguel P, Mohlke KL, Willer C, Olsson T, et al. Replication of the association between variants in WFS1 and risk of type 2 diabetes in European populations. Diabetologia. 2008;51:458–463. doi: 10.1007/s00125-007-0887-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Florez JC, Jablonski KA, McAteer J, Sandhu MS, Wareham NJ, Barroso I, Franks PW, Altshuler D, Knowler WC. Testing of diabetes-associated WFS1 polymorphisms in the Diabetes Prevention Program. Diabetologia. 2008;51:451–457. doi: 10.1007/s00125-007-0891-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Sparso T, Andersen G, Albrechtsen A, Jorgensen T, Borch-Johnsen K, Sandbaek A, Lauritzen T, Wasson J, Permutt MA, Glaser B, et al. Impact of polymorphisms in WFS1 on prediabetic phenotypes in a population-based sample of middle-aged people with normal and abnormal glucose regulation. Diabetologia. 2008;51:1646–1652. doi: 10.1007/s00125-008-1064-2. [DOI] [PubMed] [Google Scholar]
  • 77.Schafer SA, Mussig K, Staiger H, Machicao F, Stefan N, Gallwitz B, Haring HU, Fritsche A. A common genetic variant in WFS1 determines impaired glucagon-like peptide-1-induced insulin secretion. Diabetologia. 2009;52:1075–1082. doi: 10.1007/s00125-009-1344-5. [DOI] [PubMed] [Google Scholar]

RESOURCES