Skip to main content
Top
Published in: Diabetologia 5/2007

01-05-2007 | Article

Global profiling of genes modified by endoplasmic reticulum stress in pancreatic beta cells reveals the early degradation of insulin mRNAs

Authors: P. Pirot, N. Naamane, F. Libert, N. E. Magnusson, T. F. Ørntoft, A. K. Cardozo, D. L. Eizirik

Published in: Diabetologia | Issue 5/2007

Login to get access

Abstract

Aims/hypothesis

Pancreatic beta cells respond to endoplasmic reticulum (ER) stress by activating the unfolded protein response. If the stress is prolonged, or the adaptive response fails, apoptosis is triggered. We used a ‘homemade’ microarray specifically designed for the study of beta cell apoptosis (the APOCHIP) to uncover mechanisms regulating beta cell responses to ER stress.

Materials and methods

A time course viability and microarray analysis was performed in insulin-producing INS-1E cells exposed to the reversible ER stress inducer cyclopiazonic acid (CPA). Modification of selected genes was confirmed by real-time RT-PCR, and the observed inhibition of expression of the insulin-1 (Ins1) and insulin-2 (Ins2) genes was further characterised in primary beta cells exposed to a diverse range of agents that induce ER stress.

Results

CPA-induced ER stress modified the expression of 183 genes at one or more of the time points studied. The expression of most of these genes returned to control levels after a 3 h recovery period following CPA removal, with all cells surviving. Two groups of genes were particularly affected by CPA, namely, those related to cellular responses to ER stress, which were mostly upregulated, and those related to differentiated beta cell functions, which were downregulated. Levels of Ins1 and Ins2 mRNAs were severely decreased in response to CPA treatment as a result of degradation, and there was a concomitant increase in the level of IRE1 activation.

Conclusions/interpretation

In this study we provide the first global analysis of beta cell molecular responses to a severe ER stress, and identify the early degradation of mRNA transcripts of the insulin genes as an important component of this response.
Appendix
Available only for authorised users
Literature
1.
go back to reference Schuit FC, In’t Veld PA, Pipeleers DG (1988) Glucose stimulates proinsulin biosynthesis by a dose-dependent recruitment of pancreatic beta cells. Proc Natl Acad Sci USA 85:3865–3869PubMedCrossRef Schuit FC, In’t Veld PA, Pipeleers DG (1988) Glucose stimulates proinsulin biosynthesis by a dose-dependent recruitment of pancreatic beta cells. Proc Natl Acad Sci USA 85:3865–3869PubMedCrossRef
2.
go back to reference Schroder M, Kaufman RJ (2005) The mammalian unfolded protein response. Annu Rev Biochem 74:739–789PubMedCrossRef Schroder M, Kaufman RJ (2005) The mammalian unfolded protein response. Annu Rev Biochem 74:739–789PubMedCrossRef
3.
go back to reference Harding HP, Ron D (2002) Endoplasmic reticulum stress and the development of diabetes: a review. Diabetes 51(Suppl 3):S455–S461PubMedCrossRef Harding HP, Ron D (2002) Endoplasmic reticulum stress and the development of diabetes: a review. Diabetes 51(Suppl 3):S455–S461PubMedCrossRef
4.
go back to reference Schroder M, Kaufman RJ (2006) Divergent roles of IRE1alpha and PERK in the unfolded protein response. Curr Mol Med 6:5–36PubMedCrossRef Schroder M, Kaufman RJ (2006) Divergent roles of IRE1alpha and PERK in the unfolded protein response. Curr Mol Med 6:5–36PubMedCrossRef
5.
go back to reference Hollien J, Weissman JS (2006) Decay of endoplasmic reticulum-localized mRNAs during the unfolded protein response. Science 313:104–107PubMedCrossRef Hollien J, Weissman JS (2006) Decay of endoplasmic reticulum-localized mRNAs during the unfolded protein response. Science 313:104–107PubMedCrossRef
6.
go back to reference Xu C, Bailly-Maitre B, Reed JC (2005) Endoplasmic reticulum stress: cell life and death decisions. J Clin Invest 115:2656–2664PubMedCrossRef Xu C, Bailly-Maitre B, Reed JC (2005) Endoplasmic reticulum stress: cell life and death decisions. J Clin Invest 115:2656–2664PubMedCrossRef
7.
go back to reference Hartman MG, Lu D, Kim ML et al (2004) Role for activating transcription factor 3 in stress-induced beta-cell apoptosis. Mol Cell Biol 24:5721–5732PubMedCrossRef Hartman MG, Lu D, Kim ML et al (2004) Role for activating transcription factor 3 in stress-induced beta-cell apoptosis. Mol Cell Biol 24:5721–5732PubMedCrossRef
8.
go back to reference Oyadomari S, Takeda K, Takiguchi M et al (2001) Nitric oxide-induced apoptosis in pancreatic beta cells is mediated by the endoplasmic reticulum stress pathway. Proc Natl Acad Sci USA 98:10845–10850PubMedCrossRef Oyadomari S, Takeda K, Takiguchi M et al (2001) Nitric oxide-induced apoptosis in pancreatic beta cells is mediated by the endoplasmic reticulum stress pathway. Proc Natl Acad Sci USA 98:10845–10850PubMedCrossRef
9.
go back to reference Oyadomari S, Koizumi A, Takeda K et al (2002) Targeted disruption of the Chop gene delays endoplasmic reticulum stress-mediated diabetes. J Clin Invest 109:525–532PubMedCrossRef Oyadomari S, Koizumi A, Takeda K et al (2002) Targeted disruption of the Chop gene delays endoplasmic reticulum stress-mediated diabetes. J Clin Invest 109:525–532PubMedCrossRef
10.
go back to reference Delepine M, Nicolino M, Barrett T, Golamaully M, Lathrop GM, Julier C (2000) EIF2AK3, encoding translation initiation factor 2-alpha kinase 3, is mutated in patients with Wolcott–Rallison syndrome. Nat Genet 25:406–409PubMedCrossRef Delepine M, Nicolino M, Barrett T, Golamaully M, Lathrop GM, Julier C (2000) EIF2AK3, encoding translation initiation factor 2-alpha kinase 3, is mutated in patients with Wolcott–Rallison syndrome. Nat Genet 25:406–409PubMedCrossRef
11.
go back to reference Harding HP, Zeng H, Zhang Y et al (2001) Diabetes mellitus and exocrine pancreatic dysfunction in perk−/−mice reveals a role for translational control in secretory cell survival. Mol Cell 7:1153–1163PubMedCrossRef Harding HP, Zeng H, Zhang Y et al (2001) Diabetes mellitus and exocrine pancreatic dysfunction in perk−/−mice reveals a role for translational control in secretory cell survival. Mol Cell 7:1153–1163PubMedCrossRef
12.
go back to reference Scheuner D, Mierde DV, Song B et al (2005) Control of mRNA translation preserves endoplasmic reticulum function in beta cells and maintains glucose homeostasis. Nat Med 11:757–764PubMedCrossRef Scheuner D, Mierde DV, Song B et al (2005) Control of mRNA translation preserves endoplasmic reticulum function in beta cells and maintains glucose homeostasis. Nat Med 11:757–764PubMedCrossRef
13.
go back to reference Cnop M, Ladriere L, Hekerman P et al (2007) Selective inhibition of eIF2α dephosphorylation potentiates fatty acid-induced ER stress and causes pancreatic β-cell dysfunction and apoptosis. J Biol Chem 282:3989–3997PubMedCrossRef Cnop M, Ladriere L, Hekerman P et al (2007) Selective inhibition of eIF2α dephosphorylation potentiates fatty acid-induced ER stress and causes pancreatic β-cell dysfunction and apoptosis. J Biol Chem 282:3989–3997PubMedCrossRef
14.
go back to reference Kharroubi I, Ladriere L, Cardozo AK, Dogusan Z, Cnop M, Eizirik DL (2004) Free fatty acids and cytokines induce pancreatic beta-cell apoptosis by different mechanisms: role of nuclear factor-kappaB and endoplasmic reticulum stress. Endocrinology 145:5087–5096PubMedCrossRef Kharroubi I, Ladriere L, Cardozo AK, Dogusan Z, Cnop M, Eizirik DL (2004) Free fatty acids and cytokines induce pancreatic beta-cell apoptosis by different mechanisms: role of nuclear factor-kappaB and endoplasmic reticulum stress. Endocrinology 145:5087–5096PubMedCrossRef
15.
go back to reference Cardozo AK, Ortis F, Storling J et al (2005) Cytokines downregulate the sarcoendoplasmic reticulum pump Ca2+ ATPase 2b and deplete endoplasmic reticulum Ca2+, leading to induction of endoplasmic reticulum stress in pancreatic β-cells. Diabetes 54:452–461PubMedCrossRef Cardozo AK, Ortis F, Storling J et al (2005) Cytokines downregulate the sarcoendoplasmic reticulum pump Ca2+ ATPase 2b and deplete endoplasmic reticulum Ca2+, leading to induction of endoplasmic reticulum stress in pancreatic β-cells. Diabetes 54:452–461PubMedCrossRef
16.
go back to reference Cnop M, Welsh N, Jonas JC et al (2005) Mechanisms of pancreatic β-cell death in type 1 and type 2 diabetes: many differences, few similarities. Diabetes 54(Suppl 2):S97–S107PubMedCrossRef Cnop M, Welsh N, Jonas JC et al (2005) Mechanisms of pancreatic β-cell death in type 1 and type 2 diabetes: many differences, few similarities. Diabetes 54(Suppl 2):S97–S107PubMedCrossRef
17.
go back to reference Karaskov E, Scott C, Zhang L, Teodoro T, Ravazzola M, Volchuk A (2006) Chronic palmitate but not oleate exposure induces endoplasmic reticulum stress, which may contribute to INS-1 pancreatic β-cell apoptosis. Endocrinology 147:3398–3407PubMedCrossRef Karaskov E, Scott C, Zhang L, Teodoro T, Ravazzola M, Volchuk A (2006) Chronic palmitate but not oleate exposure induces endoplasmic reticulum stress, which may contribute to INS-1 pancreatic β-cell apoptosis. Endocrinology 147:3398–3407PubMedCrossRef
18.
go back to reference Pirot P, Eizirik DL, Cardozo AK (2006) Interferon-gamma potentiates endoplasmic reticulum stress-induced death by reducing pancreatic beta cell defence mechanisms. Diabetologia 49:1229–1236PubMedCrossRef Pirot P, Eizirik DL, Cardozo AK (2006) Interferon-gamma potentiates endoplasmic reticulum stress-induced death by reducing pancreatic beta cell defence mechanisms. Diabetologia 49:1229–1236PubMedCrossRef
19.
go back to reference Zhou YP, Teng D, Dralyuk F et al (1998) Apoptosis in insulin-secreting cells. Evidence for the role of intracellular Ca2+ stores and arachidonic acid metabolism. J Clin Invest 101:1623–1632PubMed Zhou YP, Teng D, Dralyuk F et al (1998) Apoptosis in insulin-secreting cells. Evidence for the role of intracellular Ca2+ stores and arachidonic acid metabolism. J Clin Invest 101:1623–1632PubMed
20.
go back to reference Magnusson NE, Cardozo AK, Kruhoffer M, Eizirik DL, Orntoft TF, Jensen JL (2005) Construction and validation of the APOCHIP, a spotted oligo-microarray for the study of beta-cell apoptosis. BMC Bioinformatics 6:311PubMedCrossRef Magnusson NE, Cardozo AK, Kruhoffer M, Eizirik DL, Orntoft TF, Jensen JL (2005) Construction and validation of the APOCHIP, a spotted oligo-microarray for the study of beta-cell apoptosis. BMC Bioinformatics 6:311PubMedCrossRef
21.
go back to reference Rasschaert J, Ladriere L, Urbain M et al (2005) Toll-like receptor 3 and STAT-1 contribute to double-stranded RNA+ interferon-gamma-induced apoptosis in primary pancreatic beta-cells. J Biol Chem 280:33984–33991PubMedCrossRef Rasschaert J, Ladriere L, Urbain M et al (2005) Toll-like receptor 3 and STAT-1 contribute to double-stranded RNA+ interferon-gamma-induced apoptosis in primary pancreatic beta-cells. J Biol Chem 280:33984–33991PubMedCrossRef
22.
go back to reference Kutlu B, Cardozo AK, Darville MI et al (2003) Discovery of gene networks regulating cytokine-induced dysfunction and apoptosis in insulin-producing INS-1 cells. Diabetes 52:2701–2719PubMedCrossRef Kutlu B, Cardozo AK, Darville MI et al (2003) Discovery of gene networks regulating cytokine-induced dysfunction and apoptosis in insulin-producing INS-1 cells. Diabetes 52:2701–2719PubMedCrossRef
23.
go back to reference Wilson DL, Buckley MJ, Helliwell CA, Wilson IW (2003) New normalization methods for cDNA microarray data. Bioinformatics 19:1325–1332PubMedCrossRef Wilson DL, Buckley MJ, Helliwell CA, Wilson IW (2003) New normalization methods for cDNA microarray data. Bioinformatics 19:1325–1332PubMedCrossRef
24.
go back to reference Storey JD, Tibshirani R (2003) Statistical significance for genome-wide studies. Proc Natl Acad Sci USA 100:9440–9445PubMedCrossRef Storey JD, Tibshirani R (2003) Statistical significance for genome-wide studies. Proc Natl Acad Sci USA 100:9440–9445PubMedCrossRef
25.
go back to reference Kutlu B, Naamane N, Berthou L, Eizirik DL (2004) New approaches for in silico identification of cytokine-modified beta cell gene networks. Ann NY Acad Sci 1037:41–58PubMedCrossRef Kutlu B, Naamane N, Berthou L, Eizirik DL (2004) New approaches for in silico identification of cytokine-modified beta cell gene networks. Ann NY Acad Sci 1037:41–58PubMedCrossRef
26.
go back to reference Cardozo AK, Kruhoffer M, Leeman R, Orntoft T, Eizirik DL (2001) Identification of novel cytokine-induced genes in pancreatic beta-cells by high-density oligonucleotide arrays. Diabetes 50:909–920PubMedCrossRef Cardozo AK, Kruhoffer M, Leeman R, Orntoft T, Eizirik DL (2001) Identification of novel cytokine-induced genes in pancreatic beta-cells by high-density oligonucleotide arrays. Diabetes 50:909–920PubMedCrossRef
27.
go back to reference Green LC, Wagner DA, Glogowski J, Skipper PL, Wishnok JS, Tannenbaum SR (1982) Analysis of nitrate, nitrite, and [15N]nitrate in biological fluids. Anal Biochem 126:131–138PubMedCrossRef Green LC, Wagner DA, Glogowski J, Skipper PL, Wishnok JS, Tannenbaum SR (1982) Analysis of nitrate, nitrite, and [15N]nitrate in biological fluids. Anal Biochem 126:131–138PubMedCrossRef
28.
go back to reference Chepurny OG, Hussain MA, Holz GG (2002) Exendin-4 as a stimulator of rat insulin I gene promoter activity via bZIP/CRE interactions sensitive to serine/threonine protein kinase inhibitor Ro 31-8220. Endocrinology 143:2303–2313PubMedCrossRef Chepurny OG, Hussain MA, Holz GG (2002) Exendin-4 as a stimulator of rat insulin I gene promoter activity via bZIP/CRE interactions sensitive to serine/threonine protein kinase inhibitor Ro 31-8220. Endocrinology 143:2303–2313PubMedCrossRef
29.
go back to reference Dooley TP, Curto EV, Davis RL, Grammatico P, Robinson ES, Wilborn TW (2003) DNA microarrays and likelihood ratio bioinformatic methods: discovery of human melanocyte biomarkers. Pigment Cell Res 16:245–253PubMedCrossRef Dooley TP, Curto EV, Davis RL, Grammatico P, Robinson ES, Wilborn TW (2003) DNA microarrays and likelihood ratio bioinformatic methods: discovery of human melanocyte biomarkers. Pigment Cell Res 16:245–253PubMedCrossRef
30.
go back to reference Brewer JW, Hendershot LM, Sherr CJ, Diehl JA (1999) Mammalian unfolded protein response inhibits cyclin D1 translation and cell-cycle progression. Proc Natl Acad Sci USA 96:8505–8510PubMedCrossRef Brewer JW, Hendershot LM, Sherr CJ, Diehl JA (1999) Mammalian unfolded protein response inhibits cyclin D1 translation and cell-cycle progression. Proc Natl Acad Sci USA 96:8505–8510PubMedCrossRef
31.
go back to reference Hendershot LM (2004) The ER function BiP is a master regulator of ER function. Mt Sinai J Med 71:289–297PubMed Hendershot LM (2004) The ER function BiP is a master regulator of ER function. Mt Sinai J Med 71:289–297PubMed
32.
go back to reference Gelebart P, Opas M, Michalak M (2005) Calreticulin, a Ca2+-binding chaperone of the endoplasmic reticulum. Int J Biochem Cell Biol 37:260–266PubMedCrossRef Gelebart P, Opas M, Michalak M (2005) Calreticulin, a Ca2+-binding chaperone of the endoplasmic reticulum. Int J Biochem Cell Biol 37:260–266PubMedCrossRef
33.
go back to reference Argon Y, Simen BB (1999) GRP94, an ER chaperone with protein and peptide binding properties. Semin Cell Dev Biol 10:495–505PubMedCrossRef Argon Y, Simen BB (1999) GRP94, an ER chaperone with protein and peptide binding properties. Semin Cell Dev Biol 10:495–505PubMedCrossRef
34.
go back to reference Romisch K (1999) Surfing the Sec61 channel: bidirectional protein translocation across the ER membrane. J Cell Sci 112:4185–4191PubMed Romisch K (1999) Surfing the Sec61 channel: bidirectional protein translocation across the ER membrane. J Cell Sci 112:4185–4191PubMed
35.
go back to reference Harding HP, Zhang Y, Zeng H et al (2003) An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol Cell 11:619–633PubMedCrossRef Harding HP, Zhang Y, Zeng H et al (2003) An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol Cell 11:619–633PubMedCrossRef
36.
go back to reference Ma Y, Brewer JW, Diehl JA, Hendershot LM (2002) Two distinct stress signaling pathways converge upon the CHOP promoter during the mammalian unfolded protein response. J Mol Biol 318:1351–1365PubMedCrossRef Ma Y, Brewer JW, Diehl JA, Hendershot LM (2002) Two distinct stress signaling pathways converge upon the CHOP promoter during the mammalian unfolded protein response. J Mol Biol 318:1351–1365PubMedCrossRef
37.
go back to reference Pirot P, Ortis F, Cnop M et al (2007) Transcriptional regulation of the endoplasmic reticulum (ER) stress gene Chop in pancreatic insulin producing cells. Diabetes DOI: 10.2337/db06–1253 Pirot P, Ortis F, Cnop M et al (2007) Transcriptional regulation of the endoplasmic reticulum (ER) stress gene Chop in pancreatic insulin producing cells. Diabetes DOI: 10.​2337/​db06–1253
38.
go back to reference McCullough KD, Martindale JL, Klotz LO, Aw TY, Holbrook NJ (2001) Gadd153 sensitizes cells to endoplasmic reticulum stress by down- regulating Bcl2 and perturbing the cellular redox state. Mol Cell Biol 21:1249–1259PubMedCrossRef McCullough KD, Martindale JL, Klotz LO, Aw TY, Holbrook NJ (2001) Gadd153 sensitizes cells to endoplasmic reticulum stress by down- regulating Bcl2 and perturbing the cellular redox state. Mol Cell Biol 21:1249–1259PubMedCrossRef
39.
go back to reference Nakagawa T, Zhu H, Morishima N et al (2000) Caspase-12 mediates endoplasmic-reticulum-specific apoptosis and cytotoxicity by amyloid-beta. Nature 403:98–103PubMedCrossRef Nakagawa T, Zhu H, Morishima N et al (2000) Caspase-12 mediates endoplasmic-reticulum-specific apoptosis and cytotoxicity by amyloid-beta. Nature 403:98–103PubMedCrossRef
40.
go back to reference Steiner DF, Rouille Y, Gong Q, Martin S, Carroll R, Chan SJ (1996) The role of prohormone convertases in insulin biosynthesis: evidence for inherited defects in their action in man and experimental animals. Diabetes Metab 22:94–104PubMed Steiner DF, Rouille Y, Gong Q, Martin S, Carroll R, Chan SJ (1996) The role of prohormone convertases in insulin biosynthesis: evidence for inherited defects in their action in man and experimental animals. Diabetes Metab 22:94–104PubMed
41.
go back to reference Lang J (1999) Molecular mechanisms and regulation of insulin exocytosis as a paradigm of endocrine secretion. Eur J Biochem 259:3–17PubMedCrossRef Lang J (1999) Molecular mechanisms and regulation of insulin exocytosis as a paradigm of endocrine secretion. Eur J Biochem 259:3–17PubMedCrossRef
42.
go back to reference Chakrabarti SK, Mirmira RG (2003) Transcription factors direct the development and function of pancreatic beta cells. Trends Endocrinol Metab 14:78–84PubMedCrossRef Chakrabarti SK, Mirmira RG (2003) Transcription factors direct the development and function of pancreatic beta cells. Trends Endocrinol Metab 14:78–84PubMedCrossRef
43.
go back to reference Hansen BS, Nielsen JH, Linde S (1988) Effect of interleukin-1 on the biosynthesis of proinsulin and insulin in isolated rat pancreatic islets. Biomed Biochim Acta 47:305–309PubMed Hansen BS, Nielsen JH, Linde S (1988) Effect of interleukin-1 on the biosynthesis of proinsulin and insulin in isolated rat pancreatic islets. Biomed Biochim Acta 47:305–309PubMed
44.
go back to reference Hostens K, Pavlovic D, Zambre Y et al (1999) Exposure of human islets to cytokines can result in disproportionately elevated proinsulin release. J Clin Invest 104:67–72PubMedCrossRef Hostens K, Pavlovic D, Zambre Y et al (1999) Exposure of human islets to cytokines can result in disproportionately elevated proinsulin release. J Clin Invest 104:67–72PubMedCrossRef
45.
go back to reference Ohara-Imaizumi M, Cardozo AK, Kikuta T, Eizirik DL, Nagamatsu S (2004) The cytokine interleukin-1beta reduces the docking and fusion of insulin granules in pancreatic beta-cells, preferentially decreasing the first phase of exocytosis. J Biol Chem 279:41271–41274PubMedCrossRef Ohara-Imaizumi M, Cardozo AK, Kikuta T, Eizirik DL, Nagamatsu S (2004) The cytokine interleukin-1beta reduces the docking and fusion of insulin granules in pancreatic beta-cells, preferentially decreasing the first phase of exocytosis. J Biol Chem 279:41271–41274PubMedCrossRef
46.
go back to reference Spinas GA, Hansen BS, Linde S et al (1987) Interleukin 1 dose-dependently affects the biosynthesis of (pro)insulin in isolated rat islets of Langerhans. Diabetologia 30:474–480PubMedCrossRef Spinas GA, Hansen BS, Linde S et al (1987) Interleukin 1 dose-dependently affects the biosynthesis of (pro)insulin in isolated rat islets of Langerhans. Diabetologia 30:474–480PubMedCrossRef
47.
go back to reference Eizirik DL, Welsh M, Strandell E, Welsh N, Sandler S (1990) Interleukin-1 beta depletes insulin messenger ribonucleic acid and increases the heat shock protein hsp70 in mouse pancreatic islets without impairing the glucose metabolism. Endocrinology 127:2290–2297PubMedCrossRef Eizirik DL, Welsh M, Strandell E, Welsh N, Sandler S (1990) Interleukin-1 beta depletes insulin messenger ribonucleic acid and increases the heat shock protein hsp70 in mouse pancreatic islets without impairing the glucose metabolism. Endocrinology 127:2290–2297PubMedCrossRef
48.
go back to reference Eizirik DL (1991) Interleukin-1 beta induces an early decrease in insulin release, (pro)insulin biosynthesis and insulin mRNA in mouse pancreatic islets by a mechanism dependent on gene transcription and protein synthesis. Autoimmunity 10:107–113PubMed Eizirik DL (1991) Interleukin-1 beta induces an early decrease in insulin release, (pro)insulin biosynthesis and insulin mRNA in mouse pancreatic islets by a mechanism dependent on gene transcription and protein synthesis. Autoimmunity 10:107–113PubMed
49.
go back to reference Lipson KL, Fonseca SG, Ishigaki S et al (2006) Regulation of insulin biosynthesis in pancreatic beta cells by an endoplasmic reticulum-resident protein kinase IRE1. Cell Metab 4:245–254PubMedCrossRef Lipson KL, Fonseca SG, Ishigaki S et al (2006) Regulation of insulin biosynthesis in pancreatic beta cells by an endoplasmic reticulum-resident protein kinase IRE1. Cell Metab 4:245–254PubMedCrossRef
50.
Metadata
Title
Global profiling of genes modified by endoplasmic reticulum stress in pancreatic beta cells reveals the early degradation of insulin mRNAs
Authors
P. Pirot
N. Naamane
F. Libert
N. E. Magnusson
T. F. Ørntoft
A. K. Cardozo
D. L. Eizirik
Publication date
01-05-2007
Publisher
Springer-Verlag
Published in
Diabetologia / Issue 5/2007
Print ISSN: 0012-186X
Electronic ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-007-0609-0

Other articles of this Issue 5/2007

Diabetologia 5/2007 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.