Skip to main content
Erschienen in: Diabetologia 5/2016

24.02.2016 | Article

Methylation of insulin DNA in response to proinflammatory cytokines during the progression of autoimmune diabetes in NOD mice

verfasst von: Jinxiu Rui, Songyan Deng, Jasmin Lebastchi, Pamela L. Clark, Sahar Usmani-Brown, Kevan C. Herold

Erschienen in: Diabetologia | Ausgabe 5/2016

Einloggen, um Zugang zu erhalten

Abstract

Aims/hypothesis

Type 1 diabetes is caused by the immunological destruction of pancreatic beta cells. Preclinical and clinical data indicate that there are changes in beta cell function at different stages of the disease, but the fate of beta cells has not been closely studied. We studied how immune factors affect the function and epigenetics of beta cells during disease progression and identified possible triggers of these changes.

Methods

We studied FACS sorted beta cells and infiltrating lymphocytes from NOD mouse and human islets. Gene expression was measured by quantitative real-time RT-PCR (qRT-PCR) and methylation of the insulin genes was investigated by high-throughput and Sanger sequencing. To understand the role of DNA methyltransferases, Dnmt3a was knocked down with small interfering RNA (siRNA). The effects of cytokines on methylation and expression of the insulin gene were studied in humans and mice.

Results

During disease progression in NOD mice, there was an inverse relationship between the proportion of infiltrating lymphocytes and the beta cell mass. In beta cells, methylation marks in the Ins1 and Ins2 genes changed over time. Insulin gene expression appears to be most closely regulated by the methylation of Ins1 exon 2 and Ins2 exon 1. Cytokine transcription increased with age in NOD mice, and these cytokines could induce methylation marks in the insulin DNA by inducing methyltransferases. Similar changes were induced by cytokines in human beta cells in vitro.

Conclusions/interpretation

Epigenetic modification of DNA by methylation in response to immunological stressors may be a mechanism that affects insulin gene expression during the progression of type 1 diabetes.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
1.
Zurück zum Zitat Herold KC, Vignali DA, Cooke A, Bluestone JA (2013) Type 1 diabetes: translating mechanistic observations into effective clinical outcomes. Nat Rev Immunol 13:243–256CrossRefPubMedPubMedCentral Herold KC, Vignali DA, Cooke A, Bluestone JA (2013) Type 1 diabetes: translating mechanistic observations into effective clinical outcomes. Nat Rev Immunol 13:243–256CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Mathis D, Vence L, Benoist C (2001) Beta-cell death during progression to diabetes. Nature 414:792–798CrossRefPubMed Mathis D, Vence L, Benoist C (2001) Beta-cell death during progression to diabetes. Nature 414:792–798CrossRefPubMed
4.
Zurück zum Zitat Sherry NA, Chen W, Kushner JA et al (2007) Exendin-4 improves reversal of diabetes in NOD mice treated with anti-CD3 monoclonal antibody by enhancing recovery of beta-cells. Endocrinology 148:5136–5144CrossRefPubMed Sherry NA, Chen W, Kushner JA et al (2007) Exendin-4 improves reversal of diabetes in NOD mice treated with anti-CD3 monoclonal antibody by enhancing recovery of beta-cells. Endocrinology 148:5136–5144CrossRefPubMed
6.
8.
Zurück zum Zitat Takizawa F, Mizutani S, Ogawa Y, Sawada N (2013) Glucose-independent persistence of PAI-1 gene expression and H3K4 tri-methylation in type 1 diabetic mouse endothelium: implication in metabolic memory. Biochem Biophys Res Commun 433:66–72CrossRefPubMed Takizawa F, Mizutani S, Ogawa Y, Sawada N (2013) Glucose-independent persistence of PAI-1 gene expression and H3K4 tri-methylation in type 1 diabetic mouse endothelium: implication in metabolic memory. Biochem Biophys Res Commun 433:66–72CrossRefPubMed
9.
Zurück zum Zitat Ishikawa K, Tsunekawa S, Ikeniwa M et al (2015) Long-term pancreatic beta cell exposure to high levels of glucose but not palmitate induces DNA methylation within the insulin gene promoter and represses transcriptional activity. PLoS One 10:e0115350CrossRefPubMedPubMedCentral Ishikawa K, Tsunekawa S, Ikeniwa M et al (2015) Long-term pancreatic beta cell exposure to high levels of glucose but not palmitate induces DNA methylation within the insulin gene promoter and represses transcriptional activity. PLoS One 10:e0115350CrossRefPubMedPubMedCentral
10.
11.
Zurück zum Zitat El-Osta A, Brasacchio D, Yao D et al (2008) Transient high glucose causes persistent epigenetic changes and altered gene expression during subsequent normoglycemia. J Exp Med 205:2409–2417CrossRefPubMedPubMedCentral El-Osta A, Brasacchio D, Yao D et al (2008) Transient high glucose causes persistent epigenetic changes and altered gene expression during subsequent normoglycemia. J Exp Med 205:2409–2417CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Mitic T, Emanueli C (2012) Diabetes-induced epigenetic signature in vascular cells. Endocr Metab Immune Disord Drug Targets 12:107–117CrossRefPubMed Mitic T, Emanueli C (2012) Diabetes-induced epigenetic signature in vascular cells. Endocr Metab Immune Disord Drug Targets 12:107–117CrossRefPubMed
13.
Zurück zum Zitat Feng B, Ruiz MA, Chakrabarti S (2013) Oxidative-stress-induced epigenetic changes in chronic diabetic complications. Can J Physiol Pharmacol 91:213–220CrossRefPubMed Feng B, Ruiz MA, Chakrabarti S (2013) Oxidative-stress-induced epigenetic changes in chronic diabetic complications. Can J Physiol Pharmacol 91:213–220CrossRefPubMed
14.
Zurück zum Zitat Jaenisch R, Bird A (2003) Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet 33(Suppl):245–254CrossRefPubMed Jaenisch R, Bird A (2003) Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet 33(Suppl):245–254CrossRefPubMed
15.
Zurück zum Zitat Alvarado S, Fernald RD, Storey KB, Szyf M (2014) The dynamic nature of DNA methylation: a role in response to social and seasonal variation. Integr Comp Biol 54:68–76CrossRefPubMedPubMedCentral Alvarado S, Fernald RD, Storey KB, Szyf M (2014) The dynamic nature of DNA methylation: a role in response to social and seasonal variation. Integr Comp Biol 54:68–76CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Zardo G, Fazi F, Travaglini L, Nervi C (2005) Dynamic and reversibility of heterochromatic gene silencing in human disease. Cell Res 15:679–690CrossRefPubMed Zardo G, Fazi F, Travaglini L, Nervi C (2005) Dynamic and reversibility of heterochromatic gene silencing in human disease. Cell Res 15:679–690CrossRefPubMed
17.
Zurück zum Zitat Diana J, Simoni Y, Furio L et al (2013) Crosstalk between neutrophils, B-1a cells and plasmacytoid dendritic cells initiates autoimmune diabetes. Nat Med 19:65–73CrossRefPubMed Diana J, Simoni Y, Furio L et al (2013) Crosstalk between neutrophils, B-1a cells and plasmacytoid dendritic cells initiates autoimmune diabetes. Nat Med 19:65–73CrossRefPubMed
18.
Zurück zum Zitat Jayaraman S (2011) Assessment of beta cell viability. Curr Protoc Cytom 55.6.27:6.27.1–6.27.16 Jayaraman S (2011) Assessment of beta cell viability. Curr Protoc Cytom 55.6.27:6.27.1–6.27.16
19.
Zurück zum Zitat Sherry NA, Kushner JA, Glandt M, Kitamura T, Brillantes AM, Herold KC (2006) Effects of autoimmunity and immune therapy on beta-cell turnover in type 1 diabetes. Diabetes 55:3238–3245CrossRefPubMed Sherry NA, Kushner JA, Glandt M, Kitamura T, Brillantes AM, Herold KC (2006) Effects of autoimmunity and immune therapy on beta-cell turnover in type 1 diabetes. Diabetes 55:3238–3245CrossRefPubMed
20.
Zurück zum Zitat Wachlin G, Augstein P, Schroder D et al (2003) IL-1beta, IFN-gamma and TNF-alpha increase vulnerability of pancreatic beta cells to autoimmune destruction. J Autoimmun 20:303–312CrossRefPubMed Wachlin G, Augstein P, Schroder D et al (2003) IL-1beta, IFN-gamma and TNF-alpha increase vulnerability of pancreatic beta cells to autoimmune destruction. J Autoimmun 20:303–312CrossRefPubMed
21.
Zurück zum Zitat Lehuen A, Diana J, Zaccone P, Cooke A (2010) Immune cell crosstalk in type 1 diabetes. Nat Rev Immunol 10:501–513CrossRefPubMed Lehuen A, Diana J, Zaccone P, Cooke A (2010) Immune cell crosstalk in type 1 diabetes. Nat Rev Immunol 10:501–513CrossRefPubMed
22.
Zurück zum Zitat Lakey JR, Suarez-Pinzon WL, Strynadka K et al (2001) Peroxynitrite is a mediator of cytokine-induced destruction of human pancreatic islet beta cells. Lab Invest 81:1683–1692CrossRefPubMed Lakey JR, Suarez-Pinzon WL, Strynadka K et al (2001) Peroxynitrite is a mediator of cytokine-induced destruction of human pancreatic islet beta cells. Lab Invest 81:1683–1692CrossRefPubMed
23.
Zurück zum Zitat Weir GC, Bonner-Weir S (2004) Five stages of evolving beta-cell dysfunction during progression to diabetes. Diabetes 53(Suppl 3):S16–S21CrossRefPubMed Weir GC, Bonner-Weir S (2004) Five stages of evolving beta-cell dysfunction during progression to diabetes. Diabetes 53(Suppl 3):S16–S21CrossRefPubMed
24.
25.
Zurück zum Zitat Akirav EM, Lebastchi J, Galvan EM et al (2011) Detection of beta cell death in diabetes using differentially methylated circulating DNA. Proc Natl Acad Sci U S A 108:19018–19023CrossRefPubMedPubMedCentral Akirav EM, Lebastchi J, Galvan EM et al (2011) Detection of beta cell death in diabetes using differentially methylated circulating DNA. Proc Natl Acad Sci U S A 108:19018–19023CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Yang BT, Dayeh TA, Kirkpatrick CL et al (2011) Insulin promoter DNA methylation correlates negatively with insulin gene expression and positively with HbA(1c) levels in human pancreatic islets. Diabetologia 54:360–367CrossRefPubMedPubMedCentral Yang BT, Dayeh TA, Kirkpatrick CL et al (2011) Insulin promoter DNA methylation correlates negatively with insulin gene expression and positively with HbA(1c) levels in human pancreatic islets. Diabetologia 54:360–367CrossRefPubMedPubMedCentral
27.
28.
Zurück zum Zitat Gilbert ER, Liu D (2012) Epigenetics: the missing link to understanding beta-cell dysfunction in the pathogenesis of type 2 diabetes. Epigenetics 7:841–852CrossRefPubMedPubMedCentral Gilbert ER, Liu D (2012) Epigenetics: the missing link to understanding beta-cell dysfunction in the pathogenesis of type 2 diabetes. Epigenetics 7:841–852CrossRefPubMedPubMedCentral
29.
30.
Zurück zum Zitat Liu CC, Lin JH, Hsu TW et al (2015) IL-6 enriched lung cancer stem-like cell population by inhibition of cell cycle regulators via DNMT1 upregulation. Int J Cancer Journal international du cancer 136:547–559PubMed Liu CC, Lin JH, Hsu TW et al (2015) IL-6 enriched lung cancer stem-like cell population by inhibition of cell cycle regulators via DNMT1 upregulation. Int J Cancer Journal international du cancer 136:547–559PubMed
31.
Zurück zum Zitat Li Y, Deuring J, Peppelenbosch MP, Kuipers EJ, de Haar C, van der Woude CJ (2012) IL-6-induced DNMT1 activity mediates SOCS3 promoter hypermethylation in ulcerative colitis-related colorectal cancer. Carcinogenesis 33:1889–1896CrossRefPubMed Li Y, Deuring J, Peppelenbosch MP, Kuipers EJ, de Haar C, van der Woude CJ (2012) IL-6-induced DNMT1 activity mediates SOCS3 promoter hypermethylation in ulcerative colitis-related colorectal cancer. Carcinogenesis 33:1889–1896CrossRefPubMed
32.
Zurück zum Zitat Cardenas H, Vieth E, Lee J et al (2014) TGF-beta induces global changes in DNA methylation during the epithelial-to-mesenchymal transition in ovarian cancer cells. Epigenetics 9:1461–1472CrossRefPubMedPubMedCentral Cardenas H, Vieth E, Lee J et al (2014) TGF-beta induces global changes in DNA methylation during the epithelial-to-mesenchymal transition in ovarian cancer cells. Epigenetics 9:1461–1472CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Husseiny MI, Kuroda A, Kaye AN, Nair I, Kandeel F, Ferreri K (2012) Development of a quantitative methylation-specific polymerase chain reaction method for monitoring beta cell death in type 1 diabetes. PLoS One 7:e47942CrossRefPubMedPubMedCentral Husseiny MI, Kuroda A, Kaye AN, Nair I, Kandeel F, Ferreri K (2012) Development of a quantitative methylation-specific polymerase chain reaction method for monitoring beta cell death in type 1 diabetes. PLoS One 7:e47942CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Husseiny MI, Kaye A, Zebadua E, Kandeel F, Ferreri K (2014) Tissue-specific methylation of human insulin gene and PCR assay for monitoring beta cell death. PLoS One 9:e94591CrossRefPubMedPubMedCentral Husseiny MI, Kaye A, Zebadua E, Kandeel F, Ferreri K (2014) Tissue-specific methylation of human insulin gene and PCR assay for monitoring beta cell death. PLoS One 9:e94591CrossRefPubMedPubMedCentral
Metadaten
Titel
Methylation of insulin DNA in response to proinflammatory cytokines during the progression of autoimmune diabetes in NOD mice
verfasst von
Jinxiu Rui
Songyan Deng
Jasmin Lebastchi
Pamela L. Clark
Sahar Usmani-Brown
Kevan C. Herold
Publikationsdatum
24.02.2016
Verlag
Springer Berlin Heidelberg
Erschienen in
Diabetologia / Ausgabe 5/2016
Print ISSN: 0012-186X
Elektronische ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-016-3897-4

Weitere Artikel der Ausgabe 5/2016

Diabetologia 5/2016 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.