Skip to main content

Distribution of Methylated Regions Within gDNA in Acute and Chronic Phases of Diabetes Mellitus

  • Reference work entry
  • First Online:
Handbook of Nutrition, Diet, and Epigenetics

Abstract

Diabetes mellitus (DM) is a disease of metabolic dysregulation involving the induction of hyperglycemia due to (1) loss of functioning of pancreatic beta cells that produce insulin (t1 DM) or (2) the inability of systemic cells to properly respond to the insulin signal (t2 DM). The resulting hyperglycemic episodes induce long-term complications in a broad spectrum of organs/tissues such as seen in the cardiovascular system (CV). One aspect of CV dysregulation is seen in abnormalities in blood vessel formation (BVF). These abnormalities in BVF are seen in the acute and chronic states of DM, with the latter chronic effects termed “metabolic memory” (MM). The heritable nature of metabolic memory indicates a role for the epigenome as a contributing factor in DM and MM. In this regard, the epigenome is comprised of all chromatin-modifying processes such as DNA methylation and histone modifications that allow cells and organisms to quickly respond to changing environmental stimuli. The current review focuses on the global patterns of gDNA methylation in Control, DM, and MM groups using a zebrafish DM/MM model of the disease. For this review, global analysis is focused on 10 Kb upstream from the TSS (transcription start site), 1 Kb upstream from the TSS, and 300 bp downstream of the TSS for all genes of the zebrafish genome. Analysis of gDNA methylation patterns will also be studied in regard to genes that regulate BVF, and we have included genes that control DNA replication and repair in this regulatory group. As will be discussed, hyperglycemia not only induces changes in gDNA methylation but complex changes in the pattern seen in the methylation and demethylation of BVF regulatory genes.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 579.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Hardcover Book
USD 799.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Abbreviations

AGE:

Advanced glycation end products

CH3:

Methyl group

CpG:

Cytosine-phosphate-guanine

DM:

Diabetes mellitus

gDNA:

Genomic DNA

MeDIP:

Methylated DNA immunoprecipitation

MM:

Metabolic memory

MRs:

Methylated regions

MSs:

Methylated sites

ROS:

Reactive oxygen species

STZ:

Streptozotocin

TF:

Transcription factor

TSS:

Transcription start site

References

  • Baynes JW (1991) Role of oxidative stress in development of complications in diabetes. Diabetes 40(4):405–412

    Article  CAS  Google Scholar 

  • Blankenberg D, Kuster G, Coraor N, Ananda G, Lazarus R, Mangan M, Nekrutenko A, Taylor J (2010) Galaxy: a web-based genome analysis tool for experimentalists. Curr Protocol Mol Biol 19:11–21. SRC – GoogleScholar: 1–2

    Google Scholar 

  • Brownlee M (2005) The pathobiology of diabetic complications: a unifying mechanism. Diabetes 54(6):1615–1625

    Article  CAS  Google Scholar 

  • Capiotti KM, Antonioli R Jr, Kist LW, Bogo MR, Bonan CD, Da Silva RS (2014) Persistent impaired glucose metabolism in a zebrafish hyperglycemia model. Comp Biochem Physiol B Biochem Mol Biol 171:58–65

    Article  CAS  Google Scholar 

  • Chan PS, Kanwar M, Kowluru RA (2010) Resistance of retinal inflammatory mediators to suppress after reinstitution of good glycemic control: novel mechanism for metabolic memory. J Diabetes Complicat 24(1):55–63

    Article  Google Scholar 

  • Costa P, Soares R (2013) Neovascularization in diabetes and its complications. Unraveling the angiogenic paradox. Life Sci 92(22):1037–1045

    Article  CAS  Google Scholar 

  • Dolinoy DC, Jirtle RL (2008) Environmental epigenomics in human health and disease. Environ Mol Mutagen 49(1):4–8

    Article  CAS  Google Scholar 

  • Dunoyer-Geindre S, Rivier-Cordey AS, Caetano C, Fish RJ, Kruithof EK (2016) Effect of regulatory element DNA methylation on tissue-type plasminogen activator gene expression. PLoS One 11(12):e0167588

    Article  Google Scholar 

  • Engerman RL, Kern TS (1987) Progression of incipient diabetic retinopathy during good glycemic control. Diabetes 36(7):808–812

    Article  CAS  Google Scholar 

  • Goecks J, Nekrutenko A, Taylor J (2010) Galaxy: a comprehensive approach for supporting accessible, reproducible, and transparent computational research in the life sciences. Genome Biol 11(8):R86

    Article  Google Scholar 

  • Hammes HP, Klinzing I, Wiegand S, Bretzel RG, Cohen AM, Federlin K (1993) Islet transplantation inhibits diabetic retinopathy in the sucrose-fed diabetic Cohen rat. Invest Ophthalmol Vis Sci 34(6):2092–2096

    CAS  PubMed  Google Scholar 

  • Han Y, He X (2016) Integrating epigenomics into the understanding of biomedical insight. Bioinform Biol Insights 10:267–289

    Article  CAS  Google Scholar 

  • Inzucchi S, Majumdar S (2015) Glycemic targets: what is the evidence? Med Clin N Am 99(1):47–67

    Article  Google Scholar 

  • Jhamb S, Vangaveti VN, Malabu UH (2016) Genetic and molecular basis of diabetic foot ulcers: clinical review. J Tissue Viability 25(4):229–236

    Article  Google Scholar 

  • Kimmel RA, Dobler S, Schmitner N, Walsen T, Freudenblum J, Meyer D (2015) Diabetic pdx1-mutant zebrafish show conserved responses to nutrient overload and anti-glycemic treatment. Sci Rep 5:14241

    Article  CAS  Google Scholar 

  • Leontovich AAIRV, Sarras MP Jr (2016) Epigenetic studies point to DNA replicaton/repair genes as a basis for the heritable nature of long term complications in diabetes. J Diabetes Res 2016:1–10

    Article  Google Scholar 

  • Liew G, Wong VW, Ho IV (2017) Mini review: changes in the incidence of and progression to proliferative and sight-threatening diabetic retinopathy over the last 30 years. Ophthalmic Epidemiol 24:73–80. 1–8

    Article  Google Scholar 

  • Morgan DK, Whitelaw E (2008) The case for transgenerational epigenetic inheritance in humans. Mamm Genome Off J Int Mamm Genome Soc 19(6):394–397

    Article  Google Scholar 

  • Olsen AS, Sarras MP, Intine RV (2010) Limb regeneration is impaired in an adult zebrafish model of diabetes mellitus. Wound Repair Regen Off Publ Wound Healing Soc Eur Tissue Repair Soc 18(5):532–542

    Google Scholar 

  • Olsen AS, Sarras MP, Leontovich A, Intine RV,D (2012) Heritable transmission of diabetic metabolic memory in zebrafish correlates with and aberrant gene expression. Diabetes 61(2):485–491

    Article  CAS  Google Scholar 

  • Patschan D, Muller GA (2016) Acute kidney injury in diabetes mellitus. Int J Nephrol 2016:6232909

    Article  CAS  Google Scholar 

  • Prattichizzo F, Giuliani A, Ceka A, Rippo MR, Bonfigli AR, Testa R, Procopio AD, Olivieri F (2015) Epigenetic mechanisms of endothelial dysfunction in type 2 diabetes. Clin Epigenetics 7(1):56

    Article  Google Scholar 

  • Putta S, Lanting L, Sun G, Lawson G, Kato M, Natarajan R (2012) Inhibiting microRNA-192 ameliorates renal fibrosis in diabetic nephropathy. J Am Soc Nephrol JASN 23(3):458–469

    Article  CAS  Google Scholar 

  • Rando OJ (2012) Combinatorial complexity in chromatin structure and function: revisiting the histone code. Curr Opin Genet Dev 22(2):148–155

    Article  CAS  Google Scholar 

  • Riddle MC (2010) Effects of intensive glucose lowering in the management of patients with type 2 diabetes mellitus in the Action to Control Cardiovascular Risk in Diabetes (ACCORD) trial. Circulation 122(8):844–846

    Article  Google Scholar 

  • Roy S, Sala R, Cagliero E, Lorenzi M (1990) Overexpression of fibronectin induced by diabetes or high glucose: phenomenon with a memory. Proc Natl Acad Sci U S A 87(1):404–408

    Article  CAS  Google Scholar 

  • Sarras MP, Leontovich AA, Olsen AS, Intine RV (2013) Impaired tissue regeneration corresponds with altered expression of developmental genes that persists in the metabolic memory state of diabetic zebrafish. Wound Repair Regen Off Publ Wound Healing Soc Eur Tissue Repair Soc 21(2):320–328

    Google Scholar 

  • Sarras MP Jr, Mason S, McAllister G, Intine RV (2014) Inhibition of poly-ADP ribose polymerase enzyme activity prevents hyperglycemia-induced impairment of angiogenesis during wound healing. Wound Repair Regen 22(5):666–670

    Article  Google Scholar 

  • Sarras MP Jr, Leontovich AA, Intine RV (2015) Use of zebrafish as a model to investigate the role of epigenetics in propagating the secondary complications observed in diabetes mellitus. Comp Biochem Physiol C Toxicol Pharmacol 178:3–7

    Article  CAS  Google Scholar 

  • Skyler JS, Bergenstal R, Bonow RO, Buse J, Deedwania P, Gale EAM, Howard BV, Kirkman MS, Kosiborod M, Reaven P, Sherwin RS, American Diabetes Association, American College of Cardiology Foundation, American Heart Association (2009) Intensive glycemic control and the prevention of cardiovascular events: implications of the ACCORD, ADVANCE, and VA diabetes trials: a position statement of the American Diabetes Association and a scientific statement of the American College of Cardiology Foundation and the American Heart Association. Diabetes Care 32(1):187–192

    Article  Google Scholar 

  • Spitz F, Furlong EE (2012) Transcription factors: from enhancer binding to developmental control. Nat Rev Genet 13(9):613–626

    Article  CAS  Google Scholar 

  • Yoon Y-S (2013) Reprogramming diabetic stem or progenitor cells for treating diabetic complications. Trans-NIH Angiogenesis Workshop, Division of Cancer Prevention. Lister Hill, NIH Main Campus, Division of Cancer Prevention

    Google Scholar 

  • Zhang Y, Liu T, Meyer CA, Eeckhoute J, Johnson DS, Bernstein BE, Nusbaum C, Myers RM, Brown M, Li W, Liu XS (2008) Model-based analysis of ChIP-Seq (MACS). Genome Biol 9(9):R137

    Article  Google Scholar 

  • Zhang H, Cai X, Yi B, Huang J, Wang J, Sun J (2014) Correlation of CTGF gene promoter methylation with CTGF expression in type 2 diabetes mellitus with or without nephropathy. Mol Med Rep 9(6):2138–2144

    Article  CAS  Google Scholar 

  • Zhang Q, Lenardo MJ, Baltimore D (2017) 30 years of NF-kappaB: a blossoming of relevance to human pathobiology. Cell 168(1–2):37–57

    Article  CAS  Google Scholar 

  • Zhong Q, Kowluru RA (2010) Role of histone acetylation in the development of diabetic retinopathy and the metabolic memory phenomenon. J Cell Biochem 110(6):1306–1313

    Article  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Michael P. Sarras Jr. .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2019 Springer Nature Switzerland AG

About this entry

Check for updates. Verify currency and authenticity via CrossMark

Cite this entry

Leontovich, A.A., Sarras, M.P. (2019). Distribution of Methylated Regions Within gDNA in Acute and Chronic Phases of Diabetes Mellitus. In: Patel, V., Preedy, V. (eds) Handbook of Nutrition, Diet, and Epigenetics. Springer, Cham. https://doi.org/10.1007/978-3-319-55530-0_76

Download citation

  • DOI: https://doi.org/10.1007/978-3-319-55530-0_76

  • Published:

  • Publisher Name: Springer, Cham

  • Print ISBN: 978-3-319-55529-4

  • Online ISBN: 978-3-319-55530-0

  • eBook Packages: MedicineReference Module Medicine

Publish with us

Policies and ethics