Epigenetic Mechanisms in Diabetic Kidney Disease
详细信息    查看全文
Thomas MC, Brownlee M, Susztak K, et al. Diabetic kidney disease. Nature Reviews (Disease Primers). July 2015. doi: 10.​1038/​nrdp.​2015.​18 . This collaborative paper provides an expert review of the pathogenesis, epidemiology, diagnosis, management and treatment of diabetic kidney disease.
2.Thomas MC, Groop PH, Tryggvason K. Towards understanding the inherited susceptibility for nephropathy in diabetes. Curr Opin Nephrol Hypertens. 2012;21(2):195–202.CrossRef PubMed
3.Clustering of long-term complications in families with diabetes in the diabetes control and complications trial. The Diabetes Control and Complications Trial Research Group. Diabetes. 1997;46(11):1829-1839
4.Iyengar SK, Abboud HE, Goddard KA, et al. Genome-wide scans for diabetic nephropathy and albuminuria in multiethnic populations: the family investigation of nephropathy and diabetes (FIND). Diabetes. 2007;56(6):1577–85.CrossRef PubMed
5.•
Reddy MA, Zhang E, Natarajan R. Epigenetic mechanisms in diabetic complications and metabolic memory. Diabetologia. 2015;58(3):443–55. This paper provides a state-of-the-art review of evidence pertaining to the role of epigenetic modifications in diabetic complications.CrossRef PubMed PubMedCentral
6.Esteller M. Epigenetics in cancer. N Engl J Med. 2008;358(11):1148–59.CrossRef PubMed
7.Grace BS, Clayton P, McDonald SP. Increases in renal replacement therapy in Australia and New Zealand: understanding trends in diabetic nephropathy. Nephrology. 2012;17(1):76–84.CrossRef PubMed
8.Brasacchio D, Okabe J, Tikellis C, et al. Hyperglycemia induces a dynamic cooperativity of histone methylase and demethylase enzymes associated with gene-activating epigenetic marks that coexist on the lysine tail. Diabetes. 2009;58(5):1229–36.CrossRef PubMed PubMedCentral
9.Kornfeld JW, Bruning JC. Regulation of metabolism by long, non-coding RNAs. Front Genet. 2014;5:57.CrossRef PubMed PubMedCentral
10.••
Guttman M, Rinn JL. Modular regulatory principles of large non-coding RNAs. Nature. 2012;482(7385):339–46. This paper provides an excellent overview of mechanistic evidence pertaining to non-coding RNAs.CrossRef PubMed PubMedCentral
11.Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281–97.CrossRef PubMed
12.Pirola L, Balcerczyk A, Tothill RW, et al. Genome-wide analysis distinguishes hyperglycemia regulated epigenetic signatures of primary vascular cells. Genome Res. 2011;21(10):1601–15.CrossRef PubMed PubMedCentral
13.•
Cooper ME, El-Osta A. Epigenetics: mechanisms and implications for diabetic complications. Circ Res. 2010;107(12):1403–13. This paper also provides a review of evidence pertaining to the role of epigenetic modifications in diabetic complications.CrossRef PubMed
14.Marumo T, Yagi S, Kawarazaki W, et al. Diabetes induces aberrant DNA methylation in the proximal tubules of the kidney. J Am Soc Nephrol. 2015.
15.Richter K, Konzack A, Pihlajaniemi T, Heljasvaara R, Kietzmann T. Redox-fibrosis: impact of TGFbeta1 on ROS generators, mediators and functional consequences. Redox biology. 2015;6:344–52.CrossRef PubMed PubMedCentral
16.Horsburgh S, Robson-Ansley P, Adams R, Smith C. Exercise and inflammation-related epigenetic modifications: focus on DNA methylation. Exerc Immunol Rev. 2015;21:26–41.PubMed
17.Milagro FI, Mansego ML, De Miguel C, Martinez JA. Dietary factors, epigenetic modifications and obesity outcomes: progresses and perspectives. Mol Aspects Med. 2013;34(4):782–812.CrossRef PubMed
18.Caramori ML, Kim Y, Goldfine AB, et al. Differential gene expression in diabetic nephropathy in individuals with type 1 diabetes. J Clin Endocrinol Metab. 2015;100(6):E876–82.CrossRef PubMed
19.Sapienza C, Lee J, Powell J, et al. DNA methylation profiling identifies epigenetic differences between diabetes patients with ESRD and diabetes patients without nephropathy. Epigenetics. 2011;6(1):20–8.CrossRef PubMed
20.Bell CG, Teschendorff AE, Rakyan VK, Maxwell AP, Beck S, Savage DA. Genome-wide DNA methylation analysis for diabetic nephropathy in type 1 diabetes mellitus. BMC Med Genomics. 2010;3:33.CrossRef PubMed PubMedCentral
21.Hasegawa K, Wakino S, Simic P, et al. Renal tubular Sirt1 attenuates diabetic albuminuria by epigenetically suppressing Claudin-1 overexpression in podocytes. Nat Med. 2013;19(11):1496–504.CrossRef PubMed PubMedCentral
22.Swan EJ, Maxwell AP, McKnight AJ. Distinct methylation patterns in genes that affect mitochondrial function are associated with kidney disease in blood-derived DNA from individuals with type 1 diabetes. Diabet Med. 2015;32(8):1110–5.CrossRef PubMed
23.Riviere G, Lienhard D, Andrieu T, Vieau D, Frey BM, Frey FJ. Epigenetic regulation of somatic angiotensin-converting enzyme by DNA methylation and histone acetylation. Epigenetics. 2011;6(4):478–89.CrossRef PubMed
24.El-Osta A, Brasacchio D, Yao D, et al. Transient high glucose causes persistent epigenetic changes and altered gene expression during subsequent normoglycemia. J Exp Med. 2008;205(10):2409–17.CrossRef PubMed PubMedCentral
25.••Okabe J, Orlowski C, Balcerczyk A, et al. Distinguishing hyperglycemic changes by Set7 in vascular endothelial cells. Circ Res. 2012;110(8):1067–76. This paper elaborates one key mechanism through which exposure to elevated glucose levels is able to imprint persistent epigenetic markers.CrossRef PubMed
26.•Miao F, Chen Z, Genuth S, et al. Evaluating the role of epigenetic histone modifications in the metabolic memory of type 1 diabetes. Diabetes. 2014;63(5):1748–62. This paper documents the epigenetic changes induced by intensive control during the Diabetes Control and Complications Trial.CrossRef PubMed PubMedCentral
27.Reddy MA, Sumanth P, Lanting L, et al. Losartan reverses permissive epigenetic changes in renal glomeruli of diabetic db/db mice. Kidney Int. 2014;85(2):362–73.CrossRef PubMed PubMedCentral
28.Wang B, Jha JC, Hagiwara S, et al. Transforming growth factor-beta1-mediated renal fibrosis is dependent on the regulation of transforming growth factor receptor 1 expression by let-7b. Kidney Int. 2014;85(2):352–61.CrossRef PubMed
29.Wang B, Komers R, Carew R, et al. Suppression of microRNA-29 expression by TGF-beta1 promotes collagen expression and renal fibrosis. J Am Soc Nephrol. 2012;23(2):252–65.CrossRef PubMed PubMedCentral
30.Wang B, Koh P, Winbanks C, et al. miR-200a prevents renal fibrogenesis through repression of TGF-beta2 expression. Diabetes. 2011;60(1):280–7.CrossRef PubMed PubMedCentral
31.Zhong X, Chung AC, Chen HY, et al. miR-21 is a key therapeutic target for renal injury in a mouse model of type 2 diabetes. Diabetologia. 2013;56(3):663–74.CrossRef PubMed
32.Cheng X, Ku CH, Siow RC. Regulation of the Nrf2 antioxidant pathway by microRNAs: new players in micromanaging redox homeostasis. Free Radic Biol Med. 2013;64:4–11.CrossRef PubMed
33.Thallas-Bonke V, Jandeleit-Dahm KA, Cooper ME. Nox-4 and progressive kidney disease. Curr Opin Nephrol Hypertens. 2015;24(1):74–80.CrossRef PubMed
34.Mitchell PS, Parkin RK, Kroh EM, et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci U S A. 2008;105(30):10513–8.CrossRef PubMed PubMedCentral
35.Kantharidis P, Wang B, Carew RM, Lan HY. Diabetes complications: the microRNA perspective. Diabetes. 2011;60(7):1832–7.CrossRef PubMed PubMedCentral
36.McClelland A, Hagiwara S, Kantharidis P. Where are we in diabetic nephropathy: microRNAs and biomarkers? Curr Opin Nephrol Hypertens. 2014;23(1):80–6.CrossRef PubMed
37.•EDIC. Sustained effect of intensive treatment of type 1 diabetes mellitus on development and progression of diabetic nephropathy: the Epidemiology of Diabetes Interventions and Complications (EDIC) study. JAMA. 2003;290(16):2159–67. This paper describes the persistent benefits achieved by intensive glucose control during the Diabetes Control and Complications Trial.CrossRef
38.••Chalmers J, Cooper ME. UKPDS and the legacy effect. N Engl J Med. 2008;359(15):1618–20. This editorial discusses the game-changing results of the UKPDS follow-up study and coins the now widely used term “the legacy effect”.CrossRef PubMed
39.Zoungas S, Chalmers J, Neal B, et al. Follow-up of blood-pressure lowering and glucose control in type 2 diabetes. N Engl J Med. 2014;371(15):1392–406.CrossRef PubMed
40.Thomas MC. Glycemic exposure, glycemic control, and metabolic karma in diabetic complications. Adv Chronic Kidney Dis. 2014;21(3):311–7.CrossRef PubMed
41.Bianchi C, Del Prato S. Metabolic memory and individual treatment aims in type 2 diabetes—outcome-lessons learned from large clinical trials. Rev Diabet Stud. 2011;8(3):432–40.CrossRef PubMed PubMedCentral
42.Kowluru RA, Abbas SN, Odenbach S. Reversal of hyperglycemia and diabetic nephropathy: effect of reinstitution of good metabolic control on oxidative stress in the kidney of diabetic rats. J Diabetes Complications. 2004;18(5):282–8.CrossRef PubMed
43.Perera F, Herbstman J. Prenatal environmental exposures, epigenetics, and disease. Reprod Toxicol. 2011;31(3):363–73.CrossRef PubMed PubMedCentral
44.Heijmans BT, Tobi EW, Stein AD, et al. Persistent epigenetic differences associated with prenatal exposure to famine in humans. Proc Natl Acad Sci U S A. 2008;105(44):17046–9.CrossRef PubMed PubMedCentral
45.Binder AM, LaRocca J, Lesseur C, Marsit CJ, Michels KB. Epigenome-wide and transcriptome-wide analyses reveal gestational diabetes is associated with alterations in the human leukocyte antigen complex. Clinical epigenetics. 2015;7(1):79.CrossRef PubMed PubMedCentral
46.••Kaati G, Bygren LO, Edvinsson S. Cardiovascular and diabetes mortality determined by nutrition during parents’ and grandparents’ slow growth period. Eur J Hum Genet. 2002;10(11):682–8. This seminal paper documents the trans-generational effects of diet.CrossRef PubMed
47.Waterland RA, Travisano M, Tahiliani KG. Diet-induced hypermethylation at agouti viable yellow is not inherited transgenerationally through the female. FASEB J. 2007;21(12):3380–5.CrossRef PubMed
48.Slyvka Y, Zhang Y, Nowak FV. Epigenetic effects of paternal diet on offspring: emphasis on obesity. Endocrine. 2015;48(1):36–46.CrossRef PubMed
49.Eggert H, Kurtz J, Diddens-de Buhr MF. Different effects of paternal trans-generational immune priming on survival and immunity in step and genetic offspring. Proceedings Biological sciences / The Royal Society. 2014;281:1797.CrossRef
50.Smyth LJ, Duffy S, Maxwell AP, McKnight AJ. Genetic and epigenetic factors influencing chronic kidney disease. Am J Physiol Renal Physiol. 2014;307(7):F757–76.CrossRef PubMed
51.Henagan TM, Stefanska B, Fang Z, et al. Sodium butyrate epigenetically modulates high-fat diet-induced skeletal muscle mitochondrial adaptation, obesity and insulin resistance through nucleosome positioning. Br J Pharmacol. 2015;172(11):2782–98.CrossRef PubMed
  • 作者单位:Merlin C. Thomas (1) (2)

    1. Baker IDI Heart & Diabetes Institute, 75 Commercial Rd, Melbourne, 3004, Australia
    2. Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia
  • 刊物主题:Diabetes;
  • 出版者:Springer US
  • ISSN:1539-0829
  • 文摘
    Progressive kidney disease is a common companion to both type 1 and type 2 diabetes. However, the majority of people with diabetes do not develop diabetic kidney disease. This may in part be explained by good control of glucose, blood pressure, obesity and other risk factors for kidney disease. It may also be partly due to their genetic makeup or ethnicity. However, the vast majority of the variability in incident nephropathy remains unaccounted for by conventional risk factors or genetics. Epigenetics has recently emerged as an increasingly powerful paradigm to understand and potentially explain complex non-Mendelian conditions—including diabetic kidney disease. Persistent epigenetic changes can be acquired during development or as adaptations to environmental exposure, including metabolic fluctuations associated with diabetes. These epigenetic modifications—including DNA methylation, histone modifications, non-coding RNAs and other changes in chromatin structure and function—individually and co-operatively act to register, store, retain and recall past experiences in a way to shape the transcription of specific genes and, therefore, cellular functions. This review will explore the emerging evidence for the role of epigenetic modifications in programming the legacy of hyperglycaemia for kidney disease in diabetes.

    © 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

    地址:北京市海淀区学院路29号 邮编:100083

    电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700