Nutr Res Pract.  2017 Apr;11(2):105-113. 10.4162/nrp.2017.11.2.105.

Genome-wide hepatic DNA methylation changes in high-fat diet-induced obese mice

Affiliations
  • 1Department of Food and Nutrition, College of Human Ecology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea. snhan@snu.ac.kr
  • 2Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA.
  • 3Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA 02111, USA.
  • 4Research Institute of Human Ecology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea.
  • 5Chaum Life Center, CHA University School of Medicine, Seoul 06062, Korea.

Abstract

BACKGROUND/OBJECTIVES
A high-fat diet (HFD) induces obesity, which is a major risk factor for cardiovascular disease and cancer, while a calorie-restricted diet can extend life span by reducing the risk of these diseases. It is known that health effects of diet are partially conveyed through epigenetic mechanism including DNA methylation. In this study, we investigated the genome-wide hepatic DNA methylation to identify the epigenetic effects of HFD-induced obesity.
MATERIALS AND METHODS
Seven-week-old male C57BL/6 mice were fed control diet (CD), calorie-restricted control diet (CRCD), or HFD for 16 weeks (after one week of acclimation to the control diet). Food intake, body weight, and liver weight were measured. Hepatic triacylglycerol and cholesterol levels were determined using enzymatic colorimetric methods. Changes in genome-wide DNA methylation were determined by a DNA methylation microarray method combined with methylated DNA immunoprecipitation. The level of transcription of individual genes was measured by real-time PCR.
RESULTS
The DNA methylation statuses of genes in biological networks related to lipid metabolism and hepatic steatosis were influenced by HFD-induced obesity. In HFD group, a proinflammatory Casp1 (Caspase 1) gene had hypomethylated CpG sites at the 1.5-kb upstream region of its transcription start site (TSS), and its mRNA level was higher compared with that in CD group. Additionally, an energy metabolism-associated gene Ndufb9 (NADH dehydrogenase 1 beta subcomplex 9) in HFD group had hypermethylated CpG sites at the 2.6-kb downstream region of its TSS, and its mRNA level was lower compared with that in CRCD group.
CONCLUSIONS
HFD alters DNA methylation profiles in genes associated with liver lipid metabolism and hepatic steatosis. The methylation statuses of Casp1 and Ndufb9 were particularly influenced by the HFD. The expression of these genes in HFD differed significantly compared with CD and CRCD, respectively, suggesting that the expressions of Casp1 and Ndufb9 in liver were regulated by their methylation statuses.

Keyword

DNA methylation; obesity; liver; Caspase 1; NADH dehydrogenase

MeSH Terms

Acclimatization
Animals
Body Weight
Cardiovascular Diseases
Caspase 1
Cholesterol
Diet
Diet, High-Fat
DNA Methylation*
DNA*
Eating
Epigenomics
Humans
Immunoprecipitation
Lipid Metabolism
Liver
Male
Methods
Methylation
Mice
Mice, Obese*
NADH Dehydrogenase
Obesity
Oxidoreductases
Real-Time Polymerase Chain Reaction
Risk Factors
RNA, Messenger
Transcription Initiation Site
Triglycerides
Caspase 1
Cholesterol
DNA
NADH Dehydrogenase
Oxidoreductases
RNA, Messenger
Triglycerides

Figure

  • Fig. 1 Heat maps showing differentially methylated probes in each comparison. (A) CD vs. CRCD comparison. (B) HFD vs. CD comparison. (C) HFD vs. CRCD comparison. CRCD, calorie-restricted control diet; CD, control diet; HFD, high-fat diet.

  • Fig. 2 Distance between DMRs and their annotated genes. The number of DMRs falling within the designated intervals relative to their annotated genes' transcription start sites (TSSs) are presented above each bar. This graph was generated via Stanford University's GREAT Analysis. DMRs: differentially methylated regions.

  • Fig. 3 Network 1 of the HFD vs. CD comparison. This network is involved in gastrointestinal disease, hepatic system disease, and liver steatosis. Red: Genes found in our data that were hypomethylated in HFD group. Green: Genes found in our data that were hypermethylated in HFD group. HFD, high-fat diet; CD, control diet.

  • Fig. 4 Relative level of Casp1 mRNA in the HFD group compared with that of the CD group. The value are expressed as means ± SEM, n = 7 for each group. Student's t-test P-value is shown. HFD, high-fat diet; CD, control diet.

  • Fig. 5 Network 1 of the HFD vs. CRCD comparison. This network is involved in lipid metabolism, molecular transport, and small molecule biochemistry. Red: Genes found in our data that were hypomethylated in HFD group. Green: Genes found in our data that were hypermethylated in HFD group. HFD, high-fat diet; CRCD, calorie-restricted control diet.

  • Fig. 6 Relative level of Ndufb9 mRNA inthe HFD group compared with that of the CRCD group. The values are expressed as means ± SEM, n = 6 for CRCD and 7 for HFD. Student's t-test P-value is shown. HFD, high-fat diet; CRCD, calorie-restricted control diet.


Reference

1. Lim SS, Vos T, Flaxman AD, Danaei G, Shibuya K, Adair-Rohani H, Amann M, Anderson HR, Andrews KG, Aryee M, Atkinson C, Bacchus LJ, Bahalim AN, Balakrishnan K, Balmes J, Barker-Collo S, Baxter A, Bell ML, Blore JD, Blyth F, Bonner C, Borges G, Bourne R, Boussinesq M, Brauer M, Brooks P, Bruce NG, Brunekreef B, Bryan-Hancock C, Bucello C, Buchbinder R, Bull F, Burnett RT, Byers TE, Calabria B, Carapetis J, Carnahan E, Chafe Z, Charlson F, Chen H, Chen JS, Cheng AT, Child JC, Cohen A, Colson KE, Cowie BC, Darby S, Darling S, Davis A, Degenhardt L, Dentener F, Des Jarlais DC, Devries K, Dherani M, Ding EL, Dorsey ER, Driscoll T, Edmond K, Ali SE, Engell RE, Erwin PJ, Fahimi S, Falder G, Farzadfar F, Ferrari A, Finucane MM, Flaxman S, Fowkes FG, Freedman G, Freeman MK, Gakidou E, Ghosh S, Giovannucci E, Gmel G, Graham K, Grainger R, Grant B, Gunnell D, Gutierrez HR, Hall W, Hoek HW, Hogan A, Hosgood HD 3rd, Hoy D, Hu H, Hubbell BJ, Hutchings SJ, Ibeanusi SE, Jacklyn GL, Jasrasaria R, Jonas JB, Kan H, Kanis JA, Kassebaum N, Kawakami N, Khang YH, Khatibzadeh S, Khoo JP, Kok C, Laden F, Lalloo R, Lan Q, Lathlean T, Leasher JL, Leigh J, Li Y, Lin JK, Lipshultz SE, London S, Lozano R, Lu Y, Mak J, Malekzadeh R, Mallinger L, Marcenes W, March L, Marks R, Martin R, McGale P, McGrath J, Mehta S, Mensah GA, Merriman TR, Micha R, Michaud C, Mishra V, Mohd Hanafiah K, Mokdad AA, Morawska L, Mozaffarian D, Murphy T, Naghavi M, Neal B, Nelson PK, Nolla JM, Norman R, Olives C, Omer SB, Orchard J, Osborne R, Ostro B, Page A, Pandey KD, Parry CD, Passmore E, Patra J, Pearce N, Pelizzari PM, Petzold M, Phillips MR, Pope D, Pope CA 3rd, Powles J, Rao M, Razavi H, Rehfuess EA, Rehm JT, Ritz B, Rivara FP, Roberts T, Robinson C, Rodriguez-Portales JA, Romieu I, Room R, Rosenfeld LC, Roy A, Rushton L, Salomon JA, Sampson U, Sanchez-Riera L, Sanman E, Sapkota A, Seedat S, Shi P, Shield K, Shivakoti R, Singh GM, Sleet DA, Smith E, Smith KR, Stapelberg NJ, Steenland K, Stöckl H, Stovner LJ, Straif K, Straney L, Thurston GD, Tran JH, Van Dingenen R, van Donkelaar A, Veerman JL, Vijayakumar L, Weintraub R, Weissman MM, White RA, Whiteford H, Wiersma ST, Wilkinson JD, Williams HC, Williams W, Wilson N, Woolf AD, Yip P, Zielinski JM, Lopez AD, Murray CJ, Ezzati M, AlMazroa MA, Memish ZA. A comparative risk assessment of burden of disease and injury attributable to 67 risk factors and risk factor clusters in 21 regions, 1990-2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet. 2012; 380:2224–2260. PMID: 23245609.
Article
2. Eckel RH, Grundy SM, Zimmet PZ. The metabolic syndrome. Lancet. 2005; 365:1415–1428. PMID: 15836891.
Article
3. Seppälä-Lindroos A, Vehkavaara S, Häkkinen AM, Goto T, Westerbacka J, Sovijärvi A, Halavaara J, Yki-Järvinen H. Fat accumulation in the liver is associated with defects in insulin suppression of glucose production and serum free fatty acids independent of obesity in normal men. J Clin Endocrinol Metab. 2002; 87:3023–3028. PMID: 12107194.
Article
4. Kotronen A, Westerbacka J, Bergholm R, Pietiläinen KH, Yki-Järvinen H. Liver fat in the metabolic syndrome. J Clin Endocrinol Metab. 2007; 92:3490–3497. PMID: 17595248.
Article
5. Ridker PM, Buring JE, Cook NR, Rifai N. C-reactive protein, the metabolic syndrome, and risk of incident cardiovascular events: an 8-year follow-up of 14 719 initially healthy American women. Circulation. 2003; 107:391–397. PMID: 12551861.
6. Mertens I, Verrijken A, Michiels JJ, Van der Planken M, Ruige JB, Van Gaal LF. Among inflammation and coagulation markers, PAI-1 is a true component of the metabolic syndrome. Int J Obes (Lond). 2006; 30:1308–1314. PMID: 16389265.
Article
7. Park EJ, Lee JH, Yu GY, He G, Ali SR, Holzer RG, Österreicher CH, Takahashi H, Karin M. Dietary and genetic obesity promote liver inflammation and tumorigenesis by enhancing IL-6 and TNF expression. Cell. 2010; 140:197–208. PMID: 20141834.
Article
8. Choi SW, Friso S. Epigenetics: a new bridge between nutrition and health. Adv Nutr. 2010; 1:8–16. PMID: 22043447.
Article
9. Milagro FI, Campión J, García-Díaz DF, Goyenechea E, Paternain L, Martínez JA. High fat diet-induced obesity modifies the methylation pattern of leptin promoter in rats. J Physiol Biochem. 2009; 65:1–9. PMID: 19588726.
Article
10. Jiang M, Zhang Y, Liu M, Lan MS, Fei J, Fan W, Gao X, Lu D. Hypermethylation of hepatic glucokinase and L-type pyruvate kinase promoters in high-fat diet-induced obese rats. Endocrinology. 2011; 152:1284–1289. PMID: 21239437.
Article
11. Schwenk RW, Jonas W, Ernst SB, Kammel A, Jähnert M, Schürmann A. Diet-dependent alterations of hepatic Scd1 expression are accompanied by differences in promoter methylation. Horm Metab Res. 2013; 45:786–794. PMID: 23803969.
Article
12. Chang X, Yan H, Fei J, Jiang M, Zhu H, Lu D, Gao X. Berberine reduces methylation of the MTTP promoter and alleviates fatty liver induced by a high-fat diet in rats. J Lipid Res. 2010; 51:2504–2515. PMID: 20567026.
Article
13. Dudley KJ, Sloboda DM, Connor KL, Beltrand J, Vickers MH. Offspring of mothers fed a high fat diet display hepatic cell cycle inhibition and associated changes in gene expression and DNA methylation. PLoS One. 2011; 6:e21662. PMID: 21779332.
Article
14. Zhang Y, Wang H, Zhou D, Moody L, Lezmi S, Chen H, Pan YX. High-fat diet caused widespread epigenomic differences on hepatic methylome in rat. Physiol Genomics. 2015; 47:514–523. PMID: 26199400.
Article
15. Richardson B. Impact of aging on DNA methylation. Ageing Res Rev. 2003; 2:245–261. PMID: 12726774.
Article
16. Folch J, Lees M, Sloane Stanley GH. A simple method for the isolation and purification of total lipides from animal tissues. J Biol Chem. 1957; 226:497–509. PMID: 13428781.
Article
17. Jin SG, Kadam S, Pfeifer GP. Examination of the specificity of DNA methylation profiling techniques towards 5-methylcytosine and 5-hydroxymethylcytosine. Nucleic Acids Res. 2010; 38:e125. PMID: 20371518.
Article
18. Irizarry RA, Ladd-Acosta C, Carvalho B, Wu H, Brandenburg SA, Jeddeloh JA, Wen B, Feinberg AP. Comprehensive high-throughput arrays for relative methylation (CHARM). Genome Res. 2008; 18:780–790. PMID: 18316654.
Article
19. Tammen SA, Park LK, Dolnikowski GG, Ausman LM, Friso S, Choi SW. Hepatic DNA hydroxymethylation is site-specifically altered by chronic alcohol consumption and aging. Eur J Nutr. Forthcoming 2015.
Article
20. McLean CY, Bristor D, Hiller M, Clarke SL, Schaar BT, Lowe CB, Wenger AM, Bejerano G. GREAT improves functional interpretation of cis-regulatory regions. Nat Biotechnol. 2010; 28:495–501. PMID: 20436461.
Article
21. Fujita PA, Rhead B, Zweig AS, Hinrichs AS, Karolchik D, Cline MS, Goldman M, Barber GP, Clawson H, Coelho A, Diekhans M, Dreszer TR, Giardine BM, Harte RA, Hillman-Jackson J, Hsu F, Kirkup V, Kuhn RM, Learned K, Li CH, Meyer LR, Pohl A, Raney BJ, Rosenbloom KR, Smith KE, Haussler D, Kent WJ. The UCSC Genome Browser database: update 2011. Nucleic Acids Res. 2011; 39:D876–D882. PMID: 20959295.
Article
22. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Δ Δ C(T)) method. Methods. 2001; 25:402–408. PMID: 11846609.
23. Franchi L, Eigenbrod T, Muñoz-Planillo R, Nuñez G. The inflammasome: a caspase-1-activation platform that regulates immune responses and disease pathogenesis. Nat Immunol. 2009; 10:241–247. PMID: 19221555.
Article
24. Smeitink J, van den Heuvel L. Human mitochondrial complex I in health and disease. Am J Hum Genet. 1999; 64:1505–1510. PMID: 10330338.
Article
25. Haack TB, Madignier F, Herzer M, Lamantea E, Danhauser K, Invernizzi F, Koch J, Freitag M, Drost R, Hillier I, Haberberger B, Mayr JA, Ahting U, Tiranti V, Rötig A, Iuso A, Horvath R, Tesarova M, Baric I, Uziel G, Rolinski B, Sperl W, Meitinger T, Zeviani M, Freisinger P, Prokisch H. Mutation screening of 75 candidate genes in 152 complex I deficiency cases identifies pathogenic variants in 16 genes including NDUFB9. J Med Genet. 2012; 49:83–89. PMID: 22200994.
26. Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet. 2012; 13:484–492. PMID: 22641018.
Article
27. Ge ZJ, Luo SM, Lin F, Liang QX, Huang L, Wei YC, Hou Y, Han ZM, Schatten H, Sun QY. DNA methylation in oocytes and liver of female mice and their offspring: effects of high-fat-diet-induced obesity. Environ Health Perspect. 2014; 122:159–164. PMID: 24316659.
Article
28. Murphy SK, Yang H, Moylan CA, Pang H, Dellinger A, Abdelmalek MF, Garrett ME, Ashley-Koch A, Suzuki A, Tillmann HL, Hauser MA, Diehl AM. Relationship between methylome and transcriptome in patients with nonalcoholic fatty liver disease. Gastroenterology. 2013; 145:1076–1087. PMID: 23916847.
Article
29. Choi MS, Kim YJ, Kwon EY, Ryoo JY, Kim SR, Jung UJ. High-fat diet decreases energy expenditure and expression of genes controlling lipid metabolism, mitochondrial function and skeletal system development in the adipose tissue, along with increased expression of extracellular matrix remodelling- and inflammation-related genes. Br J Nutr. 2015; 113:867–877. PMID: 25744306.
Article
30. Jones PA. The DNA methylation paradox. Trends Genet. 1999; 15:34–37. PMID: 10087932.
Article
31. Lorincz MC, Dickerson DR, Schmitt M, Groudine M. Intragenic DNA methylation alters chromatin structure and elongation efficiency in mammalian cells. Nat Struct Mol Biol. 2004; 11:1068–1075. PMID: 15467727.
Article
32. National Research Council (US) Subcommittee on Laboratory Animal Nutrition. Nutrient Requirements of Laboratory Animals. 4th rev. ed. Washington (D.C.): National Academies Press;1995.
Full Text Links
  • NRP
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr