Obstet Gynecol Sci.  2020 Apr;63(3):239-250. 10.5468/ogs.2020.63.3.239.

Long-term effects of pro-opiomelanocortin methylation induced in food-restricted dams on metabolic phenotypes in male rat offspring

Affiliations
  • 1Departments of Obstetrics and Gynecology and Medical Research Institute,, College of Medicine, Ewha Womans University, Seoul, Korea
  • 2Departments of Molecular Medicine, College of Medicine, Ewha Womans University, Seoul, Korea

Abstract


Objective
Maternal malnutrition affects the growth and metabolic health of the offspring. Little is known about the longterm effect on metabolic indices of epigenetic changes in the brain caused by maternal diet. Thus, we explored the effect of maternal food restriction during pregnancy on metabolic profiles of the offspring, by evaluating the DNA methylation of hypothalamic appetite regulators at 3 weeks of age.
Methods
Sprague-Dawley rats were divided into 2 groups: a control group and a group with a 50% food-restricted (FR) diet during pregnancy. Methylation and expression of appetite regulator genes were measured in 3-week-old offspring using pyrosequencing, real-time polymerase chain reaction, and western blotting analyses. We analyzed the relationship between DNA methylation and metabolic profiles by Pearson’s correlation analysis.
Results
The expression of pro-opiomelanocortin (POMC) decreased, whereas DNA methylation significantly increased in male offspring of the FR dams, compared to the male offspring of control dams. Hypermethylation of POMC was positively correlated with the levels of high-density lipoprotein cholesterol (HDL-C) and low-density lipoprotein cholesterol in 3-week-old male offspring. In addition, there were significant positive correlations between hypermethylation of POMC and the levels of triglycerides, HDL-C, and leptin in 6-month-old male offspring.
Conclusion
Our findings suggest that maternal food restriction during pregnancy influences the expression of hypothalamic appetite regulators via epigenetic changes, leading to the development of metabolic disorders in the offspring.

Keyword

Pro-opiomelanocortin; Appetite regulation; DNA methylation; Metabolic syndrome; Malnutrition

Figure

  • Fig. 1 Experimental design. Eight-week-old male and female Sprague-Dawley (SD) rats (n=8/group) were used in this study. At day 10 of gestation, pregnant SD rats were divided into 2 groups: 1) a control group, fed a normal diet throughout the whole experimental period and 2) a food-restriction (FR) group, fed a 50% FR diet during pregnancy and a normal diet after delivery.

  • Fig. 2 Relative mRNA and protein expression levels of pro-opiomelanocortin (POMC), neuropeptide Y (NPY), and melanocortin 4 receptor (MC4R) in the hypothalamus of 3-week-old offspring. (A) The mRNA expression levels of POMC, NPY, and MC4R were analyzed using real-time PCR in the food-restricted (FR) and control (C) groups (n=9 males, 9 females/group). Data are presented as means±standard deviation (SD). (B) The protein expression levels of POMC, NPY, and MC4R were analyzed using western blotting in the FR and C groups (n=9 males, 9 females/group). Data are presented as means±SD.GAPDH, glyceraldehyde 3-phosphate dehydrogenase.a)P<0.05; b)P<0.01; c)P<0.001.

  • Fig. 3 Hypothalamic pro-opiomelanocortin (POMC) methylation in male (A) and female (B) offspring at 3 weeks of age (n=9 males, 9 females/group). POMC methylation was measured by pyrosequencing in offspring of the food-restricted (FR) and control (C) dams. Data are presented as means±standard deviation.a)P<0.05.

  • Fig. 4 Hypothalamic neuropeptide Y (NPY) methylation in male and female offspring at 3 weeks of age (n=9 males, 9 females/group). (A) Methylation of promoter region 1 and (B) promoter region 2 of the NPY gene. Data are presented as means±standard deviation.C, control; FR, food-restricted.

  • Fig. 5 Hypothalamic melanocortin-4 receptor (MC4R) methylation in male and female offspring at 3 weeks of age (n=9 males, 9 females/group). (A) DNA methylation of promoter region 1 in and (B) promoter region 2 of the MC4R gene. Values are presented as means±standard deviation.C, control; FR, food-restricted.


Cited by  1 articles

Prenatal maternal alcohol exposure: diagnosis and prevention of fetal alcohol syndrome
Young Min Hur, Jiwon Choi, Sunwha Park, Sarah Soyeon Oh, Young Ju Kim
Obstet Gynecol Sci. 2022;65(5):385-394.    doi: 10.5468/ogs.22123.


Reference

1. Barker DJ, Hales CN, Fall CH, Osmond C, Phipps K, Clark PM. Type 2 (non-insulin-dependent) diabetes mellitus, hypertension and hyperlipidaemia (syndrome X): relation to reduced fetal growth. Diabetologia. 1993; 36:62–67. PMID: 8436255.
Article
2. Barker DJ. The developmental origins of chronic adult disease. Acta Paediatr Suppl. 2004; 93:26–33.
Article
3. Coupé B, Grit I, Darmaun D, Parnet P. The timing of “catch-up growth” affects metabolism and appetite regulation in male rats born with intrauterine growth restriction. Am J Physiol Regul Integr Comp Physiol. 2009; 297:R813–24. PMID: 19605764.
Article
4. Breier BH, Vickers MH, Ikenasio BA, Chan KY, Wong WP. Fetal programming of appetite and obesity. Mol Cell Endocrinol. 2001; 185:73–79. PMID: 11738796.
Article
5. Kwon EJ, Kim YJ. What is fetal programming?: a lifetime health is under the control of in utero health. Obstet Gynecol Sci. 2017; 60:506–519. PMID: 29184858.
Article
6. Lee S, Lee KA, Choi GY, Desai M, Lee SH, Pang MG, et al. Feed restriction during pregnancy/lactation induces programmed changes in lipid, adiponectin and leptin levels with gender differences in rat offspring. J Matern Fetal Neonatal Med. 2013; 26:908–914. PMID: 23327414.
Article
7. Desai M, Gayle D, Babu J, Ross MG. The timing of nutrient restriction during rat pregnancy/lactation alters metabolic syndrome phenotype. Am J Obstet Gynecol. 2007; 196:555.e1–555.e7. PMID: 17547893.
Article
8. Roseboom TJ, van der Meulen JH, Osmond C, Barker DJ, Ravelli AC, Bleker OP. Plasma lipid profiles in adults after prenatal exposure to the Dutch famine. Am J Clin Nutr. 2000; 72:1101–1106. PMID: 11063435.
Article
9. López M, Tovar S, Vázquez MJ, Williams LM, Diéguez C. Peripheral tissue-brain interactions in the regulation of food intake. Proc Nutr Soc. 2007; 66:131–155. PMID: 17343779.
Article
10. Arora S, Anubhuti . Role of neuropeptides in appetite regulation and obesity--a review. Neuropeptides. 2006; 40:375–401. PMID: 16935329.
11. Gali Ramamoorthy T, Begum G, Harno E, White A. Developmental programming of hypothalamic neuronal circuits: impact on energy balance control. Front Neurosci. 2015; 9:126. PMID: 25954145.
Article
12. Loftus TM. An adipocyte-central nervous system regulatory loop in the control of adipose homeostasis. Semin Cell Dev Biol. 1999; 10:11–18. PMID: 10355024.
Article
13. Mul JD, van Boxtel R, Bergen DJ, Brans MA, Brakkee JH, Toonen PW, et al. Melanocortin receptor 4 deficiency affects body weight regulation, grooming behavior, and substrate preference in the rat. Obesity (Silver Spring). 2012; 20:612–621. PMID: 21527895.
Article
14. Terroni PL, Anthony FW, Hanson MA, Cagampang FR. Expression of agouti-related peptide, neuropeptide Y, pro-opiomelanocortin and the leptin receptor isoforms in fetal mouse brain from pregnant dams on a protein-restricted diet. Brain Res Mol Brain Res. 2005; 140:111–115. PMID: 16099070.
Article
15. Plagemann A, Waas T, Harder T, Rittel F, Ziska T, Rohde W. Hypothalamic neuropeptide Y levels in weaning offspring of low-protein malnourished mother rats. Neuropeptides. 2000; 34:1–6. PMID: 10688961.
Article
16. Cordero P, Li J, Nguyen V, Pombo J, Maicas N, Novelli M, et al. Developmental programming of obesity and liver metabolism by maternal perinatal nutrition involves the melanocortin system. Nutrients. 2017; 9:E1041. PMID: 28930194.
Article
17. Proulx K, Richard D, Walker CD. Leptin regulates appetite-related neuropeptides in the hypothalamus of developing rats without affecting food intake. Endocrinology. 2002; 143:4683–4692. PMID: 12446596.
Article
18. Bouret SG, Draper SJ, Simerly RB. Trophic action of leptin on hypothalamic neurons that regulate feeding. Science. 2004; 304:108–110. PMID: 15064420.
Article
19. Vickers MH. Early life nutrition, epigenetics and programming of later life disease. Nutrients. 2014; 6:2165–2178. PMID: 24892374.
Article
20. Martínez JA, Cordero P, Campión J, Milagro FI. Interplay of early-life nutritional programming on obesity, inflammation and epigenetic outcomes. Proc Nutr Soc. 2012; 71:276–283. PMID: 22390978.
Article
21. Stevens A, Begum G, White A. Epigenetic changes in the hypothalamic pro-opiomelanocortin gene: a mechanism linking maternal undernutrition to obesity in the offspring? Eur J Pharmacol. 2011; 660:194–201. PMID: 21211530.
Article
22. Stevens A, Begum G, Cook A, Connor K, Rumball C, Oliver M, et al. Epigenetic changes in the hypothalamic proopiomelanocortin and glucocorticoid receptor genes in the ovine fetus after periconceptional undernutrition. Endocrinology. 2010; 151:3652–3664. PMID: 20573728.
Article
23. Crujeiras AB, Campion J, Díaz-Lagares A, Milagro FI, Goyenechea E, Abete I, et al. Association of weight regain with specific methylation levels in the NPY and POMC promoters in leukocytes of obese men: a translational study. Regul Pept. 2013; 186:1–6. PMID: 23831408.
Article
24. Widiker S, Karst S, Wagener A, Brockmann GA. High-fat diet leads to a decreased methylation of the Mc4r gene in the obese BFMI and the lean B6 mouse lines. J Appl Genet. 2010; 51:193–197. PMID: 20453306.
25. Tabachnik T, Kisliouk T, Marco A, Meiri N, Weller A. Thyroid hormone-dependent epigenetic regulation of melanocortin 4 receptor levels in female offspring of obese rats. Endocrinology. 2017; 158:842–851. PMID: 28324105.
Article
26. Yoo JY, Lee S, Lee HA, Park H, Park YJ, Ha EH, et al. Can proopiomelanocortin methylation be used as an early predictor of metabolic syndrome? Diabetes Care. 2014; 37:734–739. PMID: 24222450.
Article
27. Kwon EJ, You YA, Park B, Ha EH, Kim HS, Park H, et al. Association between the DNA methylations of POMC, MC4R, and HNF4A and metabolic profiles in the blood of children aged 7–9 years. BMC Pediatr. 2018; 18:121. PMID: 29598821.
Article
28. Plagemann A, Harder T, Brunn M, Harder A, Roepke K, Wittrock-Staar M, et al. Hypothalamic proopiomelanocortin promoter methylation becomes altered by early overfeeding: an epigenetic model of obesity and the metabolic syndrome. J Physiol. 2009; 587:4963–4976. PMID: 19723777.
Article
29. Nowacka-Woszuk J, Madeja ZE, Chmurzynska A. Prenatal caloric restriction alters lipid metabolism but not hepatic Fasn gene expression and methylation profiles in rats. BMC Genet. 2017; 18:78. PMID: 28810876.
Article
30. Desai M, Gayle D, Babu J, Ross MG. Programmed obesity in intrauterine growth-restricted newborns: modulation by newborn nutrition. Am J Physiol Regul Integr Comp Physiol. 2005; 288:R91–6. PMID: 15297266.
Article
31. Vickers MH, Breier BH, Cutfield WS, Hofman PL, Gluckman PD. Fetal origins of hyperphagia, obesity, and hypertension and postnatal amplification by hypercaloric nutrition. Am J Physiol Endocrinol Metab. 2000; 279:E83–7. PMID: 10893326.
Article
32. Lucas A, Baker BA, Desai M, Hales CN. Nutrition in pregnant or lactating rats programs lipid metabolism in the offspring. Br J Nutr. 1996; 76:605–612. PMID: 8942366.
Article
33. Coupé B, Amarger V, Grit I, Benani A, Parnet P. Nutritional programming affects hypothalamic organization and early response to leptin. Endocrinology. 2010; 151:702–713. PMID: 20016030.
34. Begum G, Stevens A, Smith EB, Connor K, Challis JR, Bloomfield F, et al. Epigenetic changes in fetal hypothalamic energy regulating pathways are associated with maternal undernutrition and twinning. FASEB J. 2012; 26:1694–1703. PMID: 22223754.
Article
35. Kuehnen P, Mischke M, Wiegand S, Sers C, Horsthemke B, Lau S, et al. An Alu element-associated hypermethylation variant of the POMC gene is associated with childhood obesity. PLoS Genet. 2012; 8:e1002543. PMID: 22438814.
Article
36. Tung YC, Yeo GS. Central melanocortin signaling regulates cholesterol. Nat Neurosci. 2010; 13:779–780. PMID: 20581809.
Article
37. Queiroz EM, Cândido AP, Castro IM, Bastos AQ, Machado-Coelho GL, Freitas RN. IGF2, LEPR, POMC, PPARG, and PPARGC1 gene variants are associated with obesity-related risk phenotypes in Brazilian children and adolescents. Braz J Med Biol Res. 2015; 48:595–602. PMID: 25923461.
Article
38. Zheng J, Xiao X, Zhang Q, Yu M, Xu J, Wang Z, et al. Maternal and post-weaning high-fat, high-sucrose diet modulates glucose homeostasis and hypothalamic POMC promoter methylation in mouse offspring. Metab Brain Dis. 2015; 30:1129–1137. PMID: 25936720.
Article
Full Text Links
  • OGS
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