We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Nutriepigenomics and malnutrition

    Arnaud de Luca

    Research Center, Sainte-Justine University Hospital Center, Montreal, Quebec H3T 1C5, Canada

    INSERM, U 1069, F-37044 Tours, France

    ,
    Regis Hankard

    INSERM, U 1069, F-37044 Tours, France

    François Rabelais University, F-37000 Tours, France

    ,
    Jean-Michel Borys

    EPODE International Network, F-75017 Paris, France

    ,
    Daniel Sinnett

    Research Center, Sainte-Justine University Hospital Center, Montreal, Quebec H3T 1C5, Canada

    Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, Quebec H3T 1J4, Canada

    ,
    Valérie Marcil

    Research Center, Sainte-Justine University Hospital Center, Montreal, Quebec H3T 1C5, Canada

    Department of Nutrition, Faculty of Medicine, University of Montréal, Montreal, Quebec H3T 1J4, Canada

    &
    Emile Levy

    *Author for correspondence:

    E-mail Address: emile.levy@recherche-ste-justine.qc.ca

    Research Center, Sainte-Justine University Hospital Center, Montreal, Quebec H3T 1C5, Canada

    EPODE International Network, F-75017 Paris, France

    Department of Nutrition, Faculty of Medicine, University of Montréal, Montreal, Quebec H3T 1J4, Canada

    Published Online:https://doi.org/10.2217/epi-2016-0168

    Epigenetics is defined as the modulation of gene expression without changes to the underlying DNA sequence. Epigenetic alterations, as a consequence of in utero malnutrition, may play a role in susceptibility to develop adulthood diseases and inheritance. However, the mechanistic link between epigenetic modifications and abnormalities in nutrition remains elusive. This review provides an update on the association of suboptimal nutritional environment and the high propensity to produce adult-onset chronic illnesses with a particular focus on modifications in genome functions that occur without alterations to the DNA sequence. We will mention the drivers of the phenotype and pattern of epigenetic markers set down during the reprogramming along with novel preventative and therapeutic strategies. New knowledge of epigenetic alterations is opening a gate toward personalized medicine.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1 Burdge GC, Hoile SP, Lillycrop KA. Epigenetics: are there implications for personalised nutrition? Curr. Opin. Clin. Nutr. Metab. Care 15(5), 442–447 (2012).
    • 2 Ravelli AC, Van Der Meulen JH, Michels RP et al. Glucose tolerance in adults after prenatal exposure to famine. Lancet 351(9097), 173–177 (1998).
    • 3 Ravelli AC, Van Der Meulen JH, Osmond C, Barker DJ, Bleker OP. Obesity at the age of 50 y in men and women exposed to famine prenatally. Am. J. Clin. Nutr. 70(5), 811–816 (1999).
    • 4 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. 72(5), 1101–1106 (2000).
    • 5 Heijmans BT, Tobi EW, Stein AD et al. Persistent epigenetic differences associated with prenatal exposure to famine in humans. Proc. Natl Acad. Sci. USA 105(44), 17046–17049 (2008).
    • 6 Jiménez-Chillarón JC, Díaz R, Martínez D et al. The role of nutrition on epigenetic modifications and their implications on health. Biochimie 94(11), 2242–2263 (2012).
    • 7 Lillycrop KA, Burdge GC. Epigenetic mechanisms linking early nutrition to long term health. Best Pract. Res. Clin. Endocrinol. Metab. 26(5), 667–676 (2012).
    • 8 Cheung L, Gustavsson C, Norstedt G, Tollet-Egnell P. Sex-different and growth hormone-regulated expression of microRNA in rat liver. BMC Mol. Biol. 10, 13 (2009).
    • 9 De Roos B. Personalised nutrition: ready for practice? Proc. Nutr. Soc. 72(1), 48–52 (2013).
    • 10 Faulk C, Dolinoy DC. Timing is everything: the when and how of environmentally induced changes in the epigenome of animals. Epigenetics 6(7), 791–797 (2011).
    • 11 Fenech M, El-Sohemy A, Cahill L et al. Nutrigenetics and nutrigenomics: viewpoints on the current status and applications in nutrition research and practice. J. Nutrigenet. Nutrigenomics 4(2), 69–89 (2011).
    • 12 Maunakea AK, Nagarajan RP, Bilenky M et al. Conserved role of intragenic DNA methylation in regulating alternative promoters. Nature 466(7303), 253–257 (2010).
    • 13 Reik W, Dean W, Walter J. Epigenetic reprogramming in mammalian development. Science 293(5532), 1089–1093 (2001).
    • 14 Baubec T, Colombo DF, Wirbelauer C et al. Genomic profiling of DNA methyltransferases reveals a role for DNMT3B in genic methylation. Nature 520(7546), 243–247 (2015).
    • 15 Li E, Beard C, Jaenisch R. Role for DNA methylation in genomic imprinting. Nature 366(6453), 362–365 (1993).
    • 16 Wong CCY, Caspi A, Williams B, Houts R, Craig IW, Mill J. A longitudinal twin study of skewed X chromosome-inactivation. PLoS ONE 6(3), e17873 (2011).
    • 17 Ferguson-Smith AC. Genomic imprinting: the emergence of an epigenetic paradigm. Nat. Rev. Genet. 12(8), 565–575 (2011).
    • 18 Kriaucionis S, Heintz N. The nuclear DNA base 5-hydroxymethylcytosine is present in Purkinje neurons and the brain. Science 324(5929), 929–930 (2009).
    • 19 Ramiro AR, Barreto VM. Activation-induced cytidine deaminase and active cytidine demethylation. Trends Biochem. Sci. 40(3), 172–181 (2015).
    • 20 Bochtler M, Kolano A, Xu GL. DNA demethylation pathways: additional players and regulators. Bioessays 39(1), 1–13 (2017).
    • 21 Turner BM. Histone acetylation and an epigenetic code. Bioessays 22(9), 836–845 (2000).
    • 22 Raychaudhuri N, Raychaudhuri S, Thamotharan M, Devaskar SU. Histone code modifications repress glucose transporter 4 expression in the intrauterine growth-restricted offspring. J. Biol. Chem. 283(20), 13611–13626 (2008).
    • 23 Zhao J, Sun BK, Erwin JA, Song JJ, Lee JT. Polycomb proteins targeted by a short repeat RNA to the mouse X chromosome. Science 322(5902), 750–756 (2008).
    • 24 Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 9(6), 654–659 (2007).
    • 25 Vickers KC, Palmisano BT, Shoucri BM, Shamburek RD, Remaley AT. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat. Cell Biol. 13(4), 423–433 (2011). • Suggests that miRNAs can be transported in plasma via high-density lipoprotein and that high-density lipoprotein participate to intercellular communication.
    • 26 Lu H, Buchan RJ, Cook SA. MicroRNA-223 regulates Glut4 expression and cardiomyocyte glucose metabolism. Cardiovasc. Res. 86(3), 410–420 (2010).
    • 27 Vickers KC, Landstreet SR, Levin MG et al. MicroRNA-223 coordinates cholesterol homeostasis. Proc. Natl Acad. Sci. USA 111(40), 14518–14523 (2014).
    • 28 Cedar H, Bergman Y. Linking DNA methylation and histone modification: patterns and paradigms. Nat. Rev. Genet. 10(5), 295–304 (2009).
    • 29 Viré E, Brenner C, Deplus R et al. The Polycomb group protein EZH2 directly controls DNA methylation. Nature 439(7078), 871–874 (2006).
    • 30 Zardo G, Ciolfi A, Vian L et al. Polycombs and microRNA-223 regulate human granulopoiesis by transcriptional control of target gene expression. Blood 119(17), 4034–4046 (2012).
    • 31 Sandovici I, Smith NH, Nitert MD et al. Maternal diet and aging alter the epigenetic control of a promoter–enhancer interaction at the Hnf4a gene in rat pancreatic islets. Proc. Natl Acad. Sci. USA 108(13), 5449–5454 (2011).
    • 32 Stadler MB, Murr R, Burger L et al. DNA-binding factors shape the mouse methylome at distal regulatory regions. Nature 480(7378), 490–495 (2011).
    • 33 Anderson OS, Sant KE, Dolinoy DC. Nutrition and epigenetics: an interplay of dietary methyl donors, one-carbon metabolism and DNA methylation. J. Nutr. Biochem. 23(8), 853–859 (2012).
    • 34 Wu SC, Zhang Y. Active DNA demethylation: many roads lead to Rome. Nat. Rev. Mol. Cell Biol. 11(9), 607–620 (2010).
    • 35 Feng S, Jacobsen SE, Reik W. Epigenetic reprogramming in plant and animal development. Science 330(6004), 622–627 (2010).
    • 36 Balhorn R. The protamine family of sperm nuclear proteins. Genome Biol. 8(9), 227 (2007).
    • 37 Kota SK, Feil R. Epigenetic transitions in germ cell development and meiosis. Dev. Cell 19(5), 675–686 (2010).
    • 38 Hanel ML, Wevrick R. Establishment and maintenance of DNA methylation patterns in mouse Ndn: implications for maintenance of imprinting in target genes of the imprinting center. Mol. Cell. Biol. 21(7), 2384–2392 (2001).
    • 39 Bollati V, Baccarelli A. Environmental epigenetics. Heredity 105(1), 105–112 (2010).
    • 40 Yoder JA, Soman NS, Verdine GL, Bestor TH. DNA (cytosine-5)-methyltransferases in mouse cells and tissues. Studies with a mechanism-based probe. J. Mol. Biol. 270(3), 385–395 (1997).
    • 41 Santos F, Hendrich B, Reik W, Dean W. Dynamic reprogramming of DNA methylation in the early mouse embryo. Dev. Biol. 241(1), 172–182 (2002).
    • 42 Fraga MF, Ballestar E, Paz MF et al. Epigenetic differences arise during the lifetime of monozygotic twins. Proc. Natl Acad. Sci. USA 102(30), 10604–10609 (2005).
    • 43 Wong CCY, Caspi A, Williams B et al. A longitudinal study of epigenetic variation in twins. Epigenetics 5(6), 516–526 (2010).
    • 44 Loke YJ, Novakovic B, Ollikainen M et al. The Peri/postnatal epigenetic twins study (PETS). Twin Res. Hum. Genet. 16(1), 13–20 (2013).
    • 45 Gordon L, Joo JE, Powell JE et al. Neonatal DNA methylation profile in human twins is specified by a complex interplay between intrauterine environmental and genetic factors, subject to tissue-specific influence. Genome Res. 22(8), 1395–1406 (2012).
    • 46 Issa JP. Aging and epigenetic drift: a vicious cycle. J. Clin. Invest. 124(1), 24–29 (2014).
    • 47 Boyd-Kirkup JD, Green CD, Wu G, Wang D, Han JDJ. Epigenomics and the regulation of aging. Epigenomics 5(2), 205–227 (2013).
    • 48 Murgatroyd C, Wu Y, Bockmühl Y, Spengler D. The Janus face of DNA methylation in aging. Aging 2(2), 107–110 (2010).
    • 49 Vaquero A, Reinberg D. Calorie restriction and the exercise of chromatin. Genes Dev. 23(16), 1849–1869 (2009).
    • 50 Godfrey KM, Barker DJ. Fetal programming and adult health. Public Health Nutr. 4(2B), 611–624 (2001).
    • 51 Lillycrop KA, Burdge GC. Epigenetic changes in early life and future risk of obesity. Int. J. Obes. (Lond.) 35(1), 72–83 (2011).
    • 52 Godfrey KM, Reynolds RM, Prescott SL et al. Influence of maternal obesity on the long-term health of offspring. Lancet Diabetes Endocrinol. 5(1), 53–64 (2017).
    • 53 Huang C, Li Z, Wang M, Martorell R. Early life exposure to the 1959–1961 Chinese famine has long-term health consequences. J. Nutr. 140(10), 1874–1878 (2010).
    • 54 Zheng X, Wang Y, Ren W et al. Risk of metabolic syndrome in adults exposed to the great Chinese famine during the fetal life and early childhood. Eur. J. Clin. Nutr. 66(2), 231–236 (2012).
    • 55 Li Y, He Y, Qi L et al. Exposure to the Chinese famine in early life and the risk of hyperglycemia and Type 2 diabetes in adulthood. Diabetes 59(10), 2400–2406 (2010).
    • 56 Hult M, Tornhammar P, Ueda P et al. Hypertension, diabetes and overweight: looming legacies of the Biafran famine. PLoS ONE 5(10), e13582 (2010).
    • 57 Stein AD, Zybert PA, Van Der Pal-De Bruin K, Lumey LH. Exposure to famine during gestation, size at birth, and blood pressure at age 59 y: evidence from the Dutch famine. Eur. J. Epidemiol. 21(10), 759–765 (2006).
    • 58 Painter RC, Roseboom TJ, Bleker OP. Prenatal exposure to the Dutch famine and disease in later life: an overview. Reprod. Toxicol. 20(3), 345–352 (2005).
    • 59 Lumey LH. Decreased birthweights in infants after maternal in utero exposure to the Dutch famine of 1944–1945. Paediatr. Perinat. Epidemiol. 6(2), 240–253 (1992).
    • 60 Kaati G, Bygren LO, Edvinsson S. Cardiovascular and diabetes mortality determined by nutrition during parents’ and grandparents’ slow growth period. Eur. J. Hum. Genet. 10(11), 682–688 (2002).
    • 61 Bygren LO, Tinghög P, Carstensen J et al. Change in paternal grandmothers’ early food supply influenced cardiovascular mortality of the female grandchildren. BMC Genet. 15, 12 (2014). •• Reports the transgenerational impact of nutritional status in humans with a putative epigenetic mechanism.
    • 62 Kamakura M. Royalactin induces queen differentiation in honeybees. Nature 473(7348), 478–483 (2011).
    • 63 Kucharski R, Maleszka J, Foret S, Maleszka R. Nutritional control of reproductive status in honeybees via DNA methylation. Science 319(5871), 1827–1830 (2008).
    • 64 Barker DJ. The developmental origins of chronic adult disease. Acta Paediatr. Suppl. 93(446), 26–33 (2004).
    • 65 Radford EJ, Ito M, Shi H et al. In utero effects. In utero undernourishment perturbs the adult sperm methylome and intergenerational metabolism. Science 345(6198), 1255903 (2014).
    • 66 Gluckman PD, Hanson MA, Spencer HG. Predictive adaptive responses and human evolution. Trends Ecol. Evol. 20(10), 527–533 (2005).
    • 67 Tobi EW, Lumey LH, Talens RP et al. DNA methylation differences after exposure to prenatal famine are common and timing- and sex-specific. Hum. Mol. Genet. 18(21), 4046–4053 (2009). •• Shows time- and sex-depending epigenetic alterations in humans exposed in utero to malnutrition during the Dutch famine.
    • 68 Tobi EW, Heijmans BT, Kremer D et al. DNA methylation of IGF2, GNASAS, INSIGF and LEP and being born small for gestational age. Epigenetics 6(2), 171–176 (2011).
    • 69 Tosh DN, Fu Q, Callaway CW et al. Epigenetics of programmed obesity: alteration in IUGR rat hepatic IGF1 mRNA expression and histone structure in rapid vs. delayed postnatal catch-up growth. Am. J. Physiol. Gastrointest. Liver Physiol. 299(5), G1023–G1029 (2010).
    • 70 Park JH, Stoffers DA, Nicholls RD, Simmons RA. Development of Type 2 diabetes following intrauterine growth retardation in rats is associated with progressive epigenetic silencing of Pdx1. J. Clin. Invest. 118(6), 2316–2324 (2008).
    • 71 Alejandro EU, Gregg B, Wallen T et al. Maternal diet-induced microRNAs and mTOR underlie β cell dysfunction in offspring. J. Clin. Invest. 124(10), 4395–4410 (2014).
    • 72 Jousse C, Parry L, Lambert-Langlais S et al. Perinatal undernutrition affects the methylation and expression of the leptin gene in adults: implication for the understanding of metabolic syndrome. FASEB J. 25(9), 3271–3278 (2011).
    • 73 Krechowec SO, Vickers M, Gertler A, Breier BH. Prenatal influences on leptin sensitivity and susceptibility to diet-induced obesity. J. Endocrinol. 189(2), 355–363 (2006).
    • 74 Gupta RK, Gao N, Gorski RK et al. Expansion of adult β-cell mass in response to increased metabolic demand is dependent on HNF-4α. Genes Dev. 21(7), 756–769 (2007).
    • 75 Maclennan NK, James SJ, Melnyk S et al. Uteroplacental insufficiency alters DNA methylation, one-carbon metabolism, and histone acetylation in IUGR rats. Physiol. Genomics 18(1), 43–50 (2004).
    • 76 Fu Q, Mcknight RA, Yu X, Callaway CW, Lane RH. Growth retardation alters the epigenetic characteristics of hepatic dual specificity phosphatase 5. FASEB J. 20(12), 2127–2129 (2006).
    • 77 Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman BM, Puigserver P. Nutrient control of glucose homeostasis through a complex of PGC-1alpha and SIRT1. Nature 434(7029), 113–118 (2005).
    • 78 Amat R, Solanes G, Giralt M, Villarroya F. SIRT1 is involved in glucocorticoid-mediated control of uncoupling protein-3 gene transcription. J. Biol. Chem. 282(47), 34066–34076 (2007).
    • 79 Gat-Yablonski G, Pando R, Phillip M. Nutritional catch-up growth. World Rev. Nutr. Diet. 106, 83–89 (2013).
    • 80 Pando R, Shtaif B, Phillip M, Gat-Yablonski G. A serum component mediates food restriction-induced growth attenuation. Endocrinology 155(3), 932–940 (2014).
    • 81 Pando R, Even-Zohar N, Shtaif B et al. MicroRNAs in the growth plate are responsive to nutritional cues: association between miR-140 and SIRT1. J. Nutr. Biochem. 23(11), 1474–1481 (2012).
    • 82 Gluckman PD, Lillycrop KA, Vickers MH et al. Metabolic plasticity during mammalian development is directionally dependent on early nutritional status. Proc. Natl Acad. Sci. USA 104(31), 12796–12800 (2007).
    • 83 Lillycrop KA, Slater-Jefferies JL, Hanson MA, Godfrey KM, Jackson AA, Burdge GC. Induction of altered epigenetic regulation of the hepatic glucocorticoid receptor in the offspring of rats fed a protein-restricted diet during pregnancy suggests that reduced DNA methyltransferase-1 expression is involved in impaired DNA methylation and changes in histone modifications. Br. J. Nutr. 97(6), 1064–1073 (2007).
    • 84 Burns SP, Desai M, Cohen RD et al. Gluconeogenesis, glucose handling, and structural changes in livers of the adult offspring of rats partially deprived of protein during pregnancy and lactation. J. Clin. Invest. 100(7), 1768–1774 (1997).
    • 85 Burdge GC, Slater-Jefferies J, Torrens C, Phillips ES, Hanson MA, Lillycrop KA. Dietary protein restriction of pregnant rats in the F0 generation induces altered methylation of hepatic gene promoters in the adult male offspring in the F1 and F2 generations. Br. J. Nutr. 97(3), 435–439 (2007).
    • 86 Whorwood CB, Firth KM, Budge H, Symonds ME. Maternal undernutrition during early to midgestation programs tissue-specific alterations in the expression of the glucocorticoid receptor, 11beta-hydroxysteroid dehydrogenase isoforms, and type 1 angiotensin ii receptor in neonatal sheep. Endocrinology 142(7), 2854–2864 (2001).
    • 87 Zheng S, Rollet M, Pan YX. Maternal protein restriction during pregnancy induces CCAAT/enhancer-binding protein (C/EBPβ) expression through the regulation of histone modification at its promoter region in female offspring rat skeletal muscle. Epigenetics 6(2), 161–170 (2011).
    • 88 Zhou D, Pan YX. Gestational low protein diet selectively induces the amino acid response pathway target genes in the liver of offspring rats through transcription factor binding and histone modifications. Biochim. Biophys. Acta 1809(10), 549–556 (2011).
    • 89 Nebendahl C, Görs S, Albrecht E et al. Early postnatal feed restriction reduces liver connective tissue levels and affects H3K9 acetylation state of regulated genes associated with protein metabolism in low birth weight pigs. J. Nutr. Biochem. 29, 41–55 (2016).
    • 90 Pinto G, Shtaif B, Phillip M, Gat-Yablonski G. Growth attenuation is associated with histone deacetylase 10-induced autophagy in the liver. J. Nutr. Biochem. 27, 171–180 (2016).
    • 91 Gluckman PD, Hanson MA, Cooper C, Thornburg KL. Effect of in utero and early-life conditions on adult health and disease. N. Engl. J. Med. 359(1), 61–73 (2008).
    • 92 Slater-Jefferies JL, Lillycrop KA, Townsend PA et al. Feeding a protein-restricted diet during pregnancy induces altered epigenetic regulation of peroxisomal proliferator-activated receptor-α in the heart of the offspring. J. Dev. Orig. Health Dis. 2(4), 250–255 (2011).
    • 93 Carone BR, Fauquier L, Habib N et al. Paternally-induced transgenerational environmental reprogramming of metabolic gene expression in mammals. Cell 143(7), 1084–1096 (2010).
    • 94 Fuemmeler BF, Agurs-Collins T, Mcclernon FJ et al. Genes implicated in serotonergic and dopaminergic functioning predict BMI categories. Obesity 16(2), 348–355 (2008).
    • 95 Ozanne SE, Sandovici I, Constância M. Maternal diet, aging and diabetes meet at a chromatin loop. Aging 3(5), 548–554 (2011).
    • 96 He A, Zhu L, Gupta N, Chang Y, Fang F. Overexpression of micro ribonucleic acid 29, highly up-regulated in diabetic rats, leads to insulin resistance in 3T3-L1 adipocytes. Mol. Endocrinol. 21(11), 2785–2794 (2007).
    • 97 Xie X, Lin T, Zhang M et al. IUGR with infantile overnutrition programs an insulin-resistant phenotype through DNA methylation of peroxisome proliferator-activated receptor-γ coactivator-1α in rats. Pediatr. Res. 77(5), 625–632 (2015).
    • 98 Wang ND, Finegold MJ, Bradley A et al. Impaired energy homeostasis in C/EBP alpha knockout mice. Science 269(5227), 1108–1112 (1995).
    • 99 Wu KJ, Wilson DR, Shih C, Darlington GJ. The transcription factor HNF1 acts with C/EBP alpha to synergistically activate the human albumin promoter through a novel domain. J. Biol. Chem. 269(2), 1177–1182 (1994).
    • 100 Van Straten EME, Bloks VW, Huijkman NCA et al. The liver X-receptor gene promoter is hypermethylated in a mouse model of prenatal protein restriction. Am. J. Physiol. Regul. Integr. Comp. Physiol. 298(2), R275–R282 (2010).
    • 101 Ou Z, Wada T, Gramignoli R et al. MicroRNA hsa-miR-613 targets the human LXRα gene and mediates a feedback loop of LXRα autoregulation. Mol. Endocrinol. 25(4), 584–596 (2011).
    • 102 Martínez D, Pentinat T, Ribó S et al. In utero undernutrition in male mice programs liver lipid metabolism in the second-generation offspring involving altered Lxra DNA methylation. Cell. Metab. 19(6), 941–951 (2014).
    • 103 Lan F, Cacicedo JM, Ruderman N, Ido Y. SIRT1 modulation of the acetylation status, cytosolic localization, and activity of LKB1. Possible role in AMP-activated protein kinase activation. J. Biol. Chem. 283(41), 27628–27635 (2008).
    • 104 Li X, Zhang S, Blander G, Tse JG, Krieger M, Guarente L. SIRT1 deacetylates and positively regulates the nuclear receptor LXR. Mol. Cell 28(1), 91–106 (2007).
    • 105 Repa JJ, Mangelsdorf DJ. The role of orphan nuclear receptors in the regulation of cholesterol homeostasis. Annu. Rev. Cell Dev. Biol. 16, 459–481 (2000).
    • 106 Sohi G, Marchand K, Revesz A, Arany E, Hardy DB. Maternal protein restriction elevates cholesterol in adult rat offspring due to repressive changes in histone modifications at the cholesterol 7alpha-hydroxylase promoter. Mol. Endocrinol. 25(5), 785–798 (2011).
    • 107 Cong R, Jia Y, Li R et al. Maternal low-protein diet causes epigenetic deregulation of HMGCR and CYP7α1 in the liver of weaning piglets. J. Nutr. Biochem. 23(12), 1647–1654 (2012).
    • 108 Karolina DS, Tavintharan S, Armugam A et al. Circulating miRNA profiles in patients with metabolic syndrome. J. Clin. Endocrinol. Metab. 97(12), E2271–E2276 (2012). • This clinical study suggests that miRNA profiles could be specific of different items of metabolic diseases, contributing to identify potential biomarkers.
    • 109 Baselga-Escudero L, Blade C, Ribas-Latre A et al. Chronic supplementation of proanthocyanidins reduces postprandial lipemia and liver miR-33a and miR-122 levels in a dose-dependent manner in healthy rats. J. Nutr. Biochem. 25(2), 151–156 (2014).
    • 110 Baselga-Escudero L, Blade C, Ribas-Latre A et al. Resveratrol and EGCG bind directly and distinctively to miR-33a and miR-122 and modulate divergently their levels in hepatic cells. Nucleic Acids Res. 42(2), 882–892 (2014).
    • 111 Joven J, Espinel E, Rull A et al. Plant-derived polyphenols regulate expression of miRNA paralogs miR-103/107 and miR-122 and prevent diet-induced fatty liver disease in hyperlipidemic mice. Biochim. Biophys. Acta 1820(7), 894–899 (2012).
    • 112 Yang YM, Seo SY, Kim TH, Kim SG. Decrease of microRNA-122 causes hepatic insulin resistance by inducing protein tyrosine phosphatase 1B, which is reversed by licorice flavonoid. Hepatology 56(6), 2209–2220 (2012). • Shows that the reduction of hepatic insulin resistance by licorice flavonoid (polyphenol) may depend on its miR-122 regulation, as a potential therapeutics.
    • 113 Ohno M, Shibata C, Kishikawa T et al. The flavonoid apigenin improves glucose tolerance through inhibition of microRNA maturation in miRNA103 transgenic mice. Sci. Rep. 3, 2553 (2013).
    • 114 Milenkovic D, Deval C, Gouranton E et al. Modulation of miRNA expression by dietary polyphenols in apoE deficient mice: a new mechanism of the action of polyphenols. PLoS ONE 7(1), e29837 (2012).
    • 115 Milenkovic D, Jude B, Morand C. miRNA as molecular target of polyphenols underlying their biological effects. Free Radic. Biol. Med. 64, 40–51 (2013).
    • 116 Wang D, Xia M, Yan X et al. Gut microbiota metabolism of anthocyanin promotes reverse cholesterol transport in mice via repressing miRNA-10b. Circ. Res. 111(8), 967–981 (2012).
    • 117 Price NL, Gomes AP, Ling AJY et al. SIRT1 is required for AMPK activation and the beneficial effects of resveratrol on mitochondrial function. Cell. Metab. 15(5), 675–690 (2012).
    • 118 Tarry-Adkins JL, Martin-Gronert MS, Fernandez-Twinn DS et al. Poor maternal nutrition followed by accelerated postnatal growth leads to alterations in DNA damage and repair, oxidative and nitrosative stress, and oxidative defense capacity in rat heart. FASEB J. 27(1), 379–390 (2013).
    • 119 Hayes P, Knaus UG. Balancing reactive oxygen species in the epigenome: NADPH oxidases as target and perpetrator. Antioxid. Redox Signal. 18(15), 1937–1945 (2013).
    • 120 Valinluck V, Tsai HH, Rogstad DK, Burdzy A, Bird A, Sowers LC. Oxidative damage to methyl-CpG sequences inhibits the binding of the methyl-CpG binding domain (MBD) of methyl-CpG binding protein 2 (MeCP2). Nucleic acids Res. 32(14), 4100–4108 (2004).
    • 121 Mckay JA, Waltham KJ, Williams EA, Mathers JC. Folate depletion during pregnancy and lactation reduces genomic DNA methylation in murine adult offspring. Genes Nutr. 6(2), 189–196 (2011).
    • 122 Tian X, Diaz FJ. Acute dietary zinc deficiency before conception compromises oocyte epigenetic programming and disrupts embryonic development. Dev. Biol. 376(1), 51–61 (2013).
    • 123 Waterland RA, Kellermayer R, Laritsky E et al. Season of conception in rural gambia affects DNA methylation at putative human metastable epialleles. PLoS Genet. 6(12), e1001252 (2010).
    • 124 Dominguez-Salas P, Moore SE, Cole D et al. DNA methylation potential: dietary intake and blood concentrations of one-carbon metabolites and cofactors in rural African women. Am. J. Clin. Nutr. 97(6), 1217–1227 (2013).
    • 125 Dominguez-Salas P, Moore SE, Baker MS et al. Maternal nutrition at conception modulates DNA methylation of human metastable epialleles. Nat. Commun. 5, 3746 (2014). • Suggests that maternal nutrition, and particularly micronutrients involved in one-carbon metabolism, has an impact on epigenome of the offspring in humans.
    • 126 Steegers-Theunissen RP, Obermann-Borst SA, Kremer D et al. Periconceptional maternal folic acid use of 400 microg per day is related to increased methylation of the IGF2 gene in the very young child. PLoS ONE 4(11), e7845 (2009).
    • 127 Lambrot R, Xu C, Saint-Phar S et al. Low paternal dietary folate alters the mouse sperm epigenome and is associated with negative pregnancy outcomes. Nat. Commun. 4, 2889 (2013).
    • 128 Burdge GC, Lillycrop KA, Jackson AA, Gluckman PD, Hanson MA. The nature of the growth pattern and of the metabolic response to fasting in the rat are dependent upon the dietary protein and folic acid intakes of their pregnant dams and post-weaning fat consumption. Br. J. Nutr. 99(3), 540–549 (2008).
    • 129 Lillycrop KA, Phillips ES, Jackson AA, Hanson MA, Burdge GC. Dietary protein restriction of pregnant rats induces and folic acid supplementation prevents epigenetic modification of hepatic gene expression in the offspring. J. Nutr. 135(6), 1382–1386 (2005).
    • 130 Burdge GC, Lillycrop KA, Phillips ES, Slater-Jefferies JL, Jackson AA, Hanson MA. Folic acid supplementation during the juvenile-pubertal period in rats modifies the phenotype and epigenotype induced by prenatal nutrition. J. Nutr. 139(6), 1054–1060 (2009).
    • 131 Zhang L, Hou D, Chen X et al. Exogenous plant MIR168a specifically targets mammalian LDLRAP1: evidence of cross-kingdom regulation by microRNA. Cell Res. 22(1), 107–126 (2012).
    • 132 Krützfeldt J, Rajewsky N, Braich R et al. Silencing of microRNAs in vivo with ‘antagomirs’. Nature 438(7068), 685–689 (2005).
    • 133 Shibata C, Kishikawa T, Otsuka M et al. Inhibition of microRNA122 decreases SREBP1 expression by modulating suppressor of cytokine signaling 3 expression. Biochem. Biophys. Res. Commun. 438(1), 230–235 (2013).
    • 134 Elmén J, Lindow M, Schütz S et al. LNA-mediated microRNA silencing in non-human primates. Nature 452(7189), 896–899 (2008).
    • 135 Wen J, Friedman JR. miR-122 regulates hepatic lipid metabolism and tumor suppression. J. Clin. Invest. 122(8), 2773–2776 (2012).
    • 136 Iliopoulos D, Drosatos K, Hiyama Y, Goldberg IJ, Zannis VI. MicroRNA-370 controls the expression of microRNA-122 and Cpt1alpha and affects lipid metabolism. J. Lipid Res. 51(6), 1513–1523 (2010).
    • 137 Rayner KJ, Esau CC, Hussain FN et al. Inhibition of miR-33a/b in non-human primates raises plasma HDL and lowers VLDL triglycerides. Nature 478(7369), 404–407 (2011). •• This nonhuman primate study reports the pharmacological inhibition of miR-33 as a potential therapeutics in dyslipidemia.
    • 138 Rottiers V, Obad S, Petri A et al. Pharmacological inhibition of a microRNA family in nonhuman primates by a seed-targeting 8-mer antimiR. Sci. Transl. Med. 5(212), 212ra162 (2013).
    • 139 Horie T, Nishino T, Baba O et al. MicroRNA-33 regulates sterol regulatory element-binding protein 1 expression in mice. Nat. Commun. 4, 2883 (2013).
    • 140 Ramírez CM, Goedeke L, Rotllan N et al. MicroRNA 33 regulates glucose metabolism. Mol. Cell. Biol. 33(15), 2891–2902 (2013).
    • 141 Wilfred BR, Wang WX, Nelson PT. Energizing miRNA research: a review of the role of miRNAs in lipid metabolism, with a prediction that miR-103/107 regulates human metabolic pathways. Mol. Genet. Metab. 91(3), 209–217 (2007).
    • 142 Ramaswamy G, Karim MA, Murti KG, Jackowski S. PPARalpha controls the intracellular coenzyme A concentration via regulation of PANK1alpha gene expression. J. Lipid Res. 45(1), 17–31 (2004).
    • 143 Trajkovski M, Hausser J, Soutschek J et al. MicroRNAs 103 and 107 regulate insulin sensitivity. Nature 474(7353), 649–653 (2011).
    • 144 Foley NH, O'neill LA. miR-107: a Toll-like receptor-regulated miRNA dysregulated in obesity and Type 2 diabetes. J. Leukoc. Biol. 92(3), 521–527 (2012).
    • 145 Catalioto RM, Maggi CA, Giuliani S. Chemically distinct HDAC inhibitors prevent adipose conversion of subcutaneous human white preadipocytes at an early stage of the differentiation program. Exp. Cell Res. 315(19), 3267–3280 (2009).
    • 146 Hesketh J, Wybranska I, Dommels Y et al. Nutrient–gene interactions in benefit-risk analysis. Br. J. Nutr. 95(6), 1232–1236 (2006).
    • 147 Goni L, Cuervo M, Milagro FI, Martinez JA. Future perspectives of personalized weight loss interventions based on nutrigenetic, epigenetic, and metagenomic data. J. Nutr. doi:10.3945/jn.115.218354 (2016) (Epub ahead of print).
    • 148 Goni L, Milagro FI, Cuervo M, Martinez JA. Single-nucleotide polymorphisms and DNA methylation markers associated with central obesity and regulation of body weight. Nutr. Rev. 72(11), 673–690 (2014).
    • 149 Crujeiras AB, Diaz-Lagares A, Moreno-Navarrete JM et al. Genome-wide DNA methylation pattern in visceral adipose tissue differentiates insulin-resistant from insulin-sensitive obese subjects. Transl. Res. 178, 13e15–24e15 (2016).
    • 150 Kohlmeier M, De Caterina R, Ferguson LR et al. Guide and position of the international society of nutrigenetics/nutrigenomics on personalized nutrition: part 2 – ethics, challenges and endeavors of precision nutrition. J. Nutrigenet. Nutrigenomics 9(1), 28–46 (2016). • A comprehensive review on nutrigenomics and nutrigenetics in personalized medicine.