ABSTRACT
The fact that not all individuals exposed to the same environmental risk factors develop obesity supports the hypothesis of the existence of underlying genetic and epigenetic elements. There is suggestive evidence that environmental stimuli, such as dietary pattern, particularly during pregnancy and early life, but also in adult life, can induce changes in DNA methylation predisposing to obesity and related comorbidities. In this context, the DNA methylation marks of each individual have emerged not only as a promising tool for the prediction, screening, diagnosis, and prognosis of obesity and metabolic syndrome features, but also for the improvement of weight loss therapies in the context of precision nutrition. The main objectives in this field are to understand the mechanisms involved in transgenerational epigenetic inheritance, and featuring the nutritional and lifestyle factors implicated in the epigenetic modifications. Likewise, DNA methylation modulation caused by diet and environment may be a target for newer therapeutic strategies concerning the prevention and treatment of metabolic diseases.
KEYWORDS: Epigenetics, precision nutrition, gastric surgery, energy-restriction
Introduction
Obesity has been defined as an abnormal or excessive fat accumulation that contributes to the development of many associated comorbidities such as type 2 diabetes (T2D), cardiovascular diseases (CVDs), dyslipidaemia, hypertension, liver steatosis, or metabolic syndrome (MetS), among others [1]. Obesity is attributed to the maintenance of a positive energy imbalance between caloric input and output [2]. Several factors could affect the energy equation, such as lifestyle behaviours (dietary habits, exercise, and sleep patterns) [3,4], social factors (educational level and economic status) [5], endocrine disorders (hypothyroidism) [6], or prescription of certain medication (corticosteroids) [7]. The current notion on the pathogenesis of obesity is that not all individuals under the same environmental factors develop the disease, so it has been suggested that genetics play a role [8]. However, genetic risk loci identified by genome-wide association studies only explain part of the genetic variance in disease risk, suggesting that more factors must be taken into account to understand the multifactorial pathology of obesity. Epigenetics has therefore, been proposed as a factor involved in obesity development because it is influenced by both genetic and environmental factors [9]. Most recently, epigenetics has offered new explanations about the mechanisms, where environmental factors modify the expression of genes related to obesity and its comorbidities [10].
As obesity has been attributed to a positive energy balance [2], reduction of energy intake, changes in macronutrient distribution, increase of physical activity, behavioural approaches, and pharmacological or surgical treatments are commonly used to produce a negative energy balance [3]. Although many strategies have been investigated for inducing weight lowering, individual response varies widely. A better understanding of the contribution of diet and nutrients to epigenetics is necessary, as weight loss is crucial for reducing the negative medical consequences of obesity. This review describes the current advances in the use of DNA methylation in the assessment of metabolic risk and the personalization of the clinical management of obesity.
Epigenetics
Epigenetics is defined as the processes that alter gene activity without nucleotide sequence modification, but including chromatin structure changes as a direct consequence [11].
The most relevant epigenetic mechanisms involved in gene activity regulation are DNA methylation, histone modifications, and non-coding RNAs. The most extensively studied epigenetic mark in the mammalian genome in relation to gene expression regulation has been found to be DNA methylation [12], which is an epigenetic mechanism involving the covalent addition of a methyl group (-CH3) onto the five position of cytosine, resulting in 5-methylcytosine. Although in mammals methylation is generally restricted to cytosine linked by a phosphate to guanine (CpG site), a small percentage of methylation may occur in non-CpG sites (CHG and CHH, where H = A, C, or T) [13,14]. Similar to methylation, hydroxymethylation is an epigenetic mechanism that replaces, at the C5-position in cytosine, the hydrogen atom by a hydroxymethyl group.
Mammalian genomes present approximately 28 million CpG sites, among which 60–80% are generally methylated. Genomic regions with a high CpG density are known as CpG islands (CGIs). The majority of the CGIs are located in gene promoter regions, but they also appear in gene bodies, often acting as alternative promoters [15]. Contrary to CpG sites, most of the CGIs in promoter regions are usually unmethylated to maintain transcription of the active gene. A differentially methylated region (DMR) is an area of the genome where multiple adjacent CpGs show different methylation status between phenotypes. DMRs are regarded as possible functional regions involved in gene transcriptional regulation [16]. In this context, differential regional DNA methylation is more biologically interpretable and statistically powerful than CpGs measured individually [17]. These characteristics allow DMR to be powerful biomarkers of biological and pathogenic processes, or pharmacological responses to a therapeutic intervention.
Obesity, related comorbidities, and DNA methylation
Obesity is considered the pandemic of the 21st century (WHO, 2017). The prevalence of obesity closely tripled between 1975 and 2016. In 2016, more than 1.9 billion adults were overweight and 650 million were obese (WHO, 2017). Also among children and adolescents, the obesity prevalence has hugely increased in the last decades. A recent meta-analysis concerning 2,416 population-based studies in 128.9 million children and adolescents found an increase in the prevalence of obesity worldwide from 0.7 % in 1975 to 5.6% in 2016 in girls, and from 0.9% in 1975 to 7.8% in 2016 in boys, suggesting that in 2022 child and adolescent obesity would surpass moderate and severe underweight [18].
Despite extensive efforts, obesity prevalence continues to increase in every country. At the same time, a number of studies have tried to deepen into the molecular origin of obesity and related disorders, focusing on epigenetic signature, during the different stages of life [19–26]. In this context, research on novel epigenetic biomarkers is required to understand the pathophysiology of obesity and its clinical manifestations. These investigations described more than a hundred of genes distributed over all chromosomes (except sex chromosomes) (Figure 1). The lack of genes in sexual chromosomes, which is common with studies focused on other pathological conditions, is due to the methodological processing of the microarray data (which traditionally excludes sex chromosomes from the analysis), although the few studies including sex chromosomes did not find relevant genes with DNA methylation changes in obesity-related conditions.
Figure 1.

Karyogram representing genes loci whose DNA methylation levels have been associated with obesity (blue), metabolic syndrome features (green), and weight loss (red). The genes represented in Figure 1 have been described in Tables 1, 2, 3, and 4, where additional information about each gene is included.
Transgenerational epigenetic inheritance in obesity
DNA methylation modifications may be dynamic and altered as a consequence of environmental stimuli or may be stable and passed on to next generations [27]. Several studies, especially in animal models, reported that exposure to environmental factors in one generation caused phenotypical effects in unexposed future generations and considered DNA methylation mechanism in origin. This phenomenon is called transgenerational epigenetics inheritance [28]. This transfer is explained by an incomplete erasure of the epigenetic modifications during gametogenesis and early embryogenesis, some of which are transferred to the offspring [28]. A few cases of this phenomenon have been identified in mammals. For example, an intracisternal A-particle retrotransposon inserted in the agouti gene results in ectopic expression of agouti protein, causing yellow fur, obesity, diabetes, and a raised tendency to tumours in the next generations of mice [29]. Due to the public health implications of obesity and metabolic diseases, a number of investigations have studied the epigenetic inheritance of nutritional metabolic risk in mammals, especially in rodents. The adverse perinatal environment can affect the phenotype of the offspring and subsequent generations. For example, rats suffering a continuous 50% caloric restriction during 50 generations, exhibited higher obesity-related metabolic disturbances associated with modified epigenetic marks in insulin-2 (Ins2) gene promoter region [30]. Interestingly, these alterations in metabolism and epigenetic signature were not reversed after two generations of normal chow (restoration of normal nutrition) [30]. On the other hand, the exposure to chemical agents is also capable of promoting epigenetic transgenerational inheritance in rodents. For instance, exposure to plastic-derived endocrine disruptors and hydrocarbons in gestating F0 generation females derived in an increase of total abnormalities, such as testis disease, obesity, and ovarian diseases, in F3 generation male and female rodents [31,32]. These abnormalities in subsequent new-borns were associated with altered global DNA methylome in sperm of male rats [31,32].
In humans, although no studies have clearly demonstrated the transgenerational epigenetic inheritance of metabolic abnormalities, several investigations have observed the relationship between nutritional imbalances and adverse health outcomes. One of the most studied famines happened in the populated western provinces of the Netherlands during the winter of 1944–1945 (The Dutch Hongerwinter). The distributed food rations dropped to below 1000 Kcal and were reduced to extremely 500–600 Kcal [33]. Infants who were conceived during this period presented higher rates of obesity [34], hypercholesterolemia [35], and/or cardiovascular disease [36] in adulthood. Interestingly, these health problems developed in the first generation were also found in the second generation [37]. The DNA isolated from these subjects showed below-average methylation of insulin growth factor 2 (IGF2) and higher expression of the hormone, even six decades later [38]. Further studies described an increase in the methylation of 6 other genes in the Dutch Hongerwinter subjects, including IL-10, LEP, ABCA1, GNASAS1, MEG3, and INSIGF2, some of which are associated with cholesterol transport, ageing, or schizophrenia [39]. Other famine period occurred in a rural area in the Chittagong division of Bangladesh in 1974–1975 as a consequence of severe monsoon. Young adults exposed to famine during gestation presented low weight and hyperglycaemia, and significant differences in DNA methylation at seven metastable epialleles (genomic regions that exhibit inter-individual epigenetic variation), such as Vault RNA 2–1 (VTRNA2-1), paired box 8 (PAX8), PR/SET domain 9 (PRDM9), near ZFP57 zinc finger protein (ZFP57), near BolA family member (BOLA), and exonuclease 3ʹ-5ʹ domain containing 3 (EXD3) [40]. In rural Gambia the nutrient availability fluctuate with wet and dry seasons creating annual hungry periods during wet season (July–October). The studies in children gestated during famine periods demonstrated persistent DNA methylation changes in metastable epialleles [41] that were associated with different metabolic alterations in the offspring. Although nutrition during pregnancy affects the phenotype in the offspring and can affect the third generation, transgenerational epigenetic inheritance during more than three generations is not clear in humans. In any case, these studies could be included in the context of the DOHaD (developmental origins of health and disease) paradigm, which suggests that environmental conditions during embryonic development and early life modify disease susceptibility during adult life [42].
In conclusion, further epidemiological studies controlling the influence of prenatal and postnatal nutrition and environment, and well-designed experiments in humans, are needed to fully understand transgenerational epigenetic inheritance in humans in relation to obesity and other metabolic disturbances.
Factors affecting DNA methylation in the context of obesity
A number of studies have found evidence that epigenetic mechanisms are involved in the onset and development of obesity and its metabolic complications. Many of these epigenetic mechanisms are modulated by the environment, and the responsible factors can be divided into two major categories: nutritional and physiological.
Nutritional factors
In the last years, different dietary patterns, nutrients and food components have been related to epigenetic processes that may contribute to an increase in the susceptibility of obesity development and other metabolic disorders. The sensitivity of the epigenetic system to nutritional factors occurs mainly during periods of developmental transition because it is the time when epigenetic marks are critically modified [43]. Thus, inappropriate feeding practices and nutrient deficiencies during pregnancy and lactation might have long-term consequences in offspring’s health [44]. The available studies explained above (i.e., Dutch Hunger Winter and Gambian children) provide a bright example of how early and mid-gestation can lead to metabolic dysregulation later on life, having been associated with cardiovascular diseases, obesity or hyperglycaemia [37]. Furthermore, as one-carbon metabolism depends on the dietary methyl donors, DNA methylation can be influenced by folate, methionine, betaine, or choline availability during this stage. Thus, methyl donor levels in the maternal diet have been shown to modify the establishment of the DNA methylation profile in the foetus and have adverse metabolic effects during infancy and childhood [39,40]. For example, maternal deficiency of folate has been associated with higher inflammatory mediators (interleukins, TNF-α, and monocyte chemoattractant protein-1), but also with an increased risk of cardiovascular diseases, obesity and insulin resistance in the offspring, in both humans and rodents [45–47]. Maternal choline and betaine deficiencies also alter the methylation status of genes that participate in growth (IGRF2, Crh, and Nr3c1) or vascular function (VEFGC and Angpt2), resulting in developmental abnormalities, blood vessel malformation, or increased predisposition for developing steatohepatitis in the offspring [48–51]. These investigations are focused on epigenetic changes related to the development of metabolic abnormalities in offspring whose mothers have suffered nutritional stress. However, the paternal diet, lifestyle, and obesity and diabetes may also induce alterations in DNA methylation signatures in the offspring that can lead to different metabolic consequences [52]. In humans, the Överkalix study showed that grandparental access to food can modify diabetes risk and all-cause mortality in grandchildren [53]. The Uppsala Multigeneration Study found an elevated mortality (mostly from cancer) in grandsons whose paternal grandfathers had good access to food just before puberty [54]. These studies propose that maternal and paternal malnutrition can result in an obesity phenotype in the offspring and suggest that nutritional factors affect DNA methylation in humans. The malnutrition caused by overnutrition (when the intake of nutrients is oversupplied) is also able to induce epigenetic alterations in the promoters of key metabolic genes that could affect gene expression and predispose to metabolic alterations [52]. Studies about the effects of high-fat diets have associated diets rich in saturated and trans-fatty acids with damages in intestinal permeability, obesity, and inflammation as well as cardiovascular abnormalities [55,56]. For instance, palmitic acid induces global hypermethylation in human pancreatic islets, impairing insulin secretion and increasing the risk of type 2 diabetes [57]. Supplementation with saturated fatty acids and elaidic acid (trans fatty acid) induces TNF hypomethylation and PPARG1 hypermethylation, increasing and decreasing the expression of these genes and leading to an inflammatory environment in adipose tissue [58].
On the other hand, different in vitro and animal studies have reported that specific phytochemicals (i.e., catechins, curcumin, quercetin, genistein, resveratrol, and sulforaphane) are able to reverse adverse epigenetic markings by acting on DNA methylation, histone modifications, and microRNA expression (revised in [23]). It is possible that some of the beneficial metabolic effects of these compounds could be mediated by epigenetic mechanisms, due to their ability to inhibit DNMT activity by increasing S-adenosylhomocysteine levels or inhibiting the catalytic site of DNMTs [59]. For example, in humans, inverse associations have been found between flavonoid intake and body weight [60], suggesting that polyphenol-targeted epigenetics might be an attractive approach for obesity prevention. Nevertheless, few intervention studies in humans have found a positive effect of polyphenol supplementation on body weight. In any case, studies in humans are still ongoing and more information is needed about amounts, combinations and influence of gut microbiota on the epigenetic effects of phytochemicals in the context of obesity.
Obesity-related physiological factors
Several obesity-related physiological factors have also been associated with the dysregulation of epigenetic processes. Most of them have been studied in vitro but it is extremely difficult to study them isolatedly in humans. In obese patients, it is usual to find more than one of these factors, and their effects on the epigenome are also masked by the commented nutritional factors.
Low-grade chronic inflammation is considered an essential link between obesity and its comorbidities. Moreover, in some studies inflammation has been reported as a predictor of weight gain [61], whereas a lower expression of genes related to inflammation and immune response has been associated with better response to weight loss treatments [62]. Many studies have demonstrated that inflammatory mediators are able to induce changes in the epigenetic landscape. In addition, DNA methylation also has a functional role in the monocyte-to-macrophage differentiation and macrophage polarization [63]. Similarly, dynamic DNA methylation and demethylation activity is necessary to develop and differentiate T linage [64]. However, the effects and amounts of the different molecules, the interactions among them and the differential effects on the different cell types are still unknown.
Hypertrophy and hyperplasia of adipocytes in the obese adipose tissue decrease oxygen availability and induce hypoxic stress, which is considered to be an important mechanism involved in obesity-related chronic inflammation, IR and mitochondrial dysfunction [65]. Hypoxia-inducible factors (i.e., HIF3A) are both, regulated by epigenetic mechanisms, and related to adipose tissue dysfunction [65], which makes hypoxia a good epigenetic target to treat obesity-related complications. Hypoxic conditions favour the increase of reactive oxygen species and oxidative stress, both described as important inflammatory mediators [66]. Oxidative stress is the result from an imbalance between tissue oxidants and antioxidants, and is usually exacerbated in obese individuals. Not only oxidative stress, but also weight loss and dietary antioxidants, which may help to decrease this phenomenon, have been reported to induce changes in the epigenetic landscape [67].
Obesity is almost always associated with IR, where poor glycaemic control has been linked to epigenetic alterations that could be involved in the development of diabetes-associated comorbidities [68]. According to this, obese subjects would be more susceptible to episodes of transient hyperglycaemia that could impair cell structure and function by means of DNA and histone epigenetic alterations. In this context, a recent study has analysed the epigenetic signatures associated with IR, measured by the Homeostatic Model Assessment of IR (HOMA-IR), and has revealed that 478 CpGs were differentially methylated between individuals with HOMA-IR ≤ 3 and > 3, especially in genes that are mainly involved in glucose- and insulin-related pathways [69].
Psychological stress has become a typical trait of the modern way of life. It involves chronic activation of the neuroendocrine systems and has been associated with increased prevalence of obesity [70]. In this context, cortisol-based and perceived psychological measures of stress may be important predictors of obesity-related risks [71]. In relation to this, stress, particularly at early stages of life, has been associated with hormonal alterations and epigenetic modifications that can persist into adulthood and be a causative factor of obesity. For example, it has been demonstrated that DNA methylation mediates the impact of exposure to prenatal maternal stress on body mass index (BMI) and central adiposity in children [72].
Among the obesity-related physiological factors, disruption of the circadian rhythm may play a key role in obesity development. In mammals, the synchronization between circadian rhythms and environmental stimuli, including daily rhythms of natural light, external temperature and food intake, is driven by the circadian system, a hierarchical multilevel organization [73]. Sleep deprivation, shift work, bright light exposure at night, and unusual eating time have been related to obesity and diabetes [74]. Many metabolic functions, such as glucose homeostasis, lipid metabolism and fasting/feeding cycles, are regulated by the internal clock system [73]. In this context, several studies have described that different nutritional and environmental factors, including obesity, are able to affect the DNA methylation pattern of the clock genes that regulate circadian rhythm in the hypothalamus and peripheral tissues [75]. On the other hand, it has been also described that insufficient sleep (short sleep duration or insomnia) is associated with loss of DNA methylation, which could be related to impairments in processes related to neuroplasticity, neurodegeneration, and cardio-metabolic condition [76].
Moreover, there is growing evidence that a number of endocrine-disrupting chemicals (such as phthalates, p,p’-dichlorodiphenyldichloroethylene and bisphenol A), heavy metals (such as lead or cadmium), toxins, and drugs, particularly in early-life, may contribute to obesity development and metabolic impairment, at last in part, by altering epigenetic profiles and gene expression regulation [77,78]. However, more research in cells and humans is needed to understand the intimate mechanisms.
Human gastrointestinal tract is densely populated by bacteria, fungi, and viruses among others, collectively called gut microbiota [79]. These organisms play important physiological roles in digestion, vitamin synthesis, and metabolism [80]. The microbiome is a well-known epigenetic regulator, in part because of the production of bioactive molecules, such as short-chain fatty acids (propionate, butyrate, and acetate), and because of its capacity to modulate the inflammatory response [81]. Gut microbiota composition has been associated with a number of diseases including obesity [82] and is the perfect link between host genetics, diet, and epigenetics. In recent years, many studies are being conducted to analyse these interactions, with special emphasis on topics such as the role of maternal and neonatal nutrition on microbiota composition, the early establishment of the foetal and neonatal microbiome, the ability to reshape the epigenome of the inflammatory cells, the identification of the microbial metabolites that affect the mammalian epigenetic machinery, and the potential epigenetic effects on genes regulating important metabolic functions such as appetite, beta cell function and the browning of adipose tissue [83,84].
In addition, the decline in physical activity is one of the alleged perpetrators for the modern epidemic of obesity. Physical exercise has been demonstrated to induce genome-wide changes in DNA methylation in humans, both in adipose tissue [85] and skeletal muscle [86], potentially affecting cell metabolism and inducing health-enhancing phenotypic adaptations. However, there is not enough information about the amounts and types of physical activity that can be more beneficial for obesity prevention and treatment in relation to their effects on the epigenome.
Finally, other factors that could potentially affect the epigenome in relation to obesity risk, because they can exert an influence on energy intake and energy expenditure, are hormonal and neuropeptide imbalances, anxiety and depression, infections, ambient temperature, artificial sweeteners, air and noise pollution. However, all of them need to be demonstrated in humans.
DNA methylation markers in obesity
Numerous cross-sectional studies have reported a significant association between obesity or adiposity status, and DNA methylation (Table 1). For example, the offspring of obese mothers presented several CpG sites differentially methylated in cord blood in comparison with offspring from normal weight mothers [87]. Other regions studied, were mainly hypomethylated in obese children and located in the gene body region, and revealed a unique cluster of obese individuals that was differentiated from the normal weight children [88]. In addition, some of these genes are implicated in lipid and glucose metabolism, differential body size and body composition in children [89]. Another analysis in adult population found methylation changes in carnitine palmitoyltransferase 1A (CPT1A) gene, associated with obesity and T2D [90]. CPT1A is also implicated in the control of fasting triglycerides (TG) and very low-density lipoprotein (VLDL) levels [91]. These studies suggest an important role for this gene in obesity and metabolic complications.
Table 1.
Cross-sectional studies analysing the association between DNA methylation and obesity.
| Study population | Sample type | Target gene | Major finding | Age/Sex adjusted | Reference |
|---|---|---|---|---|---|
| Children (n = 2,861) | Cord blood | Genome-wide | 28 CpG differentially methylated between offspring of obese and normal weigh mothers. 0.01–1.84% change in DNA methylation per 1 kg/m2 of BMI Maternal weight modifies offspring epigenome |
x/✓ | [87] |
| Obese (n = 6) and non-obese (n = 6) children |
PBC |
21orf56, ZNF154, SDK1, KIAA0146, SKIV2L, GPR125, SORBS2,C14orf70, POLR3E, CTBP2,DLGAP2,CAPS2,GIMAP1, RNF213,MND1,SRM,TGM6,WDR27 |
Different DNA methylation profile between obese/non-obese children Markers for early diagnosis of obesity |
-/- | [88] |
| Children (n = 374) | PBC |
SNED1(IRE-BP1),KLHL6, WDR51A(POC1A),CYTH4-ELFN2, CFLAR, PRDM14, SOS1, ZNF643(ZFP69B),ST6GAL1, C3orf70, CILP2, MLLT4 |
Association between DNA methylation and BMI, fat mass, lipid, and glucose metabolism 1% change in DNA methylatin per 0.1–0.4% of variables |
✓/✓ | [89] |
| Obese (n = 24) and non-obese (n = 23) |
PBC | CERCAM, DPYD, IL12A, ZNF35, ZNF362, TSC22D2, CBX6, FOXF1, PSMD7, H1FX, PRRC2C, MSI1, COL4A1, NBPF3, USP5, PLOD2, TLE3, RPS24, DVL3, POLD3 | 20 genes differentially methylated between obese and non obese adults 2.8–30.8% differences in DNA methylation between groups Biomarkers for the understanding of obesity |
✓/✓ | [147] |
| Adults (n = 547) | Blood | 15 DMP and CPT1A | Association between DNA methylation and BMI, obesity, and abdominal adiposity | ✓/✓ | [148] |
| GOLDN (n = 991), ARIC (n = 2,106), and FHS (n = 442) cohorts |
CD4+ T-cells |
CPT1A, CD38, PHGDH | Association between DNA methylation and BMI and WC | ✓/✓ | [90] |
| Popgen biobank (n = 49) | PBC | MCHR1 | DNA methylation associated with BMI DNA methylation contributes to the age-related specific effects |
x/- | [149] |
| Non-obese (n = 90) and obese (n = 171) children |
PBC | POMC | Hypermethylaton associated with obesity 37.5% differences in DNA methylation between groups Appetite regulation |
✓/✓ | [150] |
| Obese (n = 7) and non-obese (n = 7) adults |
PBL | UBASH3A, TRIM3 | Hypermethylation of UBASH3A and hypomethylation of TRIM3 genes in obese subjects compared with lean controls 2.5–10.1% differences in DNA methylation between groups |
✓/✓ | [26] |
| Adults (n = 40) | PBMC | SERPINE1, LINE-1 | Association between hypermethylation of LINE-1 and SERPINE1, and WC and BMI | ✓/✓ | [151] |
| RESMENA (n = 48) and OBEPALIP (n = 25) cohorts | WBC | GPR13, ITGB5 | Association between DNA methylation and BMI > 10% differences in DNA methylation between groups |
✓/✓ | [152] |
| Qatari descent (n = 30) and TwinsUK (n = 32) cohorts | Blood |
CPT1A, TXNIP, ABCG1 |
Association between hypomethylation of CPT1A and TXNIP and obesity Association between hypermethylation of ABCG1 and BMI 2.7–3.77 rate of change of T2D characteristics as methylation changes |
✓/✓ | [112] |
| Men with hypercholesterolemia (n = 61) and morbid obese men(n = 30) | VAT and blood |
ADRB3 | Association between hypermethylation of ADRB3 gene and visceral obesity and fat distribution in VAT, and LDL-C and waist-to-hip ration in blood | ✓/- | [93] |
| Obese (n = 50) and non-obese girls (n = 50) |
Saliva |
NAV3, MC2R |
Association between hypermethylation and BMI 11.6% differences in DNA methylation between groups |
-/- | [96] |
| Adults (n = 431) | Saliva | LEP | Association between DNA methylation and BMI | ✓/- | [94] |
| Children (n = 92) | Saliva | Genome-wide | Association between 17 CpGs methylation and BMI | ✓/✓ | [95] |
| Adults (n = 43) | Blood and oral mucosa |
GAP43, ATP2A3, ADARB2 |
Association between genes methylation and overweight | x/x | [97] |
| NEST cohort (n = 92) | Blood | PLAGL1, NEG3 | Hypermethylation of PLAGL1 and hypomethylation of NEG3 in obese mothers 1.5–3.9% differences in DNA methylation between groups |
✓/x | [153] |
| Children (n = 88) | Cord blood |
MMP7, KCNK4, TRPM5, NFKB1 |
Association between GWG and hypermethylation of genes | ✓/✓ | [154] |
| BBC cohort (n = 309) | Blood leucocytes | Genome-wide | Association between maternal BMI and DNA methylation of 20 CpGs | ✓/✓ | [155] |
| Cardiogenic Consortium (n = 479), MARTHA (n = 339), KORA F4 (n = 1789), and MuTHER (n = 635) cohorts | Blood and adipose tissue | HIF3A | Association between DNA hypermethylation of gene and BMI 2.3–4.6% increase in BMI as DNA methylation changes in 0.1% |
✓/✓ | [130] |
| MZ twin pairs (n = 84) | PBC | SLC6A4 | Association between DNA hypermethylation of gene and BMI, weight and WC Increase 0.3 kg/m2 BMI, 0.16 kg body weight, and 0.78 cm in WC per 1% of DNA methylation |
✓/✓ | [156] |
| ARIC cohort (n = 2097) | WBC | Genome-wide | Association between eight CpG methylation and BMI changes | ✓/✓ | [157] |
| Three cohorts: BPRHS, GOLDN and FHS (n = 1986) | CD4+, WBC | APOA2 | Association between DNA methylation status of the gene with SFA intake and obesity risk | ✓/✓ | [158] |
Abbreviations: CpG, cytosine linked by a phosphate to guanine; PBC, peripheral blood cells; BMI, body mass index; DMP, differentially methylated positions; GOLDN, Genetics of Lipid Lowering
Drugs and Diet Network; ARIC, Atherosclerosis Risk in Communities; FHS, Framingham Heart Study; PBMC, peripheral blood mononuclear cells; WC, waist circumference; RESMENA, Metabolic
Syndrome Reduction in Navarra; OBEPALIP, Effects of Lipoic Acid and Eicosapentaenoic Acid in Human Obesity; WBC, white blood cells; VAT, visceral adipose tissue; LDL-c, low-density
lipoprotein cholesterol; NEST, The Newborn Epigenetic Study; GWG, gestational weight gain; BBC, Boston Birth Cohort; MARTHA, MARseille THrombosis Association; KORA, Cooperative
Health Research in the Region of Augsburg; MuTHER, Multiple Tissue Human Expression Resource; MZ, monozygotic; BPRHS, Boston Puerto Rican Health Study; SFA, saturated fatty acid. ✓,
adjusted; x, no adjusted; -, no information
A study in three populations of diverse ancestries found that the methylation levels of the apolipoprotein A2 (APOA2) regulatory region was associated with the consumption of saturated fatty acids [50], a nutritional factor that has been associated with an increase in the risk of obesity in previous studies.
Not only is the methylation profile of blood cells associated with obesity, but also the methylation of other tissues and body fluids. An EWAS identified predominantly DNA hypermethylation in white adipose tissue (WAT) from obese subjects, which was related to gene expression of proinflammatory pathways. These findings suggest that DNA methylation may link dysfunctional adipocytes to WAT inflammation and IR in obesity [92]. Hypermethylation of the beta-3 adrenoceptor (ADRB3) gene was also reported in WAT of obese subjects, which was linked with an increased susceptibility to visceral obesity and altered body fat distribution [93]. Similar results were previously described in blood cells [93], revealing that the methylation changes of ADRB3 gene in blood may reflect obesity-related DNA methylation changes of this gene in WAT. In addition, a case-control study demonstrated an association between hypomethylation of leptin (LEP) gene in obese individuals, measured in saliva, and obesity-related parameters [94]. Likewise, an EWAS performed in 92 children’s saliva samples described 17 CpGs associated with maternal BMI [95]. The saliva sample analysis in 50 girls with and without obesity found two interesting genes, neuron navigator 3 (NAV3) and melanocortin 2 receptor (MC2R), whose methylation levels were associated with BMI [96]. These studies evidenced that saliva may be a viable probe for epigenetic testing in obesity. However, further studies would have to include both, saliva and blood samples, to demonstrate that saliva is consistent with whole blood findings. With this purpose, a recent study provided a novel application of noninvasive buccal samples for the identification of DNA methylation markers in relation to the management of overweight and insulin sensibility [97].
Most of the investigations analysing DNA methylation in relation to the pathophysiology of obesity are performed in a cross-section studies, where the data were collected by the observation of the variable at the same time point. In most of the populations, it has been difficult to rule out other effects such as the influence of nutritional status, nutrients, diet, or genetics. Moreover, in some of these cohorts, important covariates are missing. Even though all these studies back the DNA methylation-obesity link hypothesis, most of them are association studies and are far from causative and can only correlate DNA methylation with phenotypic outcomes. In this sense, few longitudinal studies have tried to investigate the causal effect of DNA methylation on obesity and related measures (Table 2).
Table 2.
Longitudinal studies analysing the association between DNA methylation and obesity.
| Study population | Sample type | Target gene | Major finding | Age/Sex adjusted | Reference |
|---|---|---|---|---|---|
| Botnia prospective study: T2D (n = 129); controls (n = 129), 8 years |
Blood | PHOSPHO1, ABCG1 | Associated with BMI, insulin, TG. | ✓/✓ | [104] |
| Children (n = 438), 5 years | Blood | Genome-wide | Associated with obesity, BMI z-score, HOMA-IR, insulin and glucose | ✓/✓ | [98] |
| GUSTO cohort (n = 987), 4 years | Blood | CDKN2B/P4HA3 | Associated with BMI | x/✓ | [99] |
| CHAMACOS study (n = 373), 9 years | Blood | PPARγ | Associated with body size | x/✓ | [159] |
| Two birth cohorts: ALSPAC (n = 178), 7 years, and PTBGS (n = 24), 11–13 years | Blood | CDKN1C, EPHA1, CASP10, HLA, NID1 | 2.08% and 0.80% increase in BMI per 1% increase in methylation at CDKN1C and EPHA1 respectively Increase of 5.16% and 1.84% in fat mass per 1% increase in methylation respectively. | ✓/✓ | [100] |
| Two prospective cohorts: PAH, (n = 78), 9 years and SWS (n = 239), 6 years | Blood | RXRA | Higher methylation of RXRA, was associated with lower maternal carbohydrate intake and neonatal adiposity and fat mass | ✓/✓ | [101] |
| Two cohort: NPBGS (n = 94), 11 years and ALSPAC (n = 161), 7 years | Blood | TACSTD2 | Hypomethylation associated with higher fat mass | ✓/✓ | [102] |
| BSCC study (n = 600), 2.5 years | Blood | LINE-1 | Hypomethylation associated with adiposity | -/- | [103] |
| POUNDS Lost Trail (n = 48) | Blood | NFATC2IP | A 52.8% impact on weight change mediated by the cis DNA methylation at NFATC2IP | -/- | [105] |
Abbreviations: T2D, type 2 diabetes; BMI, body mass index; TG, triglycerides; HOMA-IR, homeostasis model assessment-insulin resistance; GUSTO, Growing Up in Singapore Towards healthy Outcomes; CHAMACOS, Center for the Health Assessment of Mothers and Children of Salinas; ALSPAC, Avon Longitudinal Study of Parents and Children; PTBGS, Preterm Birth Growth Study; PAH, Princess Anne Hospital; SWS, Southampton Women’s Survey; NPBGS, Newcastle Preterm Birth Growth Study; BSCC, Bogota School Children Cohort; POUNDS, The Preventing Overweight Using Novel Dietary Strategies. ✓, adjusted; x, no adjusted; -, no information.
Most of the longitudinal and prospective studies have been focused on the paediatric population, where obesity is not usually accompanied by other comorbidities that may confound the results. Prospective studies during the early life stage described DNA methylation of several weight-linked loci in new-borns that continued to show a longitudinal association with adiposity, fat mass, body size and other obesity parameters in childhood [98–103]. Interestingly, several prospective studies including adult life stage evidenced that these DNA methylation pattern changes were associated with the later risk of developing metabolic and other diseases [104,105]. These examples described an association between early cues (environmental stimulation in utero, in early infancy, and in adult stage) and the later development of disease. Actually, this is the basis of the ‘epigenotype model’ of DOHaD, in which the environment can modulate the epigenetic signature during human life.
DNA methylation and MetS
MetS is characterized by a cluster of risk factors associated with CVD, diabetes, obesity, and stroke, which include visceral adiposity, IR, hypertension, hypertriglyceridemia, and low high-density lipoprotein cholesterol (HDL-C) phenotypes [106]. There are several criteria for the diagnosis of MetS. According to the World Health Organization (WHO) definition, the diagnosis of MetS requires obesity, hyperinsulinemia plus two or more other clinical components. However, the most used definition is the revised Adult Treatment Panel-III, which requires at least three or more alterations.
The prevalence of MetS is rapidly increasing in most countries, affecting more than 20% of the adult population globally [107]. In 2010, the Spanish adult population presented higher prevalence of MetS, specifically, 38.37% in men and 29.62% in women, these percentages increasing in the elderly [108]. The high prevalence of MetS makes the implementation of new strategies for prevention necessary.
Numerous studies in humans have investigated the possible association between DNA methylation and MetS features, including visceral adiposity, IR, hypertension, HDL-C, and hypertriglyceridemia. Most of them observed that the DNA methylation patterns of different genes were associated with MetS markers, resulting in a worse prognosis or a higher risk of presenting other diseases (Table 3). Moreover, several studies have found evidence that MetS risk factors are likely to play a role in the DNA methylation process. Modification of gene methylation has been reported in an initial phase of the development of MetS.
Table 3.
Studies analysing the association between DNA methylation and MetS features.
| Study Population | Sample type | Methylation strategy | Target gene | Major results | Age/Sex adjusted | Reference |
|---|---|---|---|---|---|---|
| GDM (n = 87) and healthy pregnant (n = 81) | Umbilical cord blood | Gene-specific | GNAS | GDM increases methylation level at foetal GNAS Hypermethylation of GNAS associated with high risk for diseases in offspring |
-/- | [109] |
| GDM dietetically treated (n = 88), GDM insulin-dependent (n = 98), healthy pregnant (n = 65) | Cord blood and chorionic villi | Gene-specific | MEST | GDM decreases MEST methylation in new-born 7.8% differences in DNA methylation between groups MEST hypomethylation contributes to risk of development MetS |
✓/✓ | [110] |
| Controls (n = 70) and MetS subjects (n = 64) | VAT | Gene-specific | LPL | Association between LPL hypermethylation and aetiology of MetS | ✓/✓ | [19] |
| GDM (n = 82) and T1D in pregnancy (n = 67) | Skeletal muscle, SAT and blood | Gene-specific | TXNIP | Association between intrauterine hyperglycaemia and changes in TXNIP methylation 5.2% differences in DNA methylation between groups |
✓/✓ | [160] |
| Controls (n = 48) and T2D (n = 12) | Pancreatic islets | Gene-specific | PPARGC1A |
PPARGC1A hypermethylation in T2D subjects Association between PPARGC1A hypermethylation and lower insulin secretion 55.2% differences in DNA methylation between groups |
✓/- | [114] |
| FH (n = 61) and severely obese non-FH (n = 30) | Blood and VAT | Gene-specific | ADRB3 | Association between ADRB3 hypomethylation and higher LDL-c, WC and apoB levels 28.8% differences in DNA methylation between groups |
✓/- | [93] |
| Healthy men (n = 13) | Muscle biopsies | Gene-specific | ANGPTL4 | Association between ANGPTL4 hypomethylation and low blood glucose and insulin sensitivity | -/- | [161] |
| Adults (n = 294) | SAT and blood | EWAS | 2825 genes, 711 CpGs | Association between 2825 gene methylation and BMI Association between 711 CpGs methylation and HbA1c |
✓/✓ | [111] |
| Eight retrospective studies (n = 8,061), two case-control study (n = 687), and three cohorts (n = 1,490) | Blood | EWAS | 187 CpGs | Association between 187 CpGs methylation and BMI Future risk for the development of T2D 6.3–40.2 kg/m2 change per 1 unit increase in DNA methylation |
✓/✓ | [25] |
| Adults (n = 185) | Blood | EWAS | CPT1A, TXNIP, ABCG1 | Association between CPT1A and TXNIP hypomethylation and T2D and BMI 2.7–3.77 rate of change of T2D characteristics as methylation changes |
✓/✓ | [112] |
| LOLIPOP study (n = 25,372) | Blood | EWAS | ABCG1, PHOSPHO1, SOCS3, SREBF1 | Association between gene methylation and BMI, WC, insulin and HOMA-IR 0.5–1.1% differences in DNA methylation between groups |
✓/✓ | [113] |
| Children (n = 438) | Neonatal blood |
EWAS | 69 CpGs | Association between 69 CpGs methylation and HOMA-IR, insulin, and glucose levels | ✓/✓ | [98] |
| Adults (three cohorts, n = 1,167) | Blood | EWAS | TXNIP | Association between DM and hypomethylation of TXNIP gene TXNIP hypomethylation is a consequence of hyperglycaemia levels 0.05–5% differences in DNA methylation between groups |
✓/✓ | [162] |
| LBW (n = 3), NBW (n = 3) and HBW (n = 3) | Cord blood and placenta | EWAS | 360 DMR in LBW and 773 DMR in HBW | Genes encoding p38 MAPK signalling components are hypermethylated in LBW and HBW PI3K/AKT pathway and glucose transport components hypomethylated in HBW |
✓/✓ | [163] |
| ESTHER study (n = 527) | Blood | EWAS | TXNIP | Association between TXNIP hypomethylation and increasing fasting glucose and HbA1c | ✓/✓ | [115] |
| FH (n = 276) and controls (n = 26) | Blood | EWAS | TNNT1 |
TNNT1 methylation is related to 10% of the inter-individual variation in HDL-c levels > 5% differences in DNA methylation between groups |
✓/✓ | [164] |
| Adults (three cohorts, n = 99,994) |
WBC | EWAS | IGFBP3,KCNK3, PDE3A,PRDM6, ARHGAP24,OSR1, SLC22A7,TBX2 | Association between gene methylation and BP and hypertension | x/✓ | [118] |
| Adults (n = 850) | Blood | EWAS | CPT1A, ABCG1 | Association between gene methylation and TG storage | ✓/✓ | [116] |
| Controls (n = 59) and obese (n = 58) |
SAT | EWAS | Genome-wide | Association between global DNA hypermethylation and WAT inflammation and IR | x/x | [92] |
| Botnia study: T2D (n = 129) and controls (n = 129) | Blood | Gene-specific |
PHOSPHO1, ABCG1 |
Association between PHOSPHO1 methylation and HDL-c levels. Association between ABCG1 methylation and BMI, HbA1c, fasting insulin and TG levels |
✓/✓ | [104] |
| Human cell line (in vitro) |
SW62 cell | Gene-specific | HSD11B2 | Association between HSD11B2 methylation and blood pressure | -/- | [117] |
| Human cell line (in vitro) |
NCI H295R | Gene-specific | AGT | Hypomethylation of AGT may result in hypertension and kidney injury. | -/- | [165] |
| C57BL/6J mice with IH | Endothelial cells | Gene-specific | Ace1, Agt | Association between gene methylation and systolic hypertension in mice exposed to IH | -/- | [166] |
Abbreviations: GDM, gestational diabetes mellitus; MetS, metabolic syndrome; VAT, visceral adipose tissue; T2D, type 2 diabetes; SAT, subcutaneous adipose tissue; FH, familiar
hypercholesterolemia; LDL-c, low density lipoprotein cholesterol; WC, waist circumference; apoB, apolipoprotein B; EWAS, epigenome-wide association study; CpG, cytosine linked by a phosphate
to guanine; BMI, body mass index; HbA1c, haemoglobin A1C; LOLIPOP, London Life Sciences Prospective Population Study; HOMA-IR, homeostasis model assessment-insulin resistance; WAT,
white adipose tissue; IR, insulin resistance; LBW, low birth weight; HBW, high birth weight; DMR, differentially methylation region; ESTHER, Epidemiologische Studie zu Chancen der Verhütung,
Früherkennung und optimierten THerapie chronischer ERkrankungen in der älteren Bevölkerung; HDL-c, high density lipoprotein cholesterol; TG, triglycerides; BMI, body mass index, SW62, colon
carcinoma cell line; NCIH295R, adrenocortical cell line; IH, intermitent hipoxia. ✓, adjusted; x, no adjusted; -, no information.
Two prospective case-control studies in new-borns from mothers with gestational diabetes, observed changes in the DNA methylation levels of different genes, supporting the hypothesis that these epigenetic alterations contribute to the life-long risk of the development of obesity and other metabolic disorders [109,110]. In addition, an EWAS involving 8,165 participants integrating data from six independent cohorts, two case-control, and eight retrospective studies, showed a causal relationship between adiposity and DNA methylation alteration in blood cells and adipose tissue. This data supports an important function for altered DNA methylation, mediated by visceral adiposity, in the development of several diseases, such as T2D, obesity, CVD, and cancer [25,111]. Moreover, a case-control study with 64 subjects focused on hypertriglyceridemia found an association between lipoprotein lipase (LPL) gene promoter methylation, which was higher in these individuals, and poor metabolic profile and the development of MetS [19].
Moreover, an EWAS performed in 483 children identified a number of DMRs at birth that were associated with insulin sensitivity in childhood [98]. Many of these changes in DNA methylation were causally related to health outcomes [98]. In addition, the methylation patterns of different genes have been associated with increased risk of MetS and T2D [112–114]. Some of these genes were also involved in the dysregulation of glucose metabolism, hypertriglyceridemia, and decreased HDL-C levels, suggesting a role in the development of IR and CVD [104,115,116]. Other studies have associated the methylation patterns of different genes with alterations in blood pressure (BP) and endothelial vascular function resulting in hypertriglyceridemia [117,118].
DNA methylation markers of weight loss
DNA methylation has been associated with body weight regulation, since it has been involved in weight-related functions such as appetite, adiposity, adipogenesis, glucose, and lipid metabolism [119]. In addition, dietary factors, such as FAs, polyphenols or methyl donors, physical exercise, or pharmacological and surgical treatments, can induce changes in the DNA methylation pattern [23,75,120–122]. In this context, several studies have identified DNA methylation as a mechanism that could be implicated in the inter-individual variability to weight loss response (Table 4).
Table 4.
Studies analysing the association between DNA methylation and weight loss.
| Weight loss strategy | Sample study | Sample type | Target gene | Main results | Age/Sex adjusted | Reference |
|---|---|---|---|---|---|---|
| Genotypic information | Obese and overweight (n = 95) | Blood | Global DNA | Association between global DNA hypomethylation and increased weight loss | ✓/✓ | [128] |
| Physical activity | Healthy women (n = 20) | Blood |
NAMPT,RUNX3, BR,SLCO4C, WNT7A,RASGRP3,CYP2E,CA13, KANK4,SMOC2, SLIT3,GABRG3 |
Baseline DNA methylation was able to predict the percent body weight change over the six-month period | -/- | [126] |
| Behavioural and ER | NW (n = 20), overweight/obese (n = 20), and morbid obese women (n = 20) | Blood | CLOCK, BMAL1, PER2 | Association between the baseline methylation of genes and the magnitude of weight loss. 10–60% differences in DNA methylation between groups at baseline |
-/- | [75] |
| No treatment | Adults (n = 51) | Blood | POMC | Association between DNA hypermethylation and body weight Increase 10% of DNA methylation per 2.8 kg/m2 |
✓/✓ | [167] |
| RYGB | Obese (n = 5) and control women (n = 6) | Skeletal muscle | 409 DMR | 409 DMR after weight loss >50% differences in DNA methylation between groups at baseline |
✓/- | [120] |
| ER | Overweight and obese women (n = 14) | Adipose tissue | 35 CpGs | Association between 35 loci methylation and weight control | ✓/- | [123] |
| ER | Obese women (n = 27) | Adipose tissue | LEP, TNF | Association between hypomethylation of genes at baseline with better response to the dietary intervention. | -/- | [125] |
| ER | Overweight and obese adolescents (n = 24) | Blood | AQP9,DUSP22, HIPK3,TNNT1, TNNI3 | Association between basal DNA methylation with changes in body weight, BMI-SDS, WC, and body fat mass after the weight loss intervention > 5% differences in DNA methylation between groups |
-/- | [168] |
| ER | Obese men (n = 18) |
Blood | POMC, NPY | Association between baseline NPY hypomethylation with weight-loss regain, and POMC hypomethylation with success in weight-loss maintenance |
-/- | [124] |
| ER, BS | Control (n = 9), obese + ER (n = 22), obese + BS women (n = 14) | Blood | SERPINE1 | Baseline DNA methylation may be a predictor of weight loss after BS | ✓/- | [127] |
| ER | Obese and overweight men (n = 12) | Blood | ATP10A, CD44, WT1 | Association between genes methylation at baseline and weight-loss outcome > 20% differences in DNA methylation between groups |
-/- | [20] |
Abbreviations: ER, energy-restriction; NW, normal weight; RYGB, Roux-en-Y gastric bypass; DMR, differentially methylated region; CpG, cytosine linked by a phosphate to guanine; BMI-SD, body mass index-standard deviation; WC, waist circumference; BS, bariatric surgery. ✓, adjusted; x, no adjusted; -, no information.
Investigations based on energy restriction have reported many CpGs whose methylation status was associated with the response to the weight loss intervention. For example, 35 different loci were differentially methylated between high and low responders to an energy-restricted diet in adipose tissue from overweight and obese women [123]. Many of these genes were associated with body weight control and insulin secretion [123].
In addition, the methylation status in adipose tissue and blood of several genes that participate in the regulation of BP, inflammation, lipid metabolism, appetite, and energy homeostasis has been related to differences in the weight loss response after a low-calorie diet (30% of energy-restriction) prescription [20,124,125]. On the other hand, DNA methylation of several clock genes implicated in the regulation of the circadian clockwork has been associated with body-weight loss in a women population under a behavioural and energy-restricted intervention [75].
Some studies have analysed other factors, such as physical activity or gastric surgery, in relation to the interaction between DNA methylation and weight loss. The analysis of blood samples from 20 healthy women within a physical activity programme identified significant associations between the methylation profile of 12 CpGs and weight loss [126]. A lifestyle and nutritional educational weight loss programme in 24 overweight and obese adolescents associated the methylation status of genes related to glucose metabolism, IR, inflammation, and CVD, with weight control and glucose and lipid metabolism [120,127].
Although genetic factors are involved in the regulation of body weight, genetic variants only partially explain the individual variation observed in the response to weight loss treatments. Thus, DNA methylation is thought to play a role in the environment-gene interaction in body weight regulation. In this context, Pirini et al. [128], studied the genetic and epigenetic alterations associated with weight loss within a personalized weight reduction programme designed on the basis of genotypic information, and identified an inverse association between global DNA methylation and weight loss depending on individual genetic variants for different genes.
Mechanisms linking DNA methylation and obesity
The present review has summarized the most relevant epidemiological studies analysing the association between DNA methylation and obesity during pregnancy, infancy, and adult life. Most of these analyses only describe a putative role of DNA methylation on the pathophysiology of obesity but are unable to deepen into the cause-and-effect relationship between epigenetics and obesity, which remains largely unknown.
Considering the epigenetic control of gene expression in the cellular activities, it is likely that the changes in DNA methylation contributed to the deregulation of important metabolic pathways and increased the susceptibility to develop obesity and its comorbidities. Despite the numerous epigenetic analyses performed in different populations and cohorts, they have not yet identified the precise mechanisms by which these changes in DNA methylation are related to the disease. However, some studies in rodents are helping to unveil the intricate mechanisms. For instance, DNA methyltransferase 3a (Dnmt3a) enzyme impaired insulin tolerance by increasing the DNA methylation of the direct target Ffg21 gene and reducing its expression in in vitro and in vivo models [129]. In the present review, multiple studies have shown changes in DNA methylation at key metabolic genes associated with obesity, such as HIF3A [130], IGFBP3 [118], SREBF1 [113], TNF [125], and CLOCK [75], pointing out to having important metabolic functions such as hypoxia, adipogenesis, inflammation, and circadian rhythm.
For example, HIF-system may play a role in mechanisms involved in the pathophysiology of adipose-tissue inflammation and IR. The abnormal expansion of WAT in obesity makes the correct blood perfusion on adipose tissue resulting in hypoxia impossible. HIF transcription factors are in charge of activating hypoxia-related gene expression, facilitating the adaptation to a hypoxic environment that contributes to inflammation and IR in WAT [65]. However, molecular mechanisms implicated in WAT dysfunction are still poorly characterized [131]. Therefore, HIF-system control genes related to glycolysis, such as pyruvate dehydrogenase kinase 1 (PDK1), lactate dehydrogenase A (LDHA), or glycogen phosphorylase L (PYGL), stimulating glucose uptake or increasing glucose production by activating phosphoenolpyruvate carboxykinase (PEPCK) in liver [132]; but HIFs also have a role in driving NF-κB and NFAT activation [131]. Interestingly, HIF activity has been reported to be regulated by DNA methylation [133,134]. This hypothesis has been demonstrated in the expression of the erythropoietin and class III beta-tubulin genes, where the HIF binding depends on the tissue-specific methylation status of HRE sites in the 3ʹUTR [135]. In addition to the role of DNA methylation in regulating HIF activity, hypoxia can induce epigenetic changes itself. Human colorectal and melanoma cell lines incubated in hypoxic conditions (< 0.1% oxygen) during 24 h suffered a 15–20% decrease in CpG methylation [135]. However, studies performed in conditions of hypobaric hypoxia (at high altitude) described progressive weight loss that was attributed to the reduction of appetite and food intake [65]. In addition, as a consequence of obesity and hypoxia, uncoupling protein one (UCP1) gene was stimulated, promoting thermogenesis by lipid oxidation and leading to browning of WAT [131]. These studies raise the controversial question: is hypoxia good or bad? In this context, recent investigations consider that the positive metabolic effects resulting from the activation of HIF signalling can be more beneficial than pathological against metabolic complications [65].
The process whereby fibroblast-like progenitors become mature adipocytes and accumulate nutrients and triglycerides is known as adipogenesis. Adipocyte expansion (hyperplasia) can improve the harmful effects of obesity [136]. Although many signalling pathways and ligands modulate the process of preadipocyte differentiation into mature adipocytes, the mechanisms and regulators of this adaptive mechanism are still under investigation [136]. Two of the main proteins involved in adipogenesis are peroxisome proliferator-activated receptor gamma (PPARγ) and co-activator CCAAT/enhancer-binding protein-α (C/EBPα). Also, the insulin signalling-cascade (including IGF receptors) plays an important role in this process [136]. In this context, in sows, maternal methyl donor supplementation throughout gestation has been reported to enhance the birth weight and postnatal growth rate of the offspring, associated with an increased expression of IGF1 and IGF1R, as well as an altered DNA methylation of IGF1 gene promoter [137]. During human adipogenesis, large epigenetic and transcriptomic changes occur in the transition from preadipocytes to mature adipocytes [138]; for example, the adipocytes present higher expression and lower methylation levels in genes involved in adipogenesis-related processes such as insulin signalling, PPAR signalling, and adipocytokine signalling [138]. These results suggest that DNA methylation changes may contribute to regulate adipogenesis. For instance, silencing of DNMT1 gene increases the rate of differentiation and modifies gene expression and lipid accumulation in mouse adipocytes [139].
In addition, other key physiological states involved in obesity, such as inflammation and circadian systems, can affect the adipogenic process. Obesity is characterized by a high expression of cytokines in WAT derived from the infiltrated macrophages. Macrophages correspond to about 40% of total WAT cells in obese subjects and 18% in lean individuals [140]. Macrophages secrete pro-inflammatory cytokines (i.e., TNF-α, IL-1β, and IL-6) and the inflammatory response stimulates Toll signalling pathways, which are able to phosphorylate insulin receptor substrate 1 (IRS-1) and, consequently, inhibit the insulin signal cascade [141], thus contributing to IR. The link between DNA methylation and inflammation in obesity-related comorbidities has been demonstrated. For example, epigenetic mechanisms play a key role in the regulation of macrophage activation and inflammation through the direct methylation of the PPARγ promoter by DNMT1 [142].
Recent investigations have revealed a wide interplay between the circadian system and metabolism. Disruptions in the physiological day-night cycles could be implicated in the development of obesity, as has been evidenced by studies of jet lag, shift work or disturbance in the timing of eating [143]. These conditions result in loss of internal rhythms and lead to weight gain and metabolic alterations [143]. Circadian rhythms are controlled by a set of core clock genes such as CLOCK, BMAL1, and NR1D1 (REV-ERBα). The CLOCK/BMAL1 heterodimer activates other core clock genes, such as PER and CRY [144]. These transcription factors drive the expression of many genes implicated in the regulation of lipid and glucose metabolism [145]. For instance, the circulating levels of lipids and the activity of enzymes involved in their metabolism, such as fatty acid synthase, fatty acyl-CoA synthetase 1, apolipoproteins, and low-density lipoprotein receptor, follow circadian rhythms in mammals [145]. CRY clock components also control gluconeogenesis by decreasing cAMP signalling, whereas CLOCK drives glycogen synthesis by enhancing Gys2 gene transcription [145]. In addition, the adipogenic factor PPARγ also oscillates with circadian rhythmicity in WAT [136]. PER activity is also cyclic in preadipocytes and, when it is activated, the adipogenic process is suppressed by the inhibition of PPARγ and KLF15 [146]. On the contrary, BMAL1, which oscillates in a contrary manner to PER, promotes adipogenesis by upregulating PPARγ [136]. Therefore, the abnormal DNA methylation of core clock genes could disrupt their oscillation causing metabolic disturbances.
Further studies are needed to elucidate the exact molecular mechanisms whereby DNA methylation and obesity are linked. However, transcriptional regulation of these genes, which participate in crucial pathways involved in the pathophysiology of obesity and its comorbidities, are partly mediated by DNA methylation. In this context, changes in the DNA methylation pattern of genes could contribute to modify the expression and disrupt the function of important metabolic pathways.
Critical analysis of the literature
Although there is increasing evidence of a link between epigenetic modifications and obesity and accompanying comorbidities in humans, yet several aspects remain obscure. The issue of causality remains the most pressing challenge because it is extremely difficult to establish and because most studies in humans only report correlations. To respond to this issue, it is necessary to follow big cohorts since conception, to compare their epigenetic marks (in different cell types) with those of their parents, to properly register the lifestyle and dietary patterns influences, to link DNA methylation results with gene expression in the same cells, and to analyse the interactions between the epigenetic marks, the genetic makeup and the environment. Currently, this task seems too vast and difficult.
Very few CpGs have repeatedly appeared in different human studies as significantly associated with obesity. This situation suggests that there is not probably a unique epigenetic pattern that is associated with the disease, but that the genetic, environmental, and physiological factors interact in a very personal way. On the other hand, one of the biggest limitations is that most studies in humans have been focused on peripheral blood cells, but very few have used liver, pancreas, muscle, adipose or other metabolically relevant tissues. As epigenetic marks are very dependent on cell type, it is imperative to broaden our knowledge of the epigenetic differences in these tissues, where most relevant biological functions (i.e., inflammation, hypoxia, IR,…) occur.
There are many studies focusing on individual CpGs that are differential between obese and non-obese subjects, but less have analysed DMR changes that could impact the regulation of the expression of metabolically relevant genes. On the other hand, the differences in DNA methylation are usually rather small (not higher than 15–20%) and very few studies have evidenced that these epigenetic changes are linked to changes in gene expression in normal conditions or in response to specific stressors. Theoretically, these small changes in methylation might be relevant when they are within a regulatory element of the gene, but evidence from human studies are lacking.
Another aspect that must be envisaged in the near future is the interplay between genetic, metagenomic and epigenetic characteristics of the individuals, and their environmental factors. Since epigenetic changes are a response to the environment, it can be hypothesized that each individual’s epigenetic pattern forms a kind of system shaped by his genetic background, perinatal history, long-term diet and lifestyle, pollutants, clinical history, drugs, usual gut microbiota, and many other factors. This extreme heterogeneity in the epigenetic patterns in the different cell types of the different individuals is one of the reasons that makes it difficult to find reliable epigenetic biomarkers associated with obesity.
As mentioned above, human studies are, currently, unable to deepen into the biological mechanisms that are linked to the epigenetic variations. There are so many environmental and biological factors that can mediate the relationships between epigenetic profile and health status, that it is very difficult to attribute specific epigenetic effects to different putative causative factors. The lack of adequate in vitro approaches and the inaccessibility to simulate the combination of several of these factors in a controlled manner adds additional difficulties to the analysis of possible causal nexi between all the variables simultaneously.
Nevertheless, in the last years considerable progress has been made in the identification and characterization of novel epigenetic signatures related to obesity and metabolic health, which could be relevant as risk biomarkers as well as causal factors. However, very few of them (if any) have been validated in independent cohorts. In this context, the use of large-scale population-based studies is necessary to untangle the complex interactions between epigenetics and environmental factors in the developmental origins of obesity and metabolic syndrome. To accomplish this objective, new instruments must be used to measure the nutritional, environmental, and parental interactions with the epigenetic modifications found in the individuals, including new indicators or markers of exposure, novel systems for collecting and processing nutritional data, and more control of the environmental conditions that may modify the epigenome.
Future perspectives
While these epigenetic studies allow understanding, at least partially, in the variability between obesity-related phenotypes and the interactions between genetics and environmental factors, it is still necessary to clarify how each specific environmental factor, such as diet or gut microbiota composition, targets specific genes to be methylated and demethylated. In addition, although all the studies found that DNA methylation is linked to obesity and obesity-related measures, until now, few studies have investigated the causal direction of phenotypic outcomes and DNA methylation profile. This question remains unclear and longitudinal analyses in other populations are required. It is also necessary to investigate how DNA methylation affects the pathophysiology of specific diseases, and the real effect on health and disease outcomes, in order to improve weight loss strategies. Finally, these studies could also help guide the use of DNA methylation regulation as a new therapeutic target for the prevention and treatment of obesity and other metabolic diseases.
Conclusion
The advances in the study of DNA methylation and the establishment of relevant longitudinal models in the study of obesity, may now allow to identify novel DNA methylation markers for obesity and related pathologies. Mainly, the first potential epigenetic markers at birth have been detected. This might help to predict obesity risk, adiposity and body size at a young age, and give opportunity for the development of prevention strategies. On the other hand, DNA methylation marks could improve the success of weight loss therapies in the context of precision nutrition. In summary, all these studies confirm that DNA methylation, in combination with genetic variants, gut microbiota and other biomarkers, could be useful in the assessment of metabolic risk and the personalization of the clinical management of obesity. However, it is necessary to conduct new studies in humans analysing the effects that nutritional, lifestyle and psychological factors can have on human health via epigenetic mechanisms.
Funding Statement
This work was supported by the Ministerio de Economía, Industria y Competitividad, Gobierno de España [AGL2013-4554-R]; Ministerio de Educación, Cultura y Deporte [BES-2014-068409].
Acknowledgments
The authors are grateful to Nora Goodwin (Technological University Dublin, Ireland) for English proofreading. The authors also acknowledge the financial support of MINECO (Nutrigenio Project reference AGL2013-45554-R), Spanish Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn) and University of Navarra. Mirian Samblas holds a FPI grant from the MECD (BES-2014-068409).
Disclosure statement
No potential conflict of interest was reported by the authors.
References
- [1].Afshin A, Forouzanfar Mh, Global Burden of Disease (GBD) 2015 Obesity Collaborators, et al. Health effects of overweight and obesity in 195 countries over 25 years. N Engl J Med. 2017;377:13–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Chatzigeorgiou A, Kandaraki E, Papavassiliou AG, et al. Peripheral targets in obesity treatment: a comprehensive update. Obes Rev. 2014;15:487–503. [DOI] [PubMed] [Google Scholar]
- [3].Biddle SJH, Bengoechea Garcia E, Pedisic Z, et al. Screen time, other sedentary behaviours, and obesity risk in adults: a review of reviews. Curr Obes Rep. 2017;6:134–147. [DOI] [PubMed] [Google Scholar]
- [4].St-Onge MP. Sleep-obesity relation: underlying mechanisms and consequences for treatment. Obes Rev. 2017;18:34–39. [DOI] [PubMed] [Google Scholar]
- [5].Kim TJ, Roesler NM, von Dem Knesebeck O. Causation or selection – examining the relation between education and overweight/obesity in prospective observational studies: a meta-analysis. Obes Rev. 2017;18:660–672. [DOI] [PubMed] [Google Scholar]
- [6].Weaver J. Classical endocrine diseases causing obesity. Front Horm Res. 2007;36:212–228. [DOI] [PubMed] [Google Scholar]
- [7].Martínez de Morentin-Aldabe BE, Hernández-Ruiz de Eguilaz M, Pérez-Díez S, et al. Influence of psychotropic drugs prescription on body weight increase. Rev Esp Nutr Hum Diet. 2013;17:17–26, Spanish. [Google Scholar]
- [8].Singh RK, Kumar P, Mahalingam K. Molecular genetics of human obesity: a comprehensive review. Comptes Rendus - Biol. 2017;340:87–108. [DOI] [PubMed] [Google Scholar]
- [9].Campion J, Milagro FI, Martinez JA. Individuality and epigenetics in obesity. Obes Rev. 2009;10:383–392. [DOI] [PubMed] [Google Scholar]
- [10].Huang Q, Ma C, Chen L, et al. Mechanistic insights into the interaction between transcription factors and epigenetic modifications and the contribution to the development of obesity. Front Endocrinol. 2018;9:370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Dupont C, Armant DR, Brenner CA. Epigenetics: definition, mechanisms and clinical perspective. Semin Reprod Med. 2009;27:351–357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Robertson KD. DNA methylation and human disease. Nat Rev Genet. 2005;6:597–610. [DOI] [PubMed] [Google Scholar]
- [13].Lister R, Pelizzola M, Dowen RH, et al. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature. 2009;462:315–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Smith ZD, Meissner A. DNA methylation: roles in mammalian development. Nat Rev Genet. 2013;14:204–220. [DOI] [PubMed] [Google Scholar]
- [15].Saxonov S, Berg P, Brutlag DL. A genome-wide analysis of CpG dinucleotides in the human genome distinguishes two distinct classes of promoters. Proc Natl Acad Sci. 2006;103:1412–1417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Shen L, Zhu J, Robert LSY, et al. Detect differentially methylated regions using non-homogeneous hidden Markov model for methylation array data. Bioinformatics. 2017;33:3701–3708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Zhang Y, Liu H, Lv J, et al. QDMR: a quantitative method for identification of differentially methylated regions by entropy. Nucleic Acids Res. 2011;39:e58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].NCD Risc Factor Collaboration (NCD-RisC) Worldwide trends in body-mass index, underweight, overweight, and obesity from 1975 to 2016: a pooled analysis of 2416 population-based measurement studies in 128.9 million children, adolescents, and adults. Lancet. 2017;390:2627–2642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Castellano-Castillo D, Moreno-Indias I, Fernandez-Garcia JC, et al. Adipose tissue LPL methylation is associated with triglyceride concentrations in the metabolic syndrome. Clin Chem. 2018;64:210–218. [DOI] [PubMed] [Google Scholar]
- [20].Milagro FI, Campion J, Cordero P, et al. A dual epigenomic approach for the search of obesity biomarkers: DNA methylation in relation to diet-induced weight loss. FASEB J. 2011;25:1378–1389. [DOI] [PubMed] [Google Scholar]
- [21].Milagro FI, Mansego ML, De Miguel C, et al. Dietary factors, epigenetic modifications and obesity outcomes: progresses and perspectives. Mol Aspects Med. 2013;34:782–812. [DOI] [PubMed] [Google Scholar]
- [22].Nilsson E, Ling C. DNA methylation links genetics, fetal environment, and an unhealthy lifestyle to the development of type 2 diabetes. Clin Epigenetics. 2017;9:105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Remely M, Lovrecic L, De La Garza AL, et al. Therapeutic perspectives of epigenetically active nutrients. Br J Pharmacol. 2015;172:2756–2768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].van Dijk SJ, Tellam RL, Morrison JL, et al. Recent developments on the role of epigenetics in obesity and metabolic disease. Clin Epigenetics. 2015;7:66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Wahl S, Drong A, Lehne B, et al. Epigenome-wide association study of body mass index, and the adverse outcomes of adiposity. Nature. 2016;541:81–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Wang X, Zhu H, Snieder H, et al. Obesity related methylation changes in DNA of peripheral blood leukocytes. BMC Med. 2010;8:87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Abdul QA, Yu BP, Chung HY, et al. Epigenetic modifications of gene expression by lifestyle and environment. Arch Pharm Res. 2017;40:1219–1237. [DOI] [PubMed] [Google Scholar]
- [28].Sharma A. Transgenerational epigenetics: integrating soma to germline communication with gametic inheritance. Mech Ageing Dev. 2017;163:15–22. [DOI] [PubMed] [Google Scholar]
- [29].Daxinger L, Whitelaw E. Understanding transgenerational epigenetic inheritance via the gametes in mammals. Nat Rev Genet. 2012;13:153–162. [DOI] [PubMed] [Google Scholar]
- [30].Hardikar AA, Satoor SN, Karandikar MS, et al. Multigenerational undernutrition increases susceptibility to obesity and diabetes that is not reversed after dietary recuperation. Cell Metab. 2015;22:312–319. [DOI] [PubMed] [Google Scholar]
- [31].Manikkam M, Tracey R, Guerrero-Bosagna C, et al. Plastics derived endocrine disruptors (BPA, DEHP and DBP) induce epigenetic transgenerational inheritance of obesity, reproductive disease and sperm epimutations. PLoS One. 2013;8:e55387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Tracey R, Manikkam M, Guerrero-Bosagna C, et al. Hydrocarbons (jet fuel JP-8) induce epigenetic transgenerational inheritance of obesity, reproductive disease and sperm epimutations. Reprod Toxicol. 2013;36:104–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Lumey LH, Van Poppel FWA. The Dutch famine of 1944–45: mortality and morbidity in past and present generations. Soc Hist Med. 1994;7:229–246. [DOI] [PubMed] [Google Scholar]
- [34].Ravelli AC, van Der Meulen JH, Osmond C, et al. Obesity at the age of 50 y in men and women exposed to famine prenatally. Am J Clin Nutr. 1999;70:811–816. [DOI] [PubMed] [Google Scholar]
- [35].Lumey LH, Stein AD, Kahn HS, et al. Lipid profiles in middle-aged men and women after famine exposure during gestation: the Dutch Hunger Winter Families Study. Am J Clin Nutr. 2009;89:1737–1743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Bossuyt PM, Painter RC, de Rooij SR, et al. Early onset of coronary artery disease after prenatal exposure to the Dutch famine. Am J Clin Nutr. 2018;84:322–327. [DOI] [PubMed] [Google Scholar]
- [37].Painter R, Osmond C, Gluckman P, et al. Transgenerational effects of prenatal exposure to the dutch famine on neonatal adiposity and health in later life. BJOG An Int J Obstet Gynaecol. 2008;115:1243–1249. [DOI] [PubMed] [Google Scholar]
- [38].Heijmans BT, Tobi EW, Stein AD, et al. Persistent epigenetic differences associated with prenatal exposure to famine in humans. Proc Natl Acad Sci. 2008;105:17046–17049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].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. 2009;18:4046–4053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Finer S, Iqbal MS, Lowe R, et al. Is famine exposure during developmental life in rural Bangladesh associated with a metabolic and epigenetic signature in young adulthood? a historical cohort study. BMJ Open. 2016;6:e011768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Waterland RA, Kellermayer R, Laritsky E, et al. Season of conception in rural Gambia affects DNA methylation at putative human metastable epialleles. PLoS Genet. 2010;6:1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Hochberg Z, Feil R, Constancia M, et al. Child health, developmental plasticity, and epigenetic programming. Endocr Rev. 2011;32:159–224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Barouki R, Gluckman PD, Grandjean P, et al. Developmental origins of non-communicable disease: implications for research and public health. Environ Heal. 2012;11:42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Li Y. Epigenetic mechanisms link maternal diets and gut microbiome to obesity in the offspring. Front Genet. 2018;9:342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].McKay JA, Mathers JC. Maternal folate deficiency and metabolic dysfunction in offspring. Proc Nutr Soc. 2016;75:90–95. [DOI] [PubMed] [Google Scholar]
- [46].Li HB, Tong J, Zhu S, et al. M6A mRNA methylation controls Tcell homeostasis by targeting the IL-7/STAT5/SOCS pathways. Nature. 2017;548:338–342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Thomas-Valdés S, Tostes MDGV, Anunciação PC, et al. Association between vitamin deficiency and metabolic disorders related to obesity. Crit Rev Food Sci Nutr. 2017;57:3332–3343. [DOI] [PubMed] [Google Scholar]
- [48].Kovacheva VP, Mellott TJ, Davison JM, et al. Gestational choline deficiency causes global and IGF2 gene DNA hypermethylation by up-regulation of DNMT1 expression. J Biol Chem. 2007;282:31777–31788. [DOI] [PubMed] [Google Scholar]
- [49].Medici V, Shibata NM, Kharbanda KK, et al. Maternal choline modifies fetal liver copper, gene expression, DNA methylation, and neonatal growth in the tx-j mouse model of Wilson disease. Epigenetics. 2014;9:286–296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Pauwels S, Ghosh M, Duca RC, et al. Maternal intake of methyl-group donors affects DNA methylation of metabolic genes in infants. Clin Epigenetics. 2017;9:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Zeisel SH. Choline, other methyl-donors and epigenetics. Nutrients. 2017;9 DOI: 10.3390/nu9050445 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Lomba A, Martínez JA, García-Díaz DF, et al. Weight gain induced by an isocaloric pair-fed high fat diet: a nutriepigenetic study on FASN and NDUFB6 gene promoters. Mol Genet Metab. 2010;101:273–278. [DOI] [PubMed] [Google Scholar]
- [53].Pembrey M, Saffery R, Bygren LO. Human transgenerational responses to early-life experience: potential impact on development, health and biomedical research. J Med Genet. 2014;51:563–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Vågerö D, Pinger PR, Aronsson V, et al. Paternal grandfather’s access to food predicts all-cause and cancer mortality in grandsons. Nat Commun. 2018;9:5124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Wang DD, Hu FB. Dietary fat and risk of cardiovascular disease: recent controversies and advances. Annu Rev Nutr. 2017;37:423–446. [DOI] [PubMed] [Google Scholar]
- [56].Fattore E, Fanelli R. Palm oil and palmitic acid: a review on cardiovascular effects and carcinogenicity. Int J Food Sci Nutr. 2013;64:648–659. [DOI] [PubMed] [Google Scholar]
- [57].Hall E, Volkov P, Dayeh T, et al. Effects of palmitate on genome-wide mRNA expression and DNA methylation patterns in human pancreatic islets. BMC Med. 2014;12:103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Flores-Sierra J, Arredondo-Guerrero M, Cervantes-Paz B, et al. The trans fatty acid elaidate affects the global DNA methylation profile of cultured cells and in vivo. Lipids Health Dis. 2016;15:75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Lim U, Song MA. Dietary and lifestyle factors of DNA methylation. Methods Mol Biol. 2012;863:359–376. [DOI] [PubMed] [Google Scholar]
- [60].Marranzano M, Ray S, Godos J, et al. Association between dietary flavonoids intake and obesity in a cohort of adults living in the mediterranean area. Int J Food Sci Nutr. 2018;69:1020–1029. [DOI] [PubMed] [Google Scholar]
- [61].Luft VC, Schmidt MI, Pankow JS, et al. Chronic inflammation role in the obesity-diabetes association: a case-cohort study. Diabetol Metab Syndr. 2013;5:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Rendo-Urteaga T, García-Calzón S, González-Muniesa P, et al. Peripheral blood mononuclear cell gene expression profile in obese boys who followed a moderate energy-restricted diet: differences between high and low responders at baseline and after the intervention. Br J Nutr. 2015;113:331–342. [DOI] [PubMed] [Google Scholar]
- [63].Fogel O, Richard-Miceli C, Tost J. Epigenetic changes in chronic inflammatory diseases. Acad Press. 2017;106: 139–189. [DOI] [PubMed] [Google Scholar]
- [64].Ansel KM, Lee DU, Rao A. An epigenetic view of helper T cell differentiation. Nat Immunol. 2003;4:616–623. [DOI] [PubMed] [Google Scholar]
- [65].Gaspar JM, Velloso LA. Hypoxia Inducible Factor as a central regulator of metabolism – implications for the development of obesity. Front Neurosci. 2018;12:813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].McGarry T, Biniecka M, Veale DJ, et al. Hypoxia, oxidative stress and inflammation. Free Radic Biol Med. 2018;125:15–24. [DOI] [PubMed] [Google Scholar]
- [67].Yara S, Lavoie JC, Levy E. Oxidative stress and DNA methylation regulation in the metabolic syndrome. Epigenomics. 2015;7:283–300. [DOI] [PubMed] [Google Scholar]
- [68].Tonna S, El-Osta A, Cooper ME, et al. Metabolic memory and diabetic nephropathy: potential role for epigenetic mechanisms. Nat Rev Nephrol. 2010;6:332–341. [DOI] [PubMed] [Google Scholar]
- [69].Arpón A, Milagro FI, Ramos-Lopez O, et al. Epigenome-wide association study in peripheral white blood cells involving insulin resistance. Sci Rep. 2019;9:2445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].Siervo M, Wells JC, Cizza G. The contribution of psychosocial stress to the obesity epidemic: an evolutionary approach. Horm Metab Res. 2009;41:261–270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Olstad DL, Ball K, Wright C, et al. Hair cortisol levels, perceived stress and body mass index in women and children living in socioeconomically disadvantaged neighborhoods: the READI study. Stress. 2016;19:158–167. [DOI] [PubMed] [Google Scholar]
- [72].Cao-Lei L, Dancause KN, Elgbeili G, et al. DNA methylation mediates the impact of exposure to prenatal maternal stress on BMI and central adiposity in children at age 13½ years: project Ice Storm. Epigenetics. 2015;10:749–761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Tarquini R, Mazzoccoli G. Clock genes, metabolism, and cardiovascular risk. Heart Fail Clin. 2017;13:645–655. [DOI] [PubMed] [Google Scholar]
- [74].Engin A. Circadian rhythms in diet-induced obesity. Adv Exp Med Biol. 2017;960:9–52. [DOI] [PubMed] [Google Scholar]
- [75].Milagro FI, Gómez-Abellán P, Campión J, et al. CLOCK, PER2 and BMAL1 DNA methylation: association with obesity and metabolic syndrome characteristics and monounsaturated fat intake. Chronobiol Int. 2012;29:1180–1194. [DOI] [PubMed] [Google Scholar]
- [76].Lahtinen A, Puttonen S, Vanttola P, et al. A distinctive DNA methylation pattern in insufficient sleep. Sci Rep. 2019;9:1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Ho SM, Cheong A, Adgent MA, et al. Environmental factors, epigenetics, and developmental origin of reproductive disorders. Reprod Toxicol. 2017;68:85–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Park SS, Skaar DA, Jirtle RL, et al. Epigenetics, obesity and early-life cadmium or lead exposure. Epigenomics. 2017;9:57–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Al-Assal K, Martinez AC, Torrinhas RS, et al. Gut microbiota and obesity. Clin Nutr Exp. 2018;20:60–64. [Google Scholar]
- [80].Baothman OA, Zamzami MA, Taher I, et al. The role of gut microbiota in the development of obesity and diabetes. Lipids Health Dis. 2016;15:108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Mischke M, Plösch T. The gut microbiota and their metabolites: potential implications for the host epigenome. Adv Exp Med Biol. 2016;902:33–44. [DOI] [PubMed] [Google Scholar]
- [82].Cuevas-Sierra A, Ramos-Lopez O, Riezu-Boj JI, et al. Diet, gut microbiota, and obesity: links with host genetics and epigenetics and potential applications. Adv Nutr. 2018;10(Suppl_1):S17–S30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Indrio F, Martini S, Francavilla R, et al. Epigenetic matters: the link between early nutrition, microbiome, and long-term health development. Front Pediatr. 2017;5:178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Nielsen JH, Haase TN, Jaksch C, et al. Impact of fetal and neonatal environment on beta cell function and development of diabetes. Acta Obstet Gynecol Scand. 2014;93:1109–1122. [DOI] [PubMed] [Google Scholar]
- [85].Rönn T, Volkov P, Davegardh C, et al. A six months exercise intervention influences the genome-wide DNA methylation pattern in human adipose tissue. PLoS Genet. 2013;9:e1003572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Lindholm ME, Marabita F, Gomez-Cabrero D, et al. An integrative analysis reveals coordinated reprogramming of the epigenome and the transcriptome in human skeletal muscle after training. Epigenetics. 2014;9:1557–1569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Sharp GC, Lawlor DA, Richmond RC, et al. Maternal pre-pregnancy BMI and gestational weight gain, offspring DNA methylation and later offspring adiposity: findings from the Avon Longitudinal Study of Parents and Children. Int J Epidemiol. 2015;44:1288–1304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Rhee JK, Lee JH, Yang HK, et al. DNA methylation profiles of blood cells are distinct between early-onset obese and control individuals. Genomics Inform. 2017;15:28–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Rzehak P, Covic M, Saffery R, et al. DNA methylation and body composition in preschool children: epigenome-wide-analysis in the European Childhood Obesity Project (ChOP)-study. Sci Rep. 2017;7:14349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Aslibekyan S, Demerath EW, Mendelson M, et al. Epigenome-wide study identifies novel methylation loci associated with body mass index and waist circumference. Obesity. 2015;23:1493–1501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Irvin MR, Zhi D, Joehanes R, et al. Epigenome-wide association study of fasting blood lipids in the genetics of lipid-lowering drugs and diet network study. Circulation. 2014;130:565–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Petrus P, Bialesova L, Checa A, et al. Adipocyte expression of SLC19A1 links DNA hypermethylation to adipose tissue inflammation and insulin resistance. J Clin Endocrinol Metab. 2017;103:710–721. [DOI] [PubMed] [Google Scholar]
- [93].Guay SP, Brisson D, Lamarche B, et al. ADRB3 gene promoter DNA methylation in blood and visceral adipose tissue is associated with metabolic disturbances in men. Epigenomics. 2014;6:33–43. [DOI] [PubMed] [Google Scholar]
- [94].Dunstan J, Bressler JP, Moran TH, et al. Associations of LEP, CRH, ICAM-1, and LINE-1 methylation, measured in saliva, with waist circumference, body mass index, and percent body fat in mid-childhood. Clin Epigenetics. 2017;9:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Oelsner KT, Guo Y, To SBC, et al. Maternal BMI as a predictor of methylation of obesity-related genes in saliva samples from preschool-age hispanic children at-risk for obesity. BMC Genomics. 2017;18:57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Rounge TB, Page CM, Lepistö M, et al. Genome-wide DNA methylation in saliva and body size of adolescent girls. Epigenomics. 2016;8:1495–1505. [DOI] [PubMed] [Google Scholar]
- [97].San-Cristobal R, Navas-Carretero S, Milagro FI, et al. Gene methylation parallelisms between peripheral blood cells and oral mucosa samples in relation to overweight. J Physiol Biochem. 2016;73:465–474. [DOI] [PubMed] [Google Scholar]
- [98].Van Dijk SJ, Peters TJ, Buckley M, et al. DNA methylation in blood from neonatal screening cards and the association with BMI and insulin sensitivity in early childhood. Int J Obes. 2018;42:28–35. [DOI] [PubMed] [Google Scholar]
- [99].Lin X, Lim IY, Wu Y, et al. Developmental pathways to adiposity begin before birth and are influenced by genotype, prenatal environment and epigenome. BMC Med. 2017;15:50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [100].Relton CL, Groom A, St. Pourcain B, et al. DNA methylation patterns in cord blood DNA and body size in childhood. PLoS One. 2012;7:e31821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [101].Godfrey KM, Sheppard A, Gluckman PD, et al. Epigenetic gene promoter methylation at birth is associated with child’s later adiposity. Diabetes. 2011;60:1528–1534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].Groom A, Potter C, Swan DC, et al. Postnatal growth and DNA methylation are associated with differential gene expression of the TACSTD2 gene and childhood fat mass. Diabetes. 2012;61:391–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].Perng W, Mora-Plazas M, Marín C, et al. A prospective study of LINE-1 DNA methylation and development of adiposity in school-age children. PLoS One. 2013;8:e62587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Dayeh T, Tuomi T, Almgren P, et al. DNA methylation of loci within ABCG1 and PHOSPHO1 in blood DNA is associated with future type 2 diabetes risk. Epigenetics. 2016;11:482–488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [105].Sun D, Heianza Y, Li X, et al. Genetic, epigenetic and transcriptional variations at NFATC2IP locus with weight loss in response to diet interventions: the POUNDS lost trial. Diabetes Obes Metab. 2018;20:2298–2303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Beilby J. Definition of metabolic syndrome : report of the national heart, lung, and blood Institute/American Heart Association conference on scientific issues related to definition. Clin Biochem Rev. 2009;25:195–198. [Google Scholar]
- [107].Onat A. Metabolic syndrome: nature, therapeutic solutions and options. Expert Opin Pharmacother. 2011;12:1887–1900. [DOI] [PubMed] [Google Scholar]
- [108].Marcuello C, Calle-Pascual AL, Fuentes M, et al. Prevalence of the metabolic syndrome in spain using regional cutoff points for waist circumference: the di@bet.es study. Acta Diabetol. 2013;50:615–623. [DOI] [PubMed] [Google Scholar]
- [109].Chen D, Zhang A, Fang M, et al. Increased methylation at differentially methylated region of GNAS in infants born to gestational diabetes. BMC Med Genet. 2014;15:108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].El Hajj N, Pliushch G, Schneider E, et al. Metabolic programming of MEST DNA methylation by intrauterine exposure to gestational diabetes mellitus. Diabetes. 2013;62:1320–1328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].Rönn T, Volkov P, Gillberg L, et al. Impact of age, BMI and HbA1c levels on the genome-wide DNA methylation and mRNA expression patterns in human adipose tissue and identification of epigenetic biomarkers in blood. Hum Mol Genet. 2015;24:3792–3813. [DOI] [PubMed] [Google Scholar]
- [112].Al Muftah WA, Al-Shafai M, Zaghlool SB, et al. Epigenetic associations of type 2 diabetes and BMI in an Arab population. Clin Epigenetics. 2016;8:13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Chambers JC, Loh M, Lehne B, et al. Epigenome-wide association of DNA methylation markers in peripheral blood from Indian Asians and Europeans with incident type 2 diabetes: a nested case-control study. Lancet Diabetes Endocrinol. 2015;3:526–534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Ling C, Del Guerra S, Lupi R, et al. Epigenetic regulation of PPARGC1A in human type 2 diabetic islets and effect on insulin secretion. Diabetologia. 2008;51:615–622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [115].Florath I, Butterbach K, Heiss J, et al. Type 2 diabetes and leucocyte DNA methylation: an epigenome-wide association study in over 1,500 older adults. Diabetologia. 2016;59:130–138. [DOI] [PubMed] [Google Scholar]
- [116].Mamtani M, Kulkarni H, Dyer TD, et al. Genome- and epigenome-wide association study of hypertriglyceridemic waist in Mexican American families. Clin Epigenetics. 2016;8:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [117].Alikhani-Koopaei R, Fouladkou F, Frey FJ, et al. Epigenetic regulation of 11β-hydroxysteroid dehydrogenase type 2 expression. J Clin Invest. 2004;114:1146–1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [118].Kato N, Loh M, Takeuchi F, et al. Trans-ancestry genome-wide association study identifies 12 genetic loci influencing blood pressure and implicates a role for DNA methylation. Nat Genet. 2015;47:1282–1293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [119].Cheng Z, Zheng L, Almeida FA. Epigenetic reprogramming in metabolic disorders: nutritional factors and beyond. J Nutr Biochem. 2018;54:1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [120].Barres R, Kirchner H, Rasmussen M, et al. Weight loss after gastric bypass surgery in human obesity remodels promoter methylation. Cell Rep. 2013;3:1020–1027. [DOI] [PubMed] [Google Scholar]
- [121].Fiorito G, Guarrera S, Valle C, et al. B-vitamins intake, DNA-methylation of one carbon metabolism and homocysteine pathway genes and myocardial infarction risk: the EPICOR study. Nutr Metab Cardiovasc Dis. 2014;24:483–488. [DOI] [PubMed] [Google Scholar]
- [122].Tremblay BL, Guénard F, Rudkowska I, et al. Epigenetic changes in blood leukocytes following an omega-3 fatty acid supplementation. Clin Epigenetics. 2017;9:43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [123].Bouchard L, Rabasa-Lhoret R, Faraj M, et al. Differential epigenomic and transcriptomic responses in subcutaneous adipose tissue between low and high responders to caloric restriction. Am J Clin Nutr. 2010;91:309–320. [DOI] [PubMed] [Google Scholar]
- [124].Crujeiras AB, Campion J, Díaz-Lagares A, 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. [DOI] [PubMed] [Google Scholar]
- [125].Cordero P, Campion J, Milagro FI, et al. Leptin and TNF-alpha promoter methylation levels measured by MSP could predict the response to a low-calorie diet. J Physiol Biochem. 2011;67:463–470. [DOI] [PubMed] [Google Scholar]
- [126].McEwen LM, Gatev EG, Jones MJ, et al. DNA methylation signatures in peripheral blood mononuclear cells from a lifestyle intervention for women at midlife: a pilot randomized controlled trial. Appl Physiol Nutr Metab. 2018;43:233–239. [DOI] [PubMed] [Google Scholar]
- [127].Nicoletti CF, Nonino CB, de Oliveira BAP, et al. DNA methylation and hydroxymethylation levels in relation to two weight loss strategies: energy-restricted diet or bariatric surgery. Obes Surg. 2016;26:603–611. [DOI] [PubMed] [Google Scholar]
- [128].Pirini F, Rodriguez‑Torres S, Ayandibu B, et al. INSIG2 rs7566605 single nucleotide variant and global DNA methylation index levels are associated with weight loss in a personalized weight reduction program. Mol Med Rep. 2017;17:1699–1709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [129].You D, Nilsson E, Tenen DE, et al. DNMT3a is an epigenetic mediator of adipose insulin resistance. Elife. 2017;6 DOI: 10.7554/eLife.30766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [130].Dick KJ, Nelson CP, Tsaprouni L, et al. DNA methylation and body-mass index: a genome-wide analysis. Lancet. 2014;383:1990–1998. [DOI] [PubMed] [Google Scholar]
- [131].Foti DP, Brunetti A. Linking hypoxia to obesity. Front Endocrinol. 2017;8:34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [132].Choi JH, Park MJ, Kim KW, et al. Molecular mechanism of hypoxia-mediated hepatic gluconeogenesis by transcriptional regulation. FEBS Lett. 2005;579:2795–2801. [DOI] [PubMed] [Google Scholar]
- [133].Pfeiffer S, Krüger J, Maierhofer A, et al. Hypoxia-inducible factor 3a gene expression and methylation in adipose tissue is related to adipose tissue dysfunction. Sci Rep. 2016;6:27969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [134].Main AM, Gillberg L, Jacobsen AL, et al. DNA methylation and gene expression of HIF3a: cross-tissue validation and associations with BMI and insulin resistance. Clin Epigenetics. 2016;8:89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [135].Watson JA, Watson CJ, McCann A, et al. Epigenetics: the epicenter of the hypoxic response. Epigenetics. 2010;5:293–296. [DOI] [PubMed] [Google Scholar]
- [136].Ghaben AL, Scherer PE. Adipogenesis and metabolic health. Nat Rev Mol Cell Biol. 2019. DOI: 10.1038/s41580-018-0093-z [DOI] [PubMed] [Google Scholar]
- [137].Jin C, Zhuo Y, Wang J, et al. Methyl donors dietary supplementation to gestating sows diet improves the growth rate of offspring and is associating with changes in expression and DNA methylation of insulin-like growth factor-1 gene. J Anim Physiol Anim Nutr. 2018;102:1340–1350. [DOI] [PubMed] [Google Scholar]
- [138].Broholm C, Olsson AH, Perfilyev A, et al. Human adipogenesis is associated with genome-wide DNA methylation and gene-expression changes. Epigenomics. 2016;8:1601–1617. [DOI] [PubMed] [Google Scholar]
- [139].Londono Gentile T, Lu C, Lodato PM, et al. DNMT1 is regulated by ATP-citrate lyase and maintains methylation patterns during adipocyte differentiation. Mol Cell Biol. 2013;33:3864–3878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [140].Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest. 2007;117:175–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [141].Rogero M, Calder P. Obesity, inflammation, toll-like receptor 4 and fatty acids. Nutrients. 2018;10:432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [142].Wang X, Cao Q, Yu L, et al. Epigenetic regulation of macrophage polarization and inflammation by DNA methylation in obesity. JCI Insight. 2016;1:e87748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [143].Laermans J, Depoortere I. Chronobesity: role of the circadian system in the obesity epidemic. Obes Rev. 2016;17:108–125. [DOI] [PubMed] [Google Scholar]
- [144].Mayeuf-Louchart A, Zecchin M, Staels B, et al. Circadian control of metabolism and pathological consequences of clock perturbations. Biochimie. 2017;143:42–50. [DOI] [PubMed] [Google Scholar]
- [145].Mazzoccoli G, Pazienza V, Vinciguerra M. Clock genes and clock-controlled genes in the regulation of metabolic rhythms. Chronobiol Int. 2012;29:227–251. [DOI] [PubMed] [Google Scholar]
- [146].Grimaldi B, Bellet MM, Katada S, et al. PER2 controls lipid metabolism by direct regulation of PPARγ. Cell Metab. 2010;12:509–520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [147].Almén MS, Jacobsson JA, Moschonis G, et al. Genome wide analysis reveals association of a FTO gene variant with epigenetic changes. Genomics. 2012;99:132–137. [DOI] [PubMed] [Google Scholar]
- [148].Meeks KAC, Henneman P, Venema A, et al. An epigenome-wide association study in whole blood of measures of adiposity among Ghanaians: the RODAM study. Clin Epigenetics. 2017;9:103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [149].Stepanow S, Reichwald K, Huse K, et al. Allele-specific, age-dependent and BMI-associated DNA methylation of human MCHR1. PLoS One. 2011;6:e17711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [150].Kuehnen P, Mischke M, Wiegand S, et al. An alu element-associated hypermethylation variant of the POMC gene is associated with childhood obesity. PLoS Genet. 2012;8:e1002543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [151].Carraro JCC, Mansego ML, Milagro FI, et al. LINE-1 and inflammatory gene methylation levels are early biomarkers of metabolic changes: association with adiposity. Biomarkers. 2016;21:625–632. [DOI] [PubMed] [Google Scholar]
- [152].Mansego ML, Milagro FI, Zulet MA, et al. Differential DNA methylation in relation to age and health risks of obesity. Int J Mol Sci. 2015;16:16816–16832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [153].Soubry A, Murphy SK, Wang F, et al. Newborns of obese parents have altered DNA methylation patterns at imprinted genes. Int J Obes. 2015;39:650–657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [154].Morales E, Groom A, Lawlor DA, et al. DNA methylation signatures in cord blood associated with maternal gestational weight gain: results from the ALSPAC cohort. BMC Res Notes. 2014;7:278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [155].Liu X, Chen Q, Tsai HJ, et al. Maternal preconception body mass index and offspring cord blood DNA methylation: exploration of early life origins of disease. Environ Mol Mutagen. 2014;55:223–230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [156].Zhao J, Goldberg J, Vaccarino V. Promoter methylation of serotonin transporter gene is associated with obesity measures: a monozygotic twin study. Int J Obes. 2013;37:140–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [157].Demerath EW, Guan W, Grove ML, et al. Epigenome-wide association study (EWAS) of BMI, BMI change and waist circumference in African American adults identifies multiple replicated loci. Hum Mol Genet. 2015;24:4464–4479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [158].Lai CQ, Smith CE, Parnell LD, et al. Epigenomics and metabolomics reveal the mechanism of the APOA2 -saturated fat intake interaction affecting obesity. Am J Clin Nutr. 2018;108:188–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [159].Volberg V, Yousefi P, Huen K, et al. CpG methylation across the adipogenic PPARγ gene and its relationship with birthweight and child BMI at 9 years. BMC Med Genet. 2017;18:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [160].Houshmand-Oeregaard A, Hjort L, Kelstrup L, et al. DNA methylation and gene expression of TXNIP in adult offspring of women with diabetes in pregnancy. PLoS One. 2017;12:e0187038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [161].Laker RC, Garde C, Camera DM, et al. Transcriptomic and epigenetic responses to short-term nutrient-exercise stress in humans. Sci Rep. 2017;7:15134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [162].Soriano-Tárraga C, Jiménez-Conde J, Giralt-Steinhauer E, et al. Epigenome-wide association study identifies TXNIP gene associated with type 2 diabetes mellitus and sustained hyperglycemia. Hum Mol Genet. 2016;25:609–619. [DOI] [PubMed] [Google Scholar]
- [163].Mao LL, Xiao XH, Zhang Q, et al. DNA methylation and birth weight: a genome-wide analysis. Biomed Environ Sci. 2017;30:667–670. [DOI] [PubMed] [Google Scholar]
- [164].Guay SP, Voisin G, Brisson D, et al. Epigenome-wide analysis in familial hypercholesterolemia identified new loci associated with high-density lipoprotein cholesterol concentration. Epigenomics. 2012;4:623–639. [DOI] [PubMed] [Google Scholar]
- [165].Wang F, Demura M, Cheng Y, et al. Dynamic CCAAT/enhancer binding protein-associated changes of DNA methylation in the angiotensinogen gene. Hypertension. 2014;63:281–288. [DOI] [PubMed] [Google Scholar]
- [166].Chu A, Gozal D, Cortese R, et al. Cardiovascular dysfunction in adult mice following postnatal intermittent hypoxia. Pediatr Res. 2015;77:425–433. [DOI] [PubMed] [Google Scholar]
- [167].Kühnen P, Handke D, Waterland RA, et al. Interindividual variation in DNA methylation at a putative POMC metastable epiallele is associated with obesity. Cell Metab. 2016;24:502–509. [DOI] [PubMed] [Google Scholar]
- [168].Moleres A, Campion J, Milagro FI, et al. Differential DNA methylation patterns between high and low responders to a weight loss intervention in overweight or obese adolescents: the EVASYON study. FASEB J. 2013;27:2504–2512. [DOI] [PubMed] [Google Scholar]
